Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
JCI Insight ; 4(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31487265

ABSTRACT

Autoimmune diseases resulting from MHC class II-restricted autoantigen-specific T cell immunity include the systemic inflammatory autoimmune conditions rheumatoid arthritis and vasculitis. While currently treated with broad-acting immunosuppressive drugs, a preferable strategy is to regulate antigen-specific effector T cells (Teffs) to restore tolerance by exploiting DC antigen presentation. We targeted draining lymph node (dLN) phagocytic DCs using liposomes encapsulating 1α,25-dihydroxyvitamin D3 (calcitriol) and antigenic peptide to elucidate mechanisms of tolerance used by DCs and responding T cells under resting and immunized conditions. PD-L1 expression was upregulated in dLNs of immunized relative to naive mice. Subcutaneous administration of liposomes encapsulating OVA323-339 and calcitriol targeted dLN PD-L1hi DCs of immunized mice and reduced their MHC class II expression. OVA323-339/calcitriol liposomes suppressed expansion, differentiation, and function of Teffs and induced Foxp3+ and IL-10+ peripheral Tregs in an antigen-specific manner, which was dependent on PD-L1. Peptide/calcitriol liposomes modulated CD40 expression by human DCs and promoted Treg induction in vitro. Liposomes encapsulating calcitriol and disease-associated peptides suppressed the severity of rheumatoid arthritis and Goodpasture's vasculitis models with suppression of antigen-specific memory T cell differentiation and function. Accordingly, peptide/calcitriol liposomes leverage DC PD-L1 for antigen-specific T cell regulation and induce antigen-specific tolerance in inflammatory autoimmune diseases.


Subject(s)
Anti-Glomerular Basement Membrane Disease/drug therapy , Arthritis, Rheumatoid/drug therapy , Calcitriol/administration & dosage , Dendritic Cells/immunology , Immunodominant Epitopes/administration & dosage , Adoptive Transfer , Animals , Anti-Glomerular Basement Membrane Disease/diagnosis , Anti-Glomerular Basement Membrane Disease/immunology , Antigen Presentation/drug effects , Arthritis, Rheumatoid/diagnosis , Arthritis, Rheumatoid/immunology , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , CHO Cells , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cricetulus , Dendritic Cells/drug effects , Dendritic Cells/transplantation , Disease Models, Animal , Female , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , Humans , Immune Tolerance/drug effects , Immunodominant Epitopes/immunology , Immunologic Memory/drug effects , Injections, Subcutaneous , Liposomes , Lymph Nodes/cytology , Mice , Mice, Transgenic , Ovalbumin/administration & dosage , Peptide Fragments/administration & dosage , Phagocytosis/drug effects , Phagocytosis/immunology , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
2.
Vaccine ; 37(12): 1546-1553, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30782490

ABSTRACT

Theileria parva kills over one million cattle annually in sub-Saharan Africa. Parasite genetic complexity, cellular response immunodominance, and bovine MHC diversity have precluded traditional vaccine development. One potential solution is gene gun (GG) immunization, which enables simultaneous administration of one or more DNA-encoded antigens. Although promising in murine, porcine, and human vaccination trials, bovine GG immunization studies are limited. We utilized the model T. parva antigen, polymorphic immunodominant molecule (PIM) to test bovine GG immunization. GG immunization using a mammalian codon optimized PIM sequence elicited significant anti-PIM antibody and cell-mediated responses in 7/8 steers, but there was no difference between immunized and control animals following T. parva challenge. The results suggest immunization with PIM, as delivered here, is insufficient to protect cattle from T. parva. Nonetheless, the robust immune responses elicited against this model antigen suggest GG immunization is a promising vaccine platform for T. parva and other bovine pathogens.


Subject(s)
Antibodies, Protozoan/immunology , Biolistics/methods , CD4-Positive T-Lymphocytes/immunology , Cattle Diseases/immunology , Immunodominant Epitopes/immunology , Theileria parva/immunology , Theileriasis/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cattle , Cattle Diseases/prevention & control , Codon , Immunity, Cellular , Immunity, Humoral , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Protozoan Vaccines/administration & dosage , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Theileriasis/prevention & control
3.
Arch Virol ; 164(2): 359-370, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30367292

ABSTRACT

In this study, an alphavirus vector platform was used to deliver replicon particles (RPs) expressing African swine fever virus (ASFV) antigens to swine. Alphavirus RPs expressing ASFV p30 (RP-30), p54 (RP-54) or pHA-72 (RP-sHA-p72) antigens were constructed and tested for expression in Vero cells and for immunogenicity in pigs. RP-30 showed the highest expression in Vero cells and was the most immunogenic in pigs, followed by RP-54 and RP-sHA-p72. Pigs primed with two doses of the RP-30 construct were then boosted with a naturally attenuated ASFV isolate, OURT88/3. Mapping of p30 identified an immunodominant region within the amino acid residues 111-130. However, the principal effect of the prime-boost was enhanced recognition of an epitope covered by the peptide sequence 61-110. The results suggest that a strategy incorporating priming with a vector-expressed antigen followed by boosting with an attenuated live virus may broaden the recognition of ASFV epitopes.


Subject(s)
African Swine Fever Virus/immunology , African Swine Fever/immunology , Antigens, Viral/immunology , Viral Vaccines/immunology , African Swine Fever/prevention & control , African Swine Fever/virology , African Swine Fever Virus/genetics , Alphavirus/genetics , Alphavirus/metabolism , Animals , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Chlorocebus aethiops , Drug Evaluation, Preclinical , Gene Expression , Immunization, Secondary , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Swine , Vero Cells , Viral Vaccines/administration & dosage
4.
Parasit Vectors ; 9(1): 435, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27502394

ABSTRACT

BACKGROUND: Vaccination is an attractive ecological alternative to the use of acaricides for parasite control. However, effective anti-parasite vaccines against sarcoptic mange have not yet been developed. The purpose of this study was first to identify Sarcoptes scabiei immunodominant antigens and second to evaluate them as vaccine candidates in a rabbit/S. scabiei var. cuniculi model. METHODS: The S. scabiei Ssλ15 immunodominant antigen was selected by immunoscreening of a S. scabiei var. hominis cDNA. The full-length cDNA was sequenced and cloned into the pGEX vector and the recombinant protein expressed in BL21 (DE3) cells and purified. A vaccination trial was performed consisting of a test group (n = 8) immunised with recAgs (a mix of two recombinant antigens, Ssλ15 and the previously described Ssλ20∆B3) and a control group (n = 8) immunised with PBS. All analyses were performed with R Statistical Environment with α set at 0.050. RESULTS: The full-length open reading frame of the 1,821 nt cloned cDNA encodes a 64 kDa polypeptide, the sequence of which had 96 % identity with a hypothetical protein of S. scabiei. Ssλ15 was localised by immunostaining of skin sections in the tegument surrounding the mouthparts and the coxa in the legs of mites. Rabbit immunisation with recAgs induced high levels of specific IgG (P < 0.010) and increased levels of total IgEs. However, no significant clinical protection against S. scabiei challenge was detected. Unexpectedly, the group immunised with the recAgs mix had significantly higher lesion scores (P = 0.050) although lower mean mite densities than those observed in the control group. These results might indicate that the lesions in the recAgs group were due not only to the mites density but also to an exacerbated immunological response after challenge, which is in agreement with the specific high levels of pro-inflammatory cytokines (IL-1 and TNFα) detected after challenge in this group. CONCLUSIONS: The selected antigens delivered as recombinant proteins had no clinical protective efficacy against S. scabiei infestation although immunisation reduced mite density. However, these results pave the way for future studies on alternative production systems, adjuvants, delivery methods and combinations of antigens in order to manage stimulation of clinical protective immune responses.


Subject(s)
Arthropod Proteins/immunology , Cytokines/immunology , Immunity, Humoral , Immunodominant Epitopes/immunology , Sarcoptes scabiei/immunology , Scabies/immunology , Animals , Antibodies/immunology , Arthropod Proteins/administration & dosage , Arthropod Proteins/genetics , Female , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Rabbits , Sarcoptes scabiei/genetics , Scabies/parasitology , Scabies/prevention & control , Vaccination
5.
J Immunol ; 196(6): 2637-45, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26864033

ABSTRACT

The currently used vaccine strategy to combat influenza A virus (IAV) aims to provide highly specific immunity to circulating seasonal IAV strains. However, the outbreak of 2009 influenza pandemic highlights the danger in this strategy. In this study, we tested the hypothesis that universal vaccination that offers broader but weaker protection would result in cross protective T cell responses after primary IAV infection, which would subsequently provide protective immunity against future pandemic strains. Specifically, we used tandem repeat extracellular domain of M2 (M2e) epitopes on virus-like particles (M2e5x VLP) that induced heterosubtypic immunity by eliciting Abs to a conserved M2e epitope. M2e5x VLP was found to be superior to strain-specific current split vaccine in conferring heterosubtypic cross protection and in equipping the host with cross-protective lung-resident nucleoprotein-specific memory CD8(+) T cell responses to a subsequent secondary infection with a new pandemic potential strain. Immune correlates for subsequent heterosubtypic immunity by M2e5x VLP vaccination were found to be virus-specific CD8(+) T cells secreting IFN-γ and expressing lung-resident memory phenotypic markers CD69(+) and CD103(+) as well as M2e Abs. Hence, vaccination with M2e5x VLP may be developable as a new strategy to combat future pandemic outbreaks.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/immunology , Lung/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Viral Matrix Proteins/administration & dosage , Animals , CD8-Positive T-Lymphocytes/virology , Cross Reactions , Female , Humans , Immunodominant Epitopes/chemistry , Immunologic Memory , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Interferon-gamma/metabolism , Lymphocyte Activation , Mass Vaccination/methods , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/prevention & control , Pandemics , Viral Matrix Proteins/chemistry , Virion/chemistry
6.
Scand J Immunol ; 82(4): 380-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26179420

ABSTRACT

Lymphatic filariasis, a mosquito-borne parasitic disease, affects more than 120 million people worldwide. Vaccination for filariasis by targeting different stages of the parasite will be a boon to the existing MDA efforts of WHO which required repeated administration of the drug to reduce the infection level and sustained transmission. Onset of a filaria-specific immune response achieved through antigen vaccines can act synergistically with these drugs to enhance the parasite killing. Multi-epitope vaccine approach has been proved to be successful against several parasitic diseases as it overcomes the limitations associated with the whole antigen vaccines. Earlier results from our group suggested the protective efficacy of multi-epitope vaccine comprising two immunodominant epitopes from Brugia malayi antioxidant thioredoxin (TRX), several epitopes from transglutaminase (TGA) and abundant larval transcript-2 (ALT-2). In this study, the prophylactic efficacy of the filarial epitope protein (FEP), a chimera of selective epitopes identified from our earlier study, was tested in a murine model (jird) of filariasis with L3 larvae. FEP conferred a significantly (P < 0.0001) high protection (69.5%) over the control in jirds. We also observed that the multi-epitope recombinant construct (FEP) induces multiple types of protective immune responses, thus ensuring the successful elimination of the parasite; this poses FEP as a potential vaccine candidate.


Subject(s)
Elephantiasis, Filarial/prevention & control , Immunodominant Epitopes/administration & dosage , Protozoan Vaccines/immunology , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Helminth/immunology , Antibodies, Protozoan/blood , Antigens, Helminth/immunology , Brugia malayi/immunology , Brugia malayi/pathogenicity , Disease Models, Animal , Gerbillinae , Helminth Proteins/immunology , Humans , Immunodominant Epitopes/immunology , Male , Mice , Protozoan Vaccines/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Proteins/immunology , Thioredoxins/immunology , Transglutaminases/immunology , Vaccination , Wuchereria bancrofti/pathogenicity
7.
Acta Biochim Pol ; 61(1): 129-32, 2014.
Article in English | MEDLINE | ID: mdl-24644555

ABSTRACT

Most Proteus lipopolysaccharides (LPSs) contain uronic acids or their amides with different amino acids, which together with other negatively charged components account for the acidic character of such LPS molecules. Previous studies have shown the significance of an amide of galacturonic acid with lysine [D-GalA(L-Lys)] for serological specificity of O-antigens from few P. mirabilis strains. In this work, the immunodominant role of GalALys was indicated for the P. penneri 42 LPS core region. The studies also showed the serological identity of core oligosaccharides from P. penneri 42 (O71), P. mirabilis 51/57 (O28) and R14/S1959 strains.


Subject(s)
Hexuronic Acids/administration & dosage , Lipopolysaccharides/administration & dosage , O Antigens/immunology , Amides , Hexuronic Acids/chemistry , Immunodominant Epitopes/administration & dosage , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Lysine/administration & dosage , Lysine/chemistry , Lysine/immunology , O Antigens/chemistry , Proteus penneri/immunology
8.
Haematologica ; 99(3): 588-96, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24441145

ABSTRACT

The offspring from pregnancies of women who have developed anti-D blood group antibodies are at risk of hemolytic disease of the newborn. We have previously mapped four peptides containing immunodominant T-helper cell epitopes from the RhD protein and the purpose of the work was to develop these into a product for suppression of established anti-D responses. A panel of each of the four immunodominant RhD peptides was synthesized with modifications to improve manufacturability and solubility, and screened for retention of recognition by human T-helper cells. A selected version of each sequence was combined in a mixture (RhDPmix), which was tested for suppressive ability in a humanized murine model of established immune responses to RhD protein. After HLA-DR15 transgenic mice had been immunized with RhD protein, a single dose of RhDPmix, given either intranasally (P=0.008, Mann-Whitney rank sum test) or subcutaneously (P=0.043), rapidly and significantly suppressed the ongoing antibody response. This was accompanied by reduced T-helper cell responsiveness, although this change was less marked for subcutaneous RhDPmix delivery, and by the recruitment of cells with a regulatory T-cell phenotype. The results support human trials of RhDPmix peptide immunotherapy in women with established antibody responses to the RhD blood group.


Subject(s)
Antibody Formation/genetics , Antibody Formation/immunology , Histocompatibility Antigens/genetics , Peptide Fragments/immunology , Rh-Hr Blood-Group System/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Adult , Animals , Female , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Immunotherapy , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Middle Aged , Peptide Fragments/administration & dosage , Rh-Hr Blood-Group System/chemistry , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Young Adult
9.
Vaccine ; 32(3): 375-82, 2014 Jan 09.
Article in English | MEDLINE | ID: mdl-24252705

ABSTRACT

Streptococcus mutans antigen I/II (AgI/II) has been widely studied as a candidate vaccine antigen against human dental caries. In this report we follow up on prior studies that indicated that anti-AgI/II immunomodulatory monoclonal antibodies (MAbs) exerted their effects by destabilizing the native protein structure and exposing cryptic epitopes. We show here that similar results can be obtained by immunizing mice with truncated polypeptides out of the context of an intra-molecular interaction that occurs within the full-length molecule and that appears to dampen the functional response against at least two important target epitopes. Putative T cell epitopes that influenced antibody specificity were identified immediately upstream of the alanine-rich repeat domain. Adherence inhibiting antibodies could be induced against two discrete domains of the protein, one corresponding to the central portion of the molecule and the other corresponding to the C-terminus.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Immunodominant Epitopes/immunology , Streptococcal Vaccines/immunology , Streptococcus mutans/immunology , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Epitopes, T-Lymphocyte/immunology , Female , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Mice , Mice, Inbred BALB C , Protein Structure, Tertiary , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/genetics
10.
Microbes Infect ; 15(4): 329-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416214

ABSTRACT

We previously reported that 5 Chlamydia muridarum antigens reacted with antisera from >90% mice urogenitally infected with C. muridarum and they are TC0660 (ABC transporter or ArtJ), TC0727 (outer membrane complex protein B or OmcB), TC0828 (macrophage infectivity potentiator or MIP), TC0726 (inclusion membrane protein or Inc) & TC0268 (hypothetical protein or HP). The orthologs of these antigens in Chlamydia trachomatis were also highly reactive with antisera from women urogenitally infected with C. trachomatis. In the current study, we evaluated these C. muridarum antigens for their ability to induce protection against a C. muridarum intravaginal challenge infection in mice. We found that only MIP induced the most pronounced protection against C. muridarum infection. The protection correlated well with robust C. muridarum MIP-specific antibody and Th1-dominant T cell responses. The MIP-immunized mice displayed significantly reduced live organism shedding from the lower genital tract and highly attenuated inflammatory pathologies in the upper genital tissues. These results demonstrate that MIP, an immunodominant antigen identified by both human and mouse antisera, may be considered a component of a multi-subunit chlamydial vaccine for inducing protective immunity.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Immunodominant Epitopes/immunology , Reproductive Tract Infections/immunology , Animals , Antigens, Bacterial/administration & dosage , Bacterial Shedding , Bacterial Vaccines/administration & dosage , Chlamydia Infections/pathology , Disease Models, Animal , Female , Immunodominant Epitopes/administration & dosage , Mice , Reproductive Tract Infections/pathology
11.
Clin Immunol ; 146(2): 120-30, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23314272

ABSTRACT

We investigated the potential of inducing additional T-cell immunity during chronic HIV-1 infection directed to subdominant HIV-1 epitopes from common HLA-supertypes. Ten treatment-naïve HIV-1-infected individuals were immunized with peptides in the adjuvant CAF01. One individual received placebo. T-cell immunogenicity was examined longitudinally by a flow cytometry (CD107a, IFNγ, TNFα, IL-2 and/or MIP1ß expression) as well as IFNγ ELISPOT. Safety was evaluated by clinical follow up combined with monitoring of biochemistry, hematology, CD4 T-cell counts and viral load. New CD4 and CD8 T-cell responses specific for one or more vaccine epitopes were induced in 10/10 vaccinees. The responses were dominated by CD107a and MIP1ß expression. There were no significant changes in HIV-1 viral load or CD4 T-cell counts. Our study demonstrates that the peptide/CAF01 vaccine is safe and that it is possible to generate new HIV-1 T-cell responses to defined epitopes in treatment-naïve HIV-1-infected individuals.


Subject(s)
Adjuvants, Immunologic/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/therapeutic use , HIV Infections/immunology , HIV Infections/therapy , Immunodominant Epitopes/therapeutic use , Peptides/therapeutic use , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , HIV Infections/prevention & control , HLA-A Antigens/administration & dosage , HLA-A Antigens/genetics , HLA-B Antigens/administration & dosage , HLA-B Antigens/genetics , HLA-C Antigens/administration & dosage , HLA-C Antigens/genetics , Humans , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Male , Middle Aged , Peptides/administration & dosage , Peptides/immunology , Single-Blind Method , Young Adult
12.
Bioorg Khim ; 38(3): 306-14, 2012.
Article in Russian | MEDLINE | ID: mdl-22997702

ABSTRACT

Multiple Sclerosis (MS) is a serve autoimmune neurodegenerative disease. Development of innovative approaches of MS treatment is of a high priority in the modern immunology and pharmacy. In the present study we showed high therapeutic efficiency of immunodominant peptides of myelin basic protein (MBP) incorporated into the monolayer mannosylated liposomes on the development of experimental autoimmune encephalomyelitis (EAE) in DA rats. MBP is a component ofoligodendrocytes' membrane, which form axonal sheath, and is one of the major autoantigens in MS. We analyzed binding pattern ofanti-MBP autoantibodies from MS patients using previously designed MBP epitope library. Utilizing the same approach we investigated pool of anti-MBP antibodies from SJL/J and C57/BL6 mice and DA rats with induced EAE. The most relevant rodent model to MS was EAE in DA rats according to the autoantibodies' binding pattern. We selected three immunodominant MBP fragments encapsulated in monolayer mannosylated liposomes for the following treatment of verified DA rodent model. MBP fragment 46-62 was the most effective in reducing of the first EAE attack, whereas MBP 124-139 and 147-160 inhibited development of pathology during remission stage. Simultaneous administration of these peptides in liposomes significantly decreased level of anti-MBP antibodies. Synergetic therapeutic effect of MBP fragments reduced integral disease score by inhibiting first EAE wave and subsequent remission, thus, our findings disclosure novel approaches for efficient treatment of Multiple Sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunodominant Epitopes/administration & dosage , Multiple Sclerosis/drug therapy , Myelin Basic Protein/administration & dosage , Nanocapsules/administration & dosage , Peptide Fragments/administration & dosage , Adult , Amino Acid Sequence , Animals , Autoantibodies/immunology , Guinea Pigs , Humans , Immunodominant Epitopes/immunology , Immunodominant Epitopes/therapeutic use , Liposomes , Mice , Mice, Inbred C57BL , Middle Aged , Molecular Sequence Data , Myelin Basic Protein/immunology , Myelin Basic Protein/therapeutic use , Nanocapsules/chemistry , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Rats , Rats, Inbred Strains
13.
PLoS One ; 7(7): e39757, 2012.
Article in English | MEDLINE | ID: mdl-22808056

ABSTRACT

The blocking of programmed death ligand-1 (PDL-1) has been shown to enhance virus-specific CD8 T cell function during chronic viral infections. Though, how PDL-1 blocking at the time of priming affects the quality of CD8 T cell response to acute infections is not well understood and remains controversial. This report demonstrates that the magnitude of the primary and secondary CD8 T cell responses to herpes simplex virus-1 (HSV-1) infection is subject to control by PDL-1. Our results showed that after footpad HSV-1 infection, PD-1 expression increases on immunodominant SSIEFARL peptide specific CD8 T cells. Additionally, post-infection, the level of PDL-1 expression also increases on CD11c+ dendritic cells. Intraperitoneal administration of anti-PDL-1 monoclonal antibody given one day prior to and three days after cutaneous HSV-1 infection, resulted in a marked increase in effector and memory CD8 T cell response to SSIEFARL peptide. This was shown by measuring the quantity and quality of SSIEFARL-specific CD8 T cells by making use of ex-vivo assays that determine antigen specific CD8 T cell function, such as intracellular cytokine assay, degranulation assay to measure cytotoxicity and viral clearance. Our results are discussed in terms of the beneficial effects of blocking PDL-1 interactions, while giving prophylactic vaccines, to generate a more effective CD8 T cell response to viral infection.


Subject(s)
Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Herpes Simplex/drug therapy , Acute Disease , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD11c Antigen/genetics , CD11c Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gene Expression/immunology , Herpes Simplex/immunology , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Humans , Immunoassay , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Immunologic Memory/drug effects , Injections, Intraperitoneal , Injections, Subcutaneous , Mice , Mice, Inbred C57BL , Oligopeptides/administration & dosage , Oligopeptides/genetics , Oligopeptides/immunology
14.
Hum Vaccin Immunother ; 8(4): 435-42, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22370518

ABSTRACT

Tuberculosis (TB) is a major cause of morbidity and mortality worldwide. This study evaluated the immune response in mice to members of the Mtb9.9 protein family. C57BL/6 mice were immunized with four different Mtb9.9 recombinant proteins to evaluate the humoral and cellular immune responses to each protein. Mouse splenocytes from these mice were stimulated in vitro for 72 h with the Mtb9.9 proteins. Concentrations of the cytokines interferon (IFN)-γ and tumor necrosis α (TNF-α) in the culture supernatants were increased compared with controls. Sera were taken from the same mice at 3 weeks after immunization and contained significantly higher levels of immunoglobulin (Ig)G2c antibody when compared with sera from control mice. Our findings indicate a bias toward T helper (Th)1-type cellular and humoral memory responses against Mtb9.9 recombinant proteins in mice. This study demonstrates that these proteins might be suitable candidates for use as subunit vaccines.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/immunology , Leukocytes, Mononuclear/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/administration & dosage , Bacterial Proteins/immunology , Female , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Mice, Inbred C57BL , Spleen/immunology , Th1 Cells/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
15.
Allergy ; 67(1): 74-82, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21950267

ABSTRACT

BACKGROUND: Allergen-specific T-cell epitopes are obvious targets for immunotherapeutic interventions in allergic disease. T-cell epitope peptides given orally may provide a practical way of inducing tolerance and preventing allergy. OBJECTIVE: This study investigates oral immunotherapy (OIT) with T-cell epitope peptides of the dominant egg-white allergen ovomucoid (Ovm) in a Balb/c mouse model of egg allergy. METHODS: Groups of mice were orally sensitized to Ovm and subsequently administered Ovm T-cell epitopes [single peptide 157-171 (SP) or multiple peptide (157-171)(3) (MP)], followed by oral challenge with Ovm. Outcomes post oral challenge were measured as clinical signs, serum histamine, antibody activity (IgG, IgE, IgG1, IgG2, IgA), cytokines (IL-4, IFN-γ, IL-12p70, IL-10, TGF-ß, and IL-17), and T regulatory cells (Tregs). RESULTS: Clinical signs were less frequent in both SP and MP groups (P ≤ 0.05). Specific IgE was less and IgA was more in both groups; however, SP-treated mice had less histamine and IgG1 and more IgG2-related antibodies indicating a bias toward the type-1 response (P ≤ 0.05). Concentration of type-2 cytokine interleukin-4 (IL-4) was significantly less in both groups and IL-12p70 and IL-10 were more in SP-treated mice (P ≤ 0.001). Interferon-γ, IL-17, and TGF-ß did not differ significantly. There was significant increase in the percentage of CD4+FOXP3+ and CD4+CD25+ cells in the SP group, indicating the significant role of Tregs in immune regulation. CONCLUSION: In summary, we demonstrated that OIT with SP and MP comprising the immunodominant regions of Ovm was safe and significantly reduced subsequent frequency of allergy to Ovm, and validated potential use of Ovm T-cell epitope as an immunoregulator.


Subject(s)
Desensitization, Immunologic/methods , Egg Hypersensitivity/prevention & control , Epitopes, T-Lymphocyte/administration & dosage , Immunodominant Epitopes/administration & dosage , Administration, Oral , Animals , Cell Separation , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Flow Cytometry , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/immunology , Mice , Mice, Inbred BALB C , Ovomucin/administration & dosage , Ovomucin/chemistry , Ovomucin/immunology , Peptides/administration & dosage , Peptides/chemistry , Protein Structure, Secondary , T-Lymphocytes, Regulatory/immunology
16.
J Immunol ; 187(3): 1184-91, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21709152

ABSTRACT

TNF-α and its two receptors (TNFR1 and 2) are known to stimulate dendritic cell (DC) maturation and T cell response. However, the specific receptor and mechanisms involved in vivo are still controversial. In this study, we show that in response to an attenuated mouse hepatitis virus infection, DCs fail to mobilize and up-regulate CD40, CD80, CD86, and MHC class I in TNFR1(-/-) mice as compared with the wild-type and TNFR2(-/-) mice. Correspondingly, virus-specific CD8 T cell response was dramatically diminished in TNFR1(-/-) mice. Adoptive transfer of TNFR1-expressing DCs into TNFR1(-/-) mice rescues CD8 T cell response. Interestingly, adoptive transfer of TNFR1-expressing naive T cells also restores DC mobilization and maturation and endogenous CD8 T cell response. These results show that TNFR1, not TNFR2, mediates TNF-α stimulation of DC maturation and T cell response to mouse hepatitis virus in vivo. They also suggest two mechanisms by which TNFR1 mediates TNF-α-driven DC maturation, as follows: a direct effect through TNFR1 expressed on immature DCs and an indirect effect through TNFR1 expressed on naive T cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Coronavirus Infections/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Murine hepatitis virus/immunology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/physiology , Adoptive Transfer , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Dendritic Cells/virology , Dose-Response Relationship, Immunologic , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Murine hepatitis virus/genetics , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/deficiency , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/physiology , Tumor Necrosis Factor-alpha/metabolism
17.
J Immunol ; 187(2): 897-904, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21653834

ABSTRACT

Septic bacterial pneumonias are a major cause of death worldwide. Several of the highest priority bioterror concerns, including anthrax, tularemia, and plague, are caused by bacteria that acutely infect the lung. Bacterial resistance to multiple antibiotics is increasingly common. Although vaccines may be our best defense against antibiotic-resistant bacteria, there has been little progress in the development of safe and effective vaccines for pulmonary bacterial pathogens. The Gram-negative bacterium Yersinia pestis causes pneumonic plague, an acutely lethal septic pneumonia. Historic pandemics of plague caused millions of deaths, and the plague bacilli's potential for weaponization sustains an ongoing quest for effective countermeasures. Subunit vaccines have failed, to date, to fully protect nonhuman primates. In mice, they induce the production of Abs that act in concert with type 1 cytokines to deliver high-level protection; however, the Y. pestis Ags recognized by cytokine-producing T cells have yet to be defined. In this study, we report that Y. pestis YopE is a dominant Ag recognized by CD8 T cells in C57BL/6 mice. After vaccinating with live attenuated Y. pestis and challenging intranasally with virulent plague, nearly 20% of pulmonary CD8 T cells recognize this single, highly conserved Ag. Moreover, immunizing mice with a single peptide, YopE(69-77), suffices to confer significant protection from lethal pulmonary challenge. These findings suggest YopE could be a valuable addition to subunit plague vaccines and provide a new animal model in which sensitive, pathogen-specific assays can be used to study CD8 T cell-mediated defense against acutely lethal bacterial infections of the lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunodominant Epitopes/immunology , Plague/prevention & control , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Acute Disease , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Clone Cells , Disease Models, Animal , Epitopes, T-Lymphocyte/administration & dosage , Immunodominant Epitopes/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Knockout , Plague/immunology , Plague/mortality , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Pneumonia, Bacterial/prevention & control , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/chemical synthesis , Vaccines, Attenuated/immunology , Vaccines, Subunit/chemical synthesis
18.
J Immunol ; 187(3): 1421-31, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21705621

ABSTRACT

Dimorphic fungi collectively account for 5-10 million new infections annually worldwide. Ongoing efforts seek to clarify mechanisms of cellular resistance to these agents and develop vaccines. A major limitation in studying the development of protective T cells in this group of organisms is the lack of tools to detect, enumerate, and characterize fungus-specific T cells during vaccination and infection. We generated a TCR transgenic mouse (Bd 1807) whose CD4(+) T cells respond to a native epitope in Blastomyces dermatitidis and also in Histoplasma capsulatum. In this study, we characterize the mouse, reveal its applications, and extend our analysis showing that 1807 cells also respond to the related dimorphic fungi Coccidioides posadasii and Paracoccidioides lutzii. On adoptive transfer into vaccinated wild-type mice, 1807 cells become activated, proliferate, and expand in the draining lymph nodes, and they differentiate into T1 effectors after trafficking to the lung upon lethal experimental challenge. Bd 1807 cells confer vaccine-induced resistance against B. dermatitidis, H. capsulatum, and C. posadasii. Transfer of naive 1807 cells at serial intervals postvaccination uncovered the prolonged duration of fungal Ag presentation. Using 1807 cells, we also found that the administration of vaccine only once induced a maximal pool of effector/memory CD4(+) cells and protective immunity by 4 wk after vaccination. The autologous adoptive transfer system described in this study reveals novel features of antifungal immunity and offers a powerful approach to study the differentiation of Ag-specific T cells responsive to multiple dimorphic fungi and the development of CD4(+) T cell memory needed to protect against fungal infection.


Subject(s)
Blastomyces/genetics , Blastomyces/immunology , Fungal Vaccines/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Coccidioidomycosis/genetics , Coccidioidomycosis/immunology , Coccidioidomycosis/pathology , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Fungal Vaccines/administration & dosage , Fungal Vaccines/genetics , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/genetics , Immunodominant Epitopes/metabolism , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Paracoccidioidomycosis/genetics , Paracoccidioidomycosis/immunology , Paracoccidioidomycosis/pathology
19.
J Immunol ; 186(7): 3847-57, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21346233

ABSTRACT

We previously reported that CD8(+) T cells are directed predominantly toward the immunodominant Her-2/neu (neu) epitope RNEU(420-429) in nontolerized FVB/N but not tolerized HER-2/neu (neu-N) mice. In this study, we screened overlapping peptides of the entire neu protein and identified six new epitopes recognized by vaccine-induced neu-N-derived T cells. Evaluation of individual nondominant responses by tetramer staining and IFN-γ secretion demonstrate that this repertoire is peripherally tolerized. To address the role that the complete CD8(+) T cell repertoire plays in vaccine-induced antitumor immunity, we created a whole-cell vaccine-expressing neu cDNA that has been mutated at the RNEU(420-429) anchor residue, thereby abrogating activation of immunodominant epitope responses. Studies comparing the mutated and nonmutated vaccines indicate that nondominant CD8(+) T cells can induce antitumor immunity when combined with regulatory T cell-depleting agents in both neu-N and FVB/N mice. Collectively, these studies demonstrate that the neu-directed T cell repertoire is not intrinsically incapable of eradicating tumors. Rather, they are suppressed by mechanisms of peripheral tolerance. Thus, these studies provide new insights into the function of the complete T cell repertoire directed toward a clinically relevant tumor Ag in tumor-bearing hosts.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Epitopes, T-Lymphocyte/metabolism , Immunodominant Epitopes/administration & dosage , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/deficiency , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Cell Line, Tumor , Dose-Response Relationship, Immunologic , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/genetics , Gene Expression Regulation, Neoplastic/immunology , Immune Tolerance/genetics , Immunodominant Epitopes/genetics , Immunodominant Epitopes/metabolism , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Transgenic , NIH 3T3 Cells , Rats , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
20.
J Clin Invest ; 121(1): 249-64, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21157037

ABSTRACT

NKT cells in the mouse recognize antigen in the context of the MHC class I-like molecule CD1d and play an important role in peripheral tolerance and protection against autoimmune and other diseases. NKT cells are usually activated by CD1d-presented lipid antigens. However, peptide recognition in the context of CD1 has also been documented, although no self-peptide ligands have been reported to date. Here, we have identified an endogenous peptide that is presented by CD1d to activate mouse NKT cells. This peptide, the immunodominant epitope from mouse collagen type II (mCII707-721), was not associated with either MHC class I or II. Activation of CD1d-restricted mCII707-721-specific NKT cells was induced via TCR signaling and classical costimulation. In addition, mCII707-721-specific NKT cells induced T cell death through Fas/FasL, in an IL-17A-independent fashion. Moreover, mCII707-721-specific NKT cells suppressed a range of in vivo inflammatory conditions, including delayed-type hypersensitivity, antigen-induced airway inflammation, collagen-induced arthritis, and EAE, which were all ameliorated by mCII707-721 vaccination. The findings presented here offer new insight into the intrinsic roles of NKT cells in health and disease. Given the results, endogenous collagen peptide activators of NKT cells may offer promise as novel therapeutics in tissue-specific autoimmune and inflammatory diseases.


Subject(s)
Antigens, CD1d/immunology , Collagen Type II/immunology , Inflammation/immunology , Inflammation/therapy , Natural Killer T-Cells/immunology , Peptide Fragments/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Animals , Antigen Presentation , Antigens, CD1d/genetics , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/therapy , Base Sequence , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Collagen Type II/administration & dosage , DNA Primers/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Fas Ligand Protein/immunology , Genes, MHC Class II , Genes, T-Cell Receptor , Hypersensitivity, Delayed/genetics , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/therapy , Immunodominant Epitopes/administration & dosage , Inflammation/genetics , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/administration & dosage , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/therapy , Th1 Cells/immunology , Th2 Cells/immunology , Vaccination , fas Receptor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...