Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(5): 613-625, 2019 05.
Article in English | MEDLINE | ID: mdl-30778243

ABSTRACT

Influenza A, B and C viruses (IAV, IBV and ICV, respectively) circulate globally and infect humans, with IAV and IBV causing the most severe disease. CD8+ T cells confer cross-protection against IAV strains, however the responses of CD8+ T cells to IBV and ICV are understudied. We investigated the breadth of CD8+ T cell cross-recognition and provide evidence of CD8+ T cell cross-reactivity across IAV, IBV and ICV. We identified immunodominant CD8+ T cell epitopes from IBVs that were protective in mice and found memory CD8+ T cells directed against universal and influenza-virus-type-specific epitopes in the blood and lungs of healthy humans. Lung-derived CD8+ T cells displayed tissue-resident memory phenotypes. Notably, CD38+Ki67+CD8+ effector T cells directed against novel epitopes were readily detected in IAV- or IBV-infected pediatric and adult subjects. Our study introduces a new paradigm whereby CD8+ T cells confer unprecedented cross-reactivity across all influenza viruses, a key finding for the design of universal vaccines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Gammainfluenzavirus/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Influenza, Human/immunology , Adolescent , Adult , Aged , Animals , CD8-Positive T-Lymphocytes/virology , Child , Epitopes, T-Lymphocyte/immunology , Female , Humans , Influenza A virus/physiology , Influenza B virus/physiology , Influenza Vaccines/immunology , Influenza, Human/virology , Gammainfluenzavirus/physiology , Male , Mice , Middle Aged , Young Adult
2.
PLoS Pathog ; 18(10): e1010891, 2022 10.
Article in English | MEDLINE | ID: mdl-36206307

ABSTRACT

Although antibody-inducing split virus vaccines (SV) are currently the most effective way to combat seasonal influenza, their efficacy can be modest, especially in immunologically-naïve individuals. We investigated immune responses towards inactivated whole influenza virus particle vaccine (WPV) formulations, predicated to be more immunogenic, in a non-human primate model, as an important step towards clinical testing in humans. Comprehensive analyses were used to capture 46 immune parameters to profile how WPV-induced responses differed to those elicited by antigenically-similar SV formulations. Naïve cynomolgus macaques vaccinated with either monovalent or quadrivalent WPV consistently induced stronger antibody responses and hemagglutination inhibition (HI) antibody titres against vaccine-matched viruses compared to SV formulations, while acute reactogenic effects were similar. Responses in WPV-primed animals were further increased by boosting with the same formulation, conversely to modest responses after priming and boosting with SV. 28-parameter multiplex bead array defined key antibody features and showed that while both WPV and SV induced elevated IgG responses against A/H1N1 nucleoprotein, only WPV increased IgG responses against A/H1N1 hemagglutinin (HA) and HA-Stem, and higher IgA responses to A/H1N1-HA after each vaccine dose. Antibodies to A/H1N1-HA and HA-Stem that could engage FcγR2a and FcγR3a were also present at higher levels after one dose of WPV compared to SV and remained elevated after the second dose. Furthermore, WPV-enhanced antibody responses were associated with higher frequencies of HA-specific B-cells and IFN-γ-producing CD4+ T-cell responses. Our data additionally demonstrate stronger boosting of HI titres by WPV following prior infection and support WPV administered as a priming dose irrespective of the follow up vaccine for the second dose. Our findings thus show that compared to SV vaccination, WPV-induced humoral responses are significantly increased in scope and magnitude, advocating WPV vaccination regimens for priming immunologically-naïve individuals and also in the event of a pandemic outbreak.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Animals , Humans , Hemagglutinins , Antibodies, Viral , Vaccination , Hemagglutination Inhibition Tests , Vaccines, Inactivated , Macaca fascicularis , Virion , Immunoglobulin A , Immunoglobulin G , Nucleoproteins
3.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055020

ABSTRACT

The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.


Subject(s)
COVID-19/virology , Nasal Mucosa/cytology , Nasal Mucosa/virology , Tissue Culture Techniques/methods , Adolescent , Adult , Angiotensin-Converting Enzyme 2/metabolism , Cell Culture Techniques , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/virology , Female , Humans , Male , Middle Aged , Models, Biological , SARS-CoV-2 , Virus Internalization
4.
Immunol Cell Biol ; 99(1): 97-106, 2021 01.
Article in English | MEDLINE | ID: mdl-32741011

ABSTRACT

Influenza remains a significant global public health burden, despite substantial annual vaccination efforts against circulating virus strains. As a result, novel vaccine approaches are needed to generate long-lasting and universal broadly cross-reactive immunity against distinct influenza virus strains and subtypes. Several new vaccine candidates are currently under development and/or in clinical trials. The successful development of new vaccines requires testing in animal models, other than mice, which capture the complexity of the human immune system. Importantly, following vaccination or challenge, the assessment of adaptive immunity at the antigen-specific level is particularly informative. In this study, using peripheral blood mononuclear cells (PBMCs) from cynomolgus macaques, we describe detection methods and in-depth analyses of influenza virus-specific B cells by recombinant hemagglutinin probes and flow cytometry, as well as the detection of influenza virus-specific CD8+ and CD4+ T cells by stimulation with live influenza A virus and intracellular cytokine staining. We highlight the potential of these assays to be used with PBMCs from other macaque species, including rhesus macaques, pigtail macaques and African green monkeys. We also demonstrate the use of a human cytometric bead array kit in detecting inflammatory cytokines and chemokines from cynomolgus macaques to assess cytokine/chemokine milieu. Overall, the detection of influenza virus-specific B and T cells, together with inflammatory responses, as described in our study, provides useful insights for evaluating novel influenza vaccines. Our data deciphering immune responses toward influenza viruses can be also adapted to understanding immunity to other infections or vaccination approaches in macaque models.


Subject(s)
Influenza Vaccines , Influenza, Human , Orthomyxoviridae Infections , Animals , Antibodies, Viral , Chlorocebus aethiops , Flow Cytometry , Hemagglutinin Glycoproteins, Influenza Virus , Humans , Leukocytes, Mononuclear , Macaca mulatta , Mice , T-Lymphocytes , Vaccination
5.
Brain Behav Immun ; 97: 371-375, 2021 10.
Article in English | MEDLINE | ID: mdl-34333113

ABSTRACT

The splanchnic anti-inflammatory pathway, the efferent arm of the endogenous inflammatory reflex, has been shown to suppress the acute inflammatory response of rats to systemic lipopolysaccharide (LPS). Here we show for the first time that this applies also to mice, and that the reflex may be engaged by a range of inflammatory stimuli. Experiments were performed on mice under deep anaesthesia. Half the animals were subjected to bilateral section of the splanchnic sympathetic nerves, to disconnect the splanchnic anti-inflammatory pathway, while the remainder underwent a sham operation. Mice were then challenged intravenously with one of three inflammatory stimuli: the toll-like receptor (TLR)-4 agonist, LPS (60 µg/kg), the TLR-3 agonist Polyinosinic:polycytidylic acid (Poly I:C, 1 mg/kg) or the TLR-2 and -6 agonist dipalmitoyl-S-glyceryl cysteine (Pam2cys, 34 µg/kg). Ninety minutes later, blood was sampled by cardiac puncture for serum cytokine analysis. The splanchnic anti-inflammatory reflex action was assessed by comparing cytokine levels between animals with cut versus those with intact splanchnic nerves. A consistent pattern emerged: Tumor necrosis factor (TNF) levels in response to all three challenges were raised by prior splanchnic nerve section, while levels of the anti-inflammatory cytokine interleukin 10 (IL-10) were reduced. The raised TNF:IL-10 ratio after splanchnic nerve section indicates an enhanced inflammatory state when the reflex is disabled. These findings show for the first time that the inflammatory reflex drives a coordinated anti-inflammatory action also in mice, and demonstrate that its anti-inflammatory action is engaged, in similar fashion, by inflammatory stimuli mimicking a range of bacterial and viral infections.


Subject(s)
Lipopolysaccharides , Splanchnic Nerves , Animals , Cytokines , Mice , Rats , Reflex , Tumor Necrosis Factor-alpha
6.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31818964

ABSTRACT

The neglected tropical disease Buruli ulcer (BU) is an infection of subcutaneous tissue with Mycobacterium ulcerans There is no effective vaccine. Here, we assessed an experimental prime-boost vaccine in a low-dose murine tail infection model. We used the enoyl reductase (ER) domain of the M. ulcerans mycolactone polyketide synthases electrostatically coupled with a previously described Toll-like receptor 2 (TLR-2) agonist-based lipopeptide adjuvant, R4Pam2Cys. Mice were vaccinated and then challenged via tail inoculation with 14 to 20 CFU of a bioluminescent strain of M. ulcerans Mice receiving either the experimental ER vaccine or Mycobacterium bovis bacillus Calmette-Guérin (BCG) were equally protected, with both groups faring significantly better than nonvaccinated animals (P < 0.05). To explore potential correlates of protection, a suite of 29 immune parameters were assessed in the mice at the end of the experimental period. Multivariate statistical approaches were used to interrogate the immune response data to develop disease-prognostic models. High levels of interleukin 2 (IL-2) and low gamma interferon (IFN-γ) produced in the spleen best predicted control of infection across all vaccine groups. Univariate logistic regression revealed vaccine-specific profiles of protection. High titers of ER-specific IgG serum antibodies together with IL-2 and IL-4 in the draining lymph node (DLN) were associated with protection induced by the ER vaccine. In contrast, high titers of IL-6, tumor necrosis factor alpha (TNF-α), IFN-γ, and IL-10 in the DLN and low IFN-γ titers in the spleen were associated with protection following BCG vaccination. This study suggests that an effective BU vaccine must induce localized, tissue-specific immune profiles with controlled inflammatory responses at the site of infection.


Subject(s)
Bacterial Vaccines/immunology , Buruli Ulcer , Mycobacterium ulcerans/immunology , Vaccination/methods , Animals , BCG Vaccine/immunology , Buruli Ulcer/immunology , Buruli Ulcer/prevention & control , Interleukins/metabolism , Mice , Multivariate Analysis
7.
Mol Pharm ; 16(5): 2037-2047, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30924661

ABSTRACT

Targeted delivery of otherwise nonimmunogenic antigens to Toll-like receptors (TLRs) expressed on dendritic cells (DCs) has proven to be an effective means of improving immunogenicity. For this purpose, we have used a branched cationic lipopeptide, R4Pam2Cys, which is an agonist for TLR2 and enables electrostatic association with antigen for this purpose. Here, we compare the immunological properties of ovalbumin formulated with different geometrical configurations of R4Pam2Cys. Our results demonstrate that notwithstanding the presence of the same adjuvant, branched forms of R4Pam2Cys are more effective at inducing immune responses than are linear geometries. CD8+ T-cell-mediated responses are particularly improved, resulting in significantly higher levels of antigen-specific cytokine secretion and cytolysis of antigen-bearing target cells in vivo. The results correlate with the ability of branched R4Pam2Cys conformations to encourage higher levels of DC maturation and facilitate superior antigen uptake, leading to increased production of proinflammatory cytokines. These differences are not attributable to particle size because both branched and linear lipopeptides associate with antigen-forming complexes of similar size, but rather the ability of branched lipopeptides to induce more efficient TLR2-mediated cell signaling. Branched lipopeptides are also more resistant to trypsin-mediated proteolysis, suggesting greater stability than their linear counterparts. The branched lipopeptide facilitates presentation of antigen more efficiently to CD8+ T cells, resulting in rapid cell division and upregulation of early cell surface activation markers. These results as well as cognate recognition of Pam2Cys by TLR2 indicate that the adjuvant's efficiency is also dependent on its geometry.


Subject(s)
Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Lipopeptides/chemistry , Lipopeptides/pharmacology , Ovalbumin/chemistry , Toll-Like Receptor 2/agonists , Animals , Antibodies/blood , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/metabolism , HEK293 Cells , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Particle Size , Protein Conformation , Signal Transduction/drug effects , Static Electricity , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/metabolism
8.
J Transl Med ; 16(1): 279, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30305097

ABSTRACT

BACKGROUND: The clinical trials conducted at Chingleput India suggest that BCG fails to protect against tuberculosis (TB) in TB-endemic population. Recent studies advocate that non-tuberculous mycobacteria and latent Mycobacterium tuberculosis (Mtb) infection interferes in the antigen processing and presentation of BCG in inducing protective immunity against Mtb. Thereby, indicating that any vaccine that require extensive antigen processing may not be efficacious in TB-endemic zones. Recently, we have demonstrated that the vaccine candidate L91, which is composed of lipidated promiscuous MHC-II binder epitope, derived from latency associated Acr1 antigen of Mtb is immunogenic in the murine and Guinea pig models of TB and conferred better protection than BCG against Mtb. METHODS: In this study, we have used a multi-stage based bi-epitope vaccine, namely L4.8, comprising of MHC-I and MHC-II binding peptides of active (TB10.4) and latent (Acr1) stages of Mtb antigens, respectively. These peptides were conjugated to the TLR-2 agonist Pam2Cys. RESULTS: L4.8 significantly elicited both CD8 T cells and CD4 T cells immunity, as evidenced by increase in the enduring polyfunctional CD8 T cells and CD4 T cells. L4.8 efficiently declined Mtb-burden and protected animals better than BCG and L91, even at the late stage of Mtb infection. CONCLUSIONS: The BCG-L4.8 prime boost strategy imparts a better protection against TB than the BCG alone. This study emphatically denotes that L4.8 can be a promising future vaccine candidate for controlling active and latent TB.


Subject(s)
BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Lipids/chemistry , Mycobacterium tuberculosis/immunology , Animals , Female , Immunity , Immunization , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice, Inbred BALB C , T-Lymphocytes, Cytotoxic/immunology , Tuberculosis/immunology
9.
J Transl Med ; 15(1): 201, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28985739

ABSTRACT

BACKGROUND: The current BCG vaccine induces only short-term protection against Mycobacterium tuberculosis (Mtb), suggesting its failure to generate long-lasting memory T cells. Previously, we have demonstrated that a self-adjuvanting peptide of Mtb (L91), successfully generated enduring memory Th1 cells. Consequently, we investigated if L91 was able to recuperate BCG potency in perpetuating the generation of memory T cells and protection against Mtb infected mice. METHODS: In the present study, we evaluated the potency of a self adjuvanting Mtb peptide vaccine L91 in invigorating BCG immune response against Mtb in mice. Female BALB/c mice were immunized with BCG. Later, they were boosted twice with L91 or an antigenically irrelevant lipidated influenza virus hemagglutinin peptide (LH). Further, PBMCs obtained from BCG vaccinated healthy subjects were cultured in vitro with L91. T cell responses were determined by surface markers and intracellular cytokine staining. Secretion of cytokines was estimated in the culture supernatants (SNs) by ELISA. RESULTS: Compared to the BCG-vaccinated controls, L91 booster significantly enhanced the percentage of memory Th1 cells and Th17 cells and reduced the mycobacterial burden in BCG primed and L91-boosted (BCG-L91) group, even after 229 days of BCG vaccination. Further, substantial augmentation in the central (CD44hiCD62LhiCD127hi) and effector memory (CD44hiCD62LloCD127lo) CD4 T cells was detected. Furthermore, greater frequency of polyfunctional Th1 cells (IFN-γ+TNF-α+) and Th17 cells (IFN-γ+IL-17A+) was observed. Importantly, BCG-L91 successfully prevented CD4 T cells from exhaustion by decreasing the expression of PD-1 and Tim-3. Additionally, augmentation in the frequency of Th1 cells, Th17 cells and memory CD4 T cells was observed in the PBMCs of the BCG-vaccinated healthy individuals following in vitro stimulation with L91. CONCLUSIONS: Our study demonstrated that L91 robustly reinvigorate BCG potency to invoke enduring protection against Mtb. This novel vaccination stratagem involving BCG-priming followed by L91-boosting can be a future prophylactic measure to control TB.


Subject(s)
BCG Vaccine/immunology , Immunity , Immunologic Memory , Lipids/chemistry , Mycobacterium tuberculosis/immunology , Peptides/pharmacology , Protective Agents/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/metabolism , Cell Proliferation/drug effects , Female , Humans , Immunity/drug effects , Immunologic Memory/drug effects , Lymphocyte Activation/drug effects , Mice, Inbred BALB C , Mycobacterium tuberculosis/drug effects , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Phenotype , Receptors, Chemokine/metabolism , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Tuberculosis/immunology , Tuberculosis/microbiology
10.
Ophthalmology ; 124(1): 43-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27817918

ABSTRACT

PURPOSE: To assess the efficacy of 2 forms of oral long-chain omega-3 (ω-3) essential fatty acid (EFA) supplements, phospholipid (krill oil) and triacylglyceride (fish oil), for treating dry eye disease (DED). DESIGN: Randomized, double-masked, placebo-controlled clinical trial. PARTICIPANTS: This study was conducted at a single site and involved 60 participants with mild to moderate DED who were randomized (1:1:1) to 1 of 3 groups: placebo (olive oil), krill oil, or fish oil supplements. METHODS: Participants received 1 of the 3 interventions: placebo (olive oil 1500 mg/day), krill oil (945 mg/day eicosapentaenoic acid [EPA], + 510 mg/day docosahexaenoic acid [DHA]), or fish oil (1000 mg/day EPA + 500 mg/day DHA) for 90 days, with monthly study visits. MAIN OUTCOME MEASURES: Primary outcome measures were mean change in (1) tear osmolarity and (2) DED symptoms (Ocular Surface Disease Index [OSDI] score) between days 1 and 90. Secondary outcomes included mean change in key clinical signs (tear stability, tear production, ocular surface staining, bulbar and limbal redness, tear volume, anterior blepharitis, meibomian gland capping) and tear inflammatory cytokine levels. RESULTS: In total, 54 participants completed the study. At day 90, tear osmolarity was reduced from baseline with both krill oil (mean ± standard error of the mean: -18.6±4.5 mOsmol/l; n = 18; P < 0.001) and fish oil (-19.8±3.9 mOsmol/l; n = 19; P < 0.001) supplements, compared with placebo (-1.5±4.4 mOsmol/l; n = 17). OSDI score was significantly reduced at day 90 relative to baseline in the krill oil group only, compared with placebo (-18.6±2.4 vs. -10.5±3.3; P = 0.02). At day 90, there were also relative improvements in tear breakup time and ocular bulbar redness, compared with placebo, for both forms of ω-3 EFAs. Basal tear levels of the proinflammatory cytokine interleukin 17A were significantly reduced in the krill oil group, compared with placebo, at day 90 (-27.1±10.9 vs. 46.5±30.4 pg/ml; P = 0.02). CONCLUSIONS: A moderate daily dose of both forms of long-chain ω-3 EFAs, for 3 months, resulted in reduced tear osmolarity and increased tear stability in people with DED. Omega-3 EFAs in a predominantly phospholipid form (krill oil) may confer additional therapeutic benefit, with improvements in DED symptoms and lower basal tear levels of interleukin 17A, relative to placebo.


Subject(s)
Dietary Supplements , Dry Eye Syndromes/drug therapy , Fatty Acids, Omega-3/therapeutic use , Fish Oils/therapeutic use , Phospholipids/therapeutic use , Adult , Animals , Cytokines/metabolism , Double-Blind Method , Dry Eye Syndromes/metabolism , Dry Eye Syndromes/physiopathology , Euphausiacea , Female , Humans , Male , Middle Aged , Osmolar Concentration , Tears/chemistry , Tears/metabolism , Visual Acuity
11.
Amino Acids ; 49(10): 1691-1704, 2017 10.
Article in English | MEDLINE | ID: mdl-28718065

ABSTRACT

The lipopeptide, R4Pam2Cys, associates electrostatically with soluble protein antigens and significantly enhances their ability to induce protective humoral and cell-mediated responses. We demonstrate that antibody titers elicited by the antigen ovalbumin (OVA) associated with R4Pam2Cys are higher than those elicited by OVA in the presence of alum and comparable to those elicited by OVA formulated with complete Freund's adjuvant (CFA). The hierarchy of anti-OVA antibody avidities was CFA > R4Pam2Cys = alum. Each of the three adjuvants facilitated IgG class-switching with significantly more IgG1 elicited by OVA when formulated with R4Pam2Cys. The effects of substituting naturally occurring L-stereoisomers of the cationic residues within R4Pam2Cys with D-stereoisomers revealed that substitution did not affect the ability of R4Pam2Cys to stimulate dendritic cell maturation or its ability to elicit antibody production when used as an adjuvant. Minor detrimental effects were, however, observed in the ensuing CD8+ T cell responses suggesting that the use of D-amino acids affects antigen processing and presentation pathways involved in generation of cell-mediated immunity at least when facilitated through TLR2. Both D- and L-forms were found to be resistant to digestion by trypsin, indicating resistance of the branched structure to protease activity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lipopeptides , Ovalbumin , Toll-Like Receptor 2/agonists , Animals , Female , Immunoglobulin G/immunology , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/immunology , Lipopeptides/pharmacology , Mice , Mice, Inbred BALB C , Ovalbumin/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Static Electricity , Structure-Activity Relationship , Toll-Like Receptor 2/immunology
12.
J Gen Virol ; 97(12): 3205-3214, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27902386

ABSTRACT

FluMist has been used in children and adults for more than 10 years. As pre-existing CD8+ T cell memory pools can provide heterologous immunity against distinct influenza viruses, it is important to understand influenza-specific CD8+ T cell responses elicited by different live attenuated influenza virus (LAIV) regimens. In this study, we immunized mice intranasally with two different doses of live-attenuated PR8 virus (PR8 ts, H1N1), low and high, and then assessed protective efficacy by challenging animals with heterosubtypic X31-H3N2 virus at 6 weeks post-vaccination. Different LAIV doses elicited influenza-specific CD8+ T cell responses in lungs and spleen, but unexpectedly not in bronchoalveolar lavage. Interestingly, the immunodominance hierarchy at the acute phase after immunization varied depending on the LAIV dose; however, these differences disappeared at 6 weeks post-vaccination, resulting in generation of comparable CD8+ T cell memory pools. After vaccination with either dose, sufficient numbers of specific CD8+ T cells were generated for recall and protection of mice against heterosubtypic H1N1→H3N2 challenge. As a result, immunized mice displayed reduced weight loss, diminished inflammatory responses and lower viral titres in lungs, when compared to unvaccinated animals. Interestingly, the higher dose led to enhanced viral clearance on day 5 post-challenge, though this was not associated with increased CD8+ T cell responses, but with higher levels of non-neutralizing antibodies against the priming virus. Our study suggests that, while different LAIV doses result in distinct immune profiles, even a low dose produces sufficient protective CD8+ T cell memory against challenge infection, though the high dose results in more rapid viral clearance and reduced inflammation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Vaccines, Attenuated/administration & dosage , Animals , Antibodies, Viral/immunology , Humans , Immunologic Memory , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Lung/virology , Mice , Mice, Inbred C57BL , Vaccines, Attenuated/immunology
13.
Immunol Cell Biol ; 94(2): 169-76, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26272554

ABSTRACT

We have previously shown that intranasal administration of the Toll-like receptor-2 agonist, S-(2,3-bis(palmitoyloxy)propyl) cysteine (Pam2Cys), provides immediate and antigen independent protection against challenge with influenza virus. Here we characterize the cellular pulmonary environments of mice which had either been treated with Pam2Cys or placebo and then challenged with influenza virus. We show that Pam2Cys treatment results in the influx of innate immune cells into the lungs and that depletion of phagocytic cells from this influx using clodronate-loaded liposomes caused a reduction in the number of interstitial macrophages and monocytes. This resulted in abolition of the protective effect indicating the importance of this cellular subset in Pam2Cys-mediated protection.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Lipopeptides/administration & dosage , Lung/drug effects , Orthomyxoviridae Infections/drug therapy , Toll-Like Receptor 2/agonists , Administration, Intranasal , Animals , Cell Movement/drug effects , Clodronic Acid/administration & dosage , Female , Immunity, Innate/drug effects , Lipopeptides/pharmacology , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/immunology
14.
Immunol Cell Biol ; 94(8): 729-40, 2016 09.
Article in English | MEDLINE | ID: mdl-27101922

ABSTRACT

CD4 T follicular helper cells (TFH) are critical in the generation of potent and long-lived B-cell responses after viral infection. However, the factors that dictate the generation and maintenance of these cells are not fully understood. Here we use adoptive transfer of OTII T-cell receptor transgenic CD4 T cells, followed by infection with recombinant influenza A virus (IAV), as a means of identifying and tracking virus-specific CD4(+) T-cell responses. We show that T-cell competition within the virus-specific CD4 T-cell pool induced by IAV infection limits the proliferation and differentiation of IAV-specific CD4(+) TFH responses. In particular, increased T-cell competition for antigen results in a diminished IAV-specific TFH CD4 T-cell responses, particularly germinal center TFH responses. Strikingly, competition in the form of preexisting cellular immunity generated by heterosubtypic IAV immunization limits de novo CD4 T-cell responses in secondary lymphoid tissue. Taken together, these data show a profound linkage between antigen availability and promotion of TFH CD4(+) T-cell responses in response to infection. These data suggest that competition within the CD4 T-cell pool limits TFH responses and may be an important regulatory mechanism for controlling immunity.


Subject(s)
Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , Cell Differentiation , Cell Division , Cell Proliferation , Clone Cells , Germinal Center/cytology , Immunity , Lymph Nodes/pathology , Mice, Inbred C57BL , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/pathology , Phenotype , Species Specificity
15.
Immunol Cell Biol ; 94(1): 101-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26134269

ABSTRACT

When administered prophylactically, we show that the Toll-like receptor-2 (TLR-2) agonist PEG-Pam2Cys (pegylated-S-(2,3-bis(palmitoyloxy)propyl)cysteine) not only mediates potent anti-viral activity against influenza virus but also reduces the impact of secondary infections with Streptococcus pneumoniae (the pneumococcus) by reducing (i) pulmonary viral and bacterial burdens, (ii) the levels of proinflammatory cytokines that normally accompany influenza and S. pneumoniae secondary infections and (iii) the vascular permeability of the pulmonary tract that can allow bacterial invasion of the blood in mice. We also show that an inactivated detergent-disrupted influenza virus vaccine formulated with the Pam2Cys-based adjuvant R4-Pam2Cys provides the host with both immediate and long-term protection against secondary pneumococcal infections following influenza virus infection through innate and specific immune mechanisms, respectively. Vaccinated animals generated influenza virus-specific immune responses that provided the host with long-term protection against influenza virus and its sequelae. This vaccine, which generates an immediate response, provides an additional countermeasure, which is ideal for use even in the midst of an influenza outbreak.


Subject(s)
Orthomyxoviridae Infections/complications , Pneumococcal Infections/complications , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Capillary Permeability/drug effects , Chemokines/metabolism , Epitopes/immunology , Female , Immunity/drug effects , Inflammation Mediators/metabolism , Lipopeptides/pharmacology , Lung/drug effects , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Orthomyxoviridae/drug effects , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/immunology , Pneumococcal Infections/blood , Pneumococcal Infections/immunology , Polyethylene Glycols/chemistry , Streptococcus pneumoniae/immunology , Survival Analysis , Vaccination
16.
J Immunol ; 192(9): 4061-8, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24696232

ABSTRACT

Virus-specific CTL responses typically fall into reproducible hierarchies with particular epitopes eliciting either immunodominant or subdominant responses after viral challenge. The recently acquired capacity to directly enumerate naive CTL precursors (CTLps) in both mice and humans has implicated CTLp frequency as a key predictor of immune response magnitude after Ag challenge. However, recent studies have indicated that naive CTLp frequencies do not necessarily predict the size of the Ag-driven response, indicating an important role for differential CTLp recruitment and/or expansion. This study characterizes the early emergence of various influenza epitope-specific CTL responses at multiple sites in C57BL/6 mice, and probes the role of Ag dose and TCR avidity in dictating immune response hierarchies. Despite large naive CTLp numbers, subdominance was found to arise largely as a consequence of the abrupt and premature cessation of CTL proliferation, at least for one epitope specificity. Investigation into the possible drivers of the poor proliferation observed for subdominant specificities showed that the immunodominance hierarchy endured irrespective of epitope abundance, and correlated with the prevalence of high-avidity T cells in both the naive and immune compartments. Our study strongly indicates that the quality, and not simply the quantity, of antiviral CTLs dictate response magnitude.


Subject(s)
Immunodominant Epitopes/immunology , Influenza A virus/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Separation , Cytotoxicity, Immunologic/immunology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL
17.
Proc Natl Acad Sci U S A ; 110(14): 5570-5, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23493558

ABSTRACT

A reverse-genetics approach has been used to probe the mechanism underlying immune escape for influenza A virus-specific CD8(+) T cells responding to the immunodominant D(b)NP366 epitope. Engineered viruses with a substitution at a critical residue (position 6, P6M) all evaded recognition by WT D(b)NP366-specific CD8(+) T cells, but only the NPM6I and NPM6T mutants altered the topography of a key residue (His155) in the MHC class I binding site. Following infection with the engineered NPM6I and NPM6T influenza viruses, both mutations were associated with a substantial "hole" in the naïve T-cell receptor repertoire, characterized by very limited T-cell receptor diversity and minimal primary responses to the NPM6I and NPM6T epitopes. Surprisingly, following respiratory challenge with a serologically distinct influenza virus carrying the same mutation, preemptive immunization against these escape variants led to the generation of secondary CD8(+) T-cell responses that were comparable in magnitude to those found for the WT NP epitope. Consequently, it might be possible to generate broadly protective T-cell immunity against commonly occurring virus escape mutants. If this is generally true for RNA viruses (like HIV, hepatitis C virus, and influenza) that show high mutation rates, priming against predicted mutants before an initial encounter could function to prevent the emergence of escape variants in infected hosts. That process could be a step toward preserving immune control of particularly persistent RNA viruses and may be worth considering for future vaccine strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immune Evasion/genetics , Influenza A virus/genetics , Influenza A virus/immunology , Models, Molecular , beta 2-Microglobulin/chemistry , Animals , Binding Sites/genetics , CD8-Positive T-Lymphocytes/virology , Crystallization , Epitopes, T-Lymphocyte/genetics , Flow Cytometry , Genes, MHC Class I/genetics , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , beta 2-Microglobulin/immunology
18.
Crit Rev Microbiol ; 41(3): 389-98, 2015.
Article in English | MEDLINE | ID: mdl-24495096

ABSTRACT

Vaccines have been successful for global eradication or control of dreaded diseases such as smallpox, diphtheria, tetanus, yellow fever, whooping cough, polio, and measles. Unfortunately, this success has not been achieved for controlling tuberculosis (TB) worldwide. Bacillus Calmette Guérin (BCG) is the only available vaccine against TB. Paradoxically, BCG has deciphered success in the Western world but has failed in TB-endemic areas. In this article, we highlight and discuss the aspects of immunity responsible for controlling Mycobacterium tuberculosis infection and factors responsible for the failure of BCG in TB-endemic countries. In addition, we also suggest strategies that contribute toward the development of successful vaccine in protecting populations where BCG has failed.


Subject(s)
Adaptive Immunity/immunology , BCG Vaccine/immunology , Immunity, Innate/immunology , Tuberculosis, Pulmonary/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/prevention & control
19.
Immunol Cell Biol ; 92(4): 377-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24394993

ABSTRACT

We have previously shown that the immunogenicity of protein antigens can be significantly enhanced if electrostatically associated with the Toll-like receptor-2 agonist-based lipopeptide R4Pam2Cys. The precise mechanisms and effectiveness of the cytotoxic T-lymphocyte (CTL)-mediated response facilitated by this agonist, however, have not been studied. Here we show that priming by dendritic cells (DCs) in the draining lymph nodes of animals vaccinated with antigen delivered using R4Pam2Cys results in significantly improved T-cell proliferation and induces their differentiation into polyfunctional effector CTLs characterised by granzyme B expression and the ability to secrete interferon-γ, interleukin-2 and tumor necrosis factor-α 7 days after vaccination. After 30 days, frequencies of antigen-specific CD62(low)CD127(high) (effector memory), CD62(high)CD127(high) (central memory) and CD43(low)CD27(high) CD8(+) T cells, a phenotype associated with strong recall responses against respiratory infections, are also increased compared with responses obtained with antigens formulated in the adjuvants Alum (alhydrogel) and CFA (complete Freund's adjuvant). The phenotypic changes observed in these mice vaccinated using R4Pam2Cys further correlated with their ability to recall specific T cells into the lung to mediate the reduction of pulmonary viral titres following challenge with a chimeric influenza virus containing the K(b)OVA257-264 epitope compared with animals vaccinated using Alum or CFA. The findings from this study not only demonstrate that better T-cell responses can be elicited using R4Pam2Cys compared with classically utilised adjuvants but also highlight the potential effectiveness of this lipopeptide-based adjuvant particularly against viral infections that require resolution through cell-mediated immunity.


Subject(s)
Adjuvants, Immunologic/pharmacology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/drug effects , Toll-Like Receptor 2/agonists , Animals , Antigen Presentation/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Proliferation/drug effects , Lipopeptides/pharmacology , Mice, Inbred C57BL , Ovalbumin/immunology , Toll-Like Receptor 2/metabolism , Vaccination
20.
J Immunol ; 188(9): 4158-70, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22467654

ABSTRACT

NK cells infiltrate human herpetic lesions, but their role has been underexplored. HSV can stimulate innate immune responses via surface TLR2, which is expressed on monocyte-derived dendritic cells (DCs) and NK cells. In this study, UV-inactivated HSV1/2 and immunodominant HSV2 glycoprotein D peptides conjugated to the TLR2 agonist dipalmitoyl-S-glyceryl cysteine stimulated CD4 T lymphocyte IFN-γ responses within PBMCs or in coculture with monocyte-derived DCs. NK cells contributed markedly to the PBMC responses. Furthermore, NK cells alone were activated directly by both Ags, also upregulating HLA-DR and HLA-DQ and then they activated autologous CD4 T lymphocytes. Using Transwells, Ag-stimulated NK cells and CD4 T lymphocytes were shown to interact through both cell-to-cell contact and cytokines, differing in relative importance in different donors. A distinct immunological synapse between Ag-stimulated NK cells and CD4 T lymphocytes was observed, indicating the significance of their cell-to-cell contact. A large proportion (57%) of NK cells was also in contact with CD4 T lymphocytes in the dermal infiltrate of human recurrent herpetic lesions. Thus, NK cells stimulated by TLR2-activating HSV Ags can present Ag alone or augment the role of DCs in vitro and perhaps in herpetic lesions or draining lymph nodes. In addition to DCs, NK cells should be considered as targets for adjuvants during HSV vaccine development.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Herpesvirus 1, Human/immunology , Herpesvirus 2, Human/immunology , Lymphocyte Activation , Toll-Like Receptor 2/immunology , Viral Envelope Proteins/immunology , Antigens, Viral/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/drug effects , Cell Communication/immunology , Cells, Cultured , Coculture Techniques , Dermis/immunology , Dermis/metabolism , Dermis/virology , Female , HLA-DQ Antigens , HLA-DR Antigens , Herpes Genitalis/immunology , Herpes Genitalis/metabolism , Herpes Simplex Virus Vaccines/immunology , Herpesvirus 1, Human/metabolism , Herpesvirus 2, Human/metabolism , Humans , Immunity, Innate/drug effects , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural , Lipopeptides/pharmacology , Male , Toll-Like Receptor 2/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL