Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cancer Immunol Res ; 9(6): 612-623, 2021 06.
Article in English | MEDLINE | ID: mdl-33674358

ABSTRACT

Merkel cell carcinomas (MCC) are immunogenic skin cancers associated with viral infection or UV mutagenesis. To study T-cell infiltrates in MCC, we analyzed 58 MCC lesions from 39 patients using multiplex-IHC/immunofluorescence (m-IHC/IF). CD4+ or CD8+ T cells comprised the majority of infiltrating T lymphocytes in most tumors. However, almost half of the tumors harbored prominent CD4/CD8 double-negative (DN) T-cell infiltrates (>20% DN T cells), and in 12% of cases, DN T cells represented the majority of T cells. Flow cytometric analysis of single-cell suspensions from fresh tumors identified DN T cells as predominantly Vδ2- γδ T cells. In the context of γδ T-cell inflammation, these cells expressed PD-1 and LAG3, which is consistent with a suppressed or exhausted phenotype, and CD103, which indicates tissue residency. Furthermore, single-cell RNA sequencing (scRNA-seq) identified a transcriptional profile of γδ T cells suggestive of proinflammatory potential. T-cell receptor (TCR) analysis confirmed clonal expansion of Vδ1 and Vδ3 clonotypes, and functional studies using cloned γδ TCRs demonstrated restriction of these for CD1c and MR1 antigen-presenting molecules. On the basis of a 13-gene γδ T-cell signature derived from scRNA-seq analysis, gene-set enrichment on bulk RNA-seq data showed a positive correlation between enrichment scores and DN T-cell infiltrates. An improved disease-specific survival was evident for patients with high enrichment scores, and complete responses to anti-PD-1/PD-L1 treatment were observed in three of four cases with high enrichment scores. Thus, γδ T-cell infiltration may serve as a prognostic biomarker and should be explored for therapeutic interventions.See related Spotlight on p. 600.


Subject(s)
Carcinoma, Merkel Cell/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/mortality , Cell Line , Computational Biology , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Male , Middle Aged , Prognosis , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Survival Analysis
2.
Cell Rep ; 34(4): 108674, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503413

ABSTRACT

Naive and memory T cells are maintained in a quiescent state, yet capable of rapid response and differentiation to antigen challenge via molecular mechanisms that are not fully understood. In naive cells, the deletion of Foxo1 following thymic development results in the increased expression of multiple AP-1 family members, rendering T cells less able to respond to antigenic challenge. Similarly, in the absence of FOXO1, post-infection memory T cells exhibit the characteristics of extended activation and senescence. Age-based analysis of human peripheral T cells reveals that levels of FOXO1 and its downstream target, TCF7, are inversely related to host age, whereas the opposite is found for AP-1 factors. These characteristics of aging also correlate with the formation of T cells manifesting features of cellular senescence. Our work illustrates a role for FOXO1 in the active maintenance of stem-like properties in T cells at the timescales of acute infection and organismal life span.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Forkhead Box Protein O1/immunology , Adult , Aged , Aged, 80 and over , Animals , CD8-Positive T-Lymphocytes/cytology , Cellular Senescence/physiology , Humans , Mice , Mice, Inbred C57BL , Middle Aged , Young Adult
3.
PLoS Pathog ; 16(8): e1008714, 2020 08.
Article in English | MEDLINE | ID: mdl-32750095

ABSTRACT

Seasonal influenza virus infections cause 290,000-650,000 deaths annually and severe morbidity in 3-5 million people. CD8+ T-cell responses towards virus-derived peptide/human leukocyte antigen (HLA) complexes provide the broadest cross-reactive immunity against human influenza viruses. Several universally-conserved CD8+ T-cell specificities that elicit prominent responses against human influenza A viruses (IAVs) have been identified. These include HLA-A*02:01-M158-66 (A2/M158), HLA-A*03:01-NP265-273, HLA-B*08:01-NP225-233, HLA-B*18:01-NP219-226, HLA-B*27:05-NP383-391 and HLA-B*57:01-NP199-207. The immunodominance hierarchies across these universal CD8+ T-cell epitopes were however unknown. Here, we probed immunodominance status of influenza-specific universal CD8+ T-cells in HLA-I heterozygote individuals expressing two or more universal HLAs for IAV. We found that while CD8+ T-cell responses directed towards A2/M158 were generally immunodominant, A2/M158+CD8+ T-cells were markedly diminished (subdominant) in HLA-A*02:01/B*27:05-expressing donors following ex vivo and in vitro analyses. A2/M158+CD8+ T-cells in non-HLA-B*27:05 individuals were immunodominant, contained optimal public TRBV19/TRAV27 TCRαß clonotypes and displayed highly polyfunctional and proliferative capacity, while A2/M158+CD8+ T cells in HLA-B*27:05-expressing donors were subdominant, with largely distinct TCRαß clonotypes and consequently markedly reduced avidity, proliferative and polyfunctional efficacy. Our data illustrate altered immunodominance patterns and immunodomination within human influenza-specific CD8+ T-cells. Accordingly, our work highlights the importance of understanding immunodominance hierarchies within individual donors across a spectrum of prominent virus-specific CD8+ T-cell specificities prior to designing T cell-directed vaccines and immunotherapies, for influenza and other infectious diseases.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , HLA-B27 Antigen/genetics , Immunodominant Epitopes/immunology , Influenza, Human/immunology , Adult , Aged , Epitopes, T-Lymphocyte/immunology , Female , HLA-B27 Antigen/immunology , Humans , Immunodominant Epitopes/genetics , Immunologic Memory , Influenza A virus/physiology , Influenza, Human/genetics , Influenza, Human/virology , Male , Middle Aged , Young Adult
4.
Nat Cancer ; 1(11): 1054-1065, 2020 11.
Article in English | MEDLINE | ID: mdl-35122066

ABSTRACT

Antibody-mediated modulation of major histocompatibility complex (MHC) molecules, or MHC class I-like molecules, could constitute an effective immunotherapeutic approach. We describe how single-domain antibodies (VHH), specific for the human MHC class I-like molecule CD1d, can modulate the function of CD1d-restricted T cells and how one VHH (1D12) specifically induced strong type I natural killer T (NKT) cell activation. The crystal structure of the VHH1D12-CD1d(α-GalCer)-NKT T-cell receptor (TCR) complex revealed that VHH1D12 simultaneously contacted CD1d and the type I NKT TCR, thereby stabilizing this interaction through intrinsic bispecificity. This led to greatly enhanced type I NKT cell-mediated antitumor activity in in vitro, including multiple myeloma and acute myeloid leukemia patient-derived bone marrow samples, and in vivo models. Our findings underscore the versatility of VHH molecules in targeting composite epitopes, in this case consisting of a complexed monomorphic antigen-presenting molecule and an invariant TCR, and represent a generalizable antitumor approach.


Subject(s)
Receptors, Antigen, T-Cell , Antigens, CD1d/chemistry , Humans , Receptors, Antigen, T-Cell/chemistry
5.
Nat Commun ; 9(1): 824, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29483513

ABSTRACT

Severe influenza A virus (IAV) infection is associated with immune dysfunction. Here, we show circulating CD8+ T-cell profiles from patients hospitalized with avian H7N9, seasonal IAV, and influenza vaccinees. Patient survival reflects an early, transient prevalence of highly activated CD38+HLA-DR+PD-1+ CD8+ T cells, whereas the prolonged persistence of this set is found in ultimately fatal cases. Single-cell T cell receptor (TCR)-αß analyses of activated CD38+HLA-DR+CD8+ T cells show similar TCRαß diversity but differential clonal expansion kinetics in surviving and fatal H7N9 patients. Delayed clonal expansion associated with an early dichotomy at a transcriptome level (as detected by single-cell RNAseq) is found in CD38+HLA-DR+CD8+ T cells from patients who succumbed to the disease, suggesting a divergent differentiation pathway of CD38+HLA-DR+CD8+ T cells from the outset during fatal disease. Our study proposes that effective expansion of cross-reactive influenza-specific TCRαß clonotypes with appropriate transcriptome signatures is needed for early protection against severe influenza disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Clonal Selection, Antigen-Mediated/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Influenza, Human/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets/immunology , Transcriptome/immunology , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cohort Studies , Critical Illness , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Hospitalization , Humans , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/genetics , Influenza, Human/mortality , Influenza, Human/virology , Lymphocyte Activation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Survival Analysis , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology
6.
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
8.
J Virol ; 90(15): 6936-6947, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27226365

ABSTRACT

UNLABELLED: Novel influenza viruses often cause differential infection patterns across different age groups, an effect that is defined as heterogeneous demographic susceptibility. This occurred during the A/H2N2 pandemic, when children experienced higher influenza attack rates than adults. Since the recognition of conserved epitopes across influenza subtypes by CD8(+) cytotoxic T lymphocytes (CTLs) limit influenza disease, we hypothesized that conservation of CTL antigenic peptides (Ag-p) in viruses circulating before the pH2N2-1957 may have resulted in differential CTL immunity. We compared viruses isolated in the years preceding the pandemic (1941 to 1957) to which children and adults were exposed to viruses circulating decades earlier (1918 to 1940), which could infect adults only. Consistent with phylogenetic models, influenza viruses circulating from 1941 to 1957, which infected children, shared with pH2N2 the majority (∼89%) of the CTL peptides within the most immunogenic nucleoprotein, matrix 1, and polymerase basic 1, thus providing evidence for minimal pH2N2 CTL escape in children. Our study, however, identified potential CTL immune evasion from pH2N2 irrespective of age, within HLA-A*03:01(+) individuals for PB1471-L473V/N476I variants and HLA-B*15:01(+) population for NP404-414-V408I mutant. Further experiments using the murine model of B-cell-deficient mice showed that multiple influenza infections resulted in superior protection from influenza-induced morbidity, coinciding with accumulation of tissue-resident memory CD8(+) T cells in the lung. Our study suggests that protection against H2N2-1957 pandemic influenza was most likely linked to the number of influenza virus infections prior to the pandemic challenge rather than differential preexisting CTL immunity. Thus, the regimen of a CTL-based vaccine/vaccine-component may benefit from periodic boosting to achieve fully protective, asymptomatic influenza infection. IMPORTANCE: Due to a lack of cross-reactive neutralizing antibodies, children are particularly susceptible to influenza infections caused by novel viral strains. Preexisting T cell immunity directed at conserved viral regions, however, can provide protection against influenza viruses, promote rapid recovery and better clinical outcomes. When we asked whether high susceptibility of children (compared to adults) to the pandemic H2N2 influenza strain was associated with immune evasion from T-cell immunity, we found high conservation within T-cell antigenic regions in pandemic H2N2. However, the number of influenza infections prior to the challenge was linked to protective, asymptomatic infections and establishment of tissue-resident memory T cells. Our study supports development of vaccines that prime and boost T cells to elicit cross-strain protective T cells, especially tissue-resident memory T cells, for lifelong immunity against distinct influenza viruses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae/immunology , Pandemics , T-Lymphocytes, Cytotoxic/immunology , Adult , Animals , B-Lymphocytes/immunology , Child , Cross Protection , Evolution, Molecular , Female , Humans , Influenza, Human/virology , Mice , Orthomyxoviridae Infections/virology , Phylogeny
9.
J Med Virol ; 88(10): 1725-32, 2016 10.
Article in English | MEDLINE | ID: mdl-26950895

ABSTRACT

Current seasonal influenza vaccines require regular updates due to antigenic drift causing loss of effectiveness and therefore providing little or no protection against novel influenza A subtypes. Next generation vaccines capable of eliciting CD8(+) T cell (CTL) mediated cross-protective immunity may offer a long-term alternative strategy. However, measuring pre- and existing levels of CTL cross-protection in humans is confounded by differences in infection histories across individuals. During 2000-2003, H1N2 viruses circulated persistently in the human population for the first time and we hypothesized that the viral nucleoprotein (NP) contained novel CTL epitopes that may have contributed to the survival of the viruses. This study describes the immunogenic NP peptides of H1N1, H2N2, and H3N2 influenza viruses isolated from humans over the past century, 1918-2003, by comparing this historical dataset to reference NP peptides from H1N2 that circulated in humans during 2000-2003. Observed peptides sequences ranged from highly conserved (15%) to highly variable (12%), with variation unrelated to reported immunodominance. No unique NP peptides which were exclusive to the H1N2 viruses were noted. However, the virus had inherited the NP from a recently emerged H3N2 variant containing novel peptides, which may have assisted its persistence. Any advantage due to this novelty was subsequently lost with emergence of a newer H3N2 variant in 2003. Our approach has potential to provide insight into the population context in which influenza viruses emerge, and may help to inform immunogenic peptide selection for CTL-inducing influenza vaccines. J. Med. Virol. 88:1725-1732, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cross Protection , Epitopes, T-Lymphocyte , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H1N2 Subtype/immunology , Animals , Antibodies, Viral/immunology , Humans , Influenza A Virus, H1N1 Subtype/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/chemistry , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Nucleocapsid Proteins , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Peptides/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Core Proteins/genetics , Viral Core Proteins/immunology
10.
Curr Opin Virol ; 16: 132-142, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26974887

ABSTRACT

Influenza A viruses (IAVs) cause significant morbidity and mortality worldwide, despite new strain-specific vaccines being available annually. As IAV-specific CD8(+) T cells promote viral control in the absence of neutralizing antibodies, and can mediate cross-reactive immunity toward distinct IAVs to drive rapid recovery from both mild and severe influenza disease, there is great interest in developing a universal T cell vaccine. However, despite detailed studies in mouse models of influenza virus infection, there is still a paucity of data on human epitope-specific CD8(+) T cell responses to IAVs. This review focuses on our current understanding of human CD8(+) T cell immunity against distinct IAVs and discusses the possibility of achieving a CD8(+) T cell mediated-vaccine that protects against multiple, distinct IAV strains across diverse human populations. We also review the importance of CD8(+) T cell immunity in individuals highly susceptible to severe influenza infection, including those hospitalised with influenza, the elderly and Indigenous populations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Pathogen Interactions/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Adaptive Immunity , Animals , Cross Reactions/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Immunodominant Epitopes/immunology , Immunologic Memory , Influenza Vaccines/immunology , Influenza, Human/diagnosis , Influenza, Human/prevention & control , Severity of Illness Index , T-Cell Antigen Receptor Specificity , Vaccination
11.
Nat Commun ; 6: 6833, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25967273

ABSTRACT

The avian origin A/H7N9 influenza virus causes high admission rates (>99%) and mortality (>30%), with ultimately favourable outcomes ranging from rapid recovery to prolonged hospitalization. Using a multicolour assay for monitoring adaptive and innate immunity, here we dissect the kinetic emergence of different effector mechanisms across the spectrum of H7N9 disease and recovery. We find that a diversity of response mechanisms contribute to resolution and survival. Patients discharged within 2-3 weeks have early prominent H7N9-specific CD8(+) T-cell responses, while individuals with prolonged hospital stays have late recruitment of CD8(+)/CD4(+) T cells and antibodies simultaneously (recovery by week 4), augmented even later by prominent NK cell responses (recovery >30 days). In contrast, those who succumbed have minimal influenza-specific immunity and little evidence of T-cell activation. Our study illustrates the importance of robust CD8(+) T-cell memory for protection against severe influenza disease caused by newly emerging influenza A viruses.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Influenza A Virus, H7N9 Subtype , Influenza, Human/pathology , Influenza, Human/virology , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes , Humans , Influenza, Human/immunology , Leukocytes, Mononuclear/physiology , Oligonucleotide Array Sequence Analysis , Receptors, KIR2DL1
12.
Front Microbiol ; 5: 285, 2014.
Article in English | MEDLINE | ID: mdl-24971078

ABSTRACT

Although an influenza vaccine has been available for 70 years, influenza virus still causes seasonal epidemics and worldwide pandemics. Currently available vaccines elicit strain-specific antibody (Ab) responses to the surface haemagglutinin (HA) and neuraminidase (NA) proteins, but these can be ineffective against serologically-distinct viral variants and novel subtypes. Thus, there is a great need for cross-protective or "universal" influenza vaccines to overcome the necessity for annual immunization against seasonal influenza and to provide immunity to reduce the severity of infection with pandemic or outbreak viruses. It is well established that natural influenza infection can provide cross-reactive immunity that can reduce the impact of infection with distinct influenza type A strains and subtypes, including H1N1, H3N2, H2N2, H5N1, and H7N9. The key to generating universal influenza immunity through vaccination is to target functionally-conserved regions of the virus, which include epitopes on the internal proteins for cross-reactive T cell immunity or on the HA stem for broadly reactive Ab responses. In the wake of the 2009 H1N1 pandemic, broadly neutralizing antibodies (bnAbs) have been characterized and isolated from convalescent and vaccinated individuals, inspiring development of new vaccination techniques to elicit such responses. Induction of influenza-specific T cell responses through vaccination has also been recently examined in clinical trials. Strong evidence is available from human and animal models of influenza to show that established influenza-specific T cell memory can reduce viral shedding and symptom severity. However, the published evidence also shows that CD8(+) T cells can efficiently select immune escape mutants early after influenza virus infection. Here, we discuss universal immunity to influenza viruses mediated by both cross-reactive T cells and Abs, the mechanisms of immune evasion in influenza, and propose how to counteract commonly occurring immune-escape variants.

13.
Crit Rev Immunol ; 34(1): 15-39, 2014.
Article in English | MEDLINE | ID: mdl-24579700

ABSTRACT

Influenza infection remains a global threat to human health. Influenza viruses are normally controlled by antibodies specific for the surface glycoproteins hemagglutinin (HA) and neuraminidase (NA). Standard influenza vaccines are aimed at inducing these antibodies, but they must be administered annually and can be rendered ineffective since different strains circulate from year to year and vary considerably in their individual HA and NA profiles. Influenza-specific T cells have been shown to be protective in animal models and typically recognize the more conserved internal influenza proteins. Improving our understanding of influenza-specific T-cell responses, including immunodominance, specific epitope sequences, strain-related epitope variation, host/virus interaction, and the balance between immunity versus immunopathology, will be important to improve future T-cell-based vaccines, which promise broader strain coverage and longer-lasting protection than current standard vaccines.


Subject(s)
Influenza A virus/immunology , Influenza Vaccines , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/immunology , Antigenic Variation/immunology , Antigens, Viral/immunology , Disease Models, Animal , Humans , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Influenza, Human/prevention & control , Orthomyxoviridae Infections/prevention & control , T-Lymphocytes/virology
14.
Proc Natl Acad Sci U S A ; 111(3): 1049-54, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395804

ABSTRACT

The absence of preexisting neutralizing antibodies specific for the novel A (H7N9) influenza virus indicates a lack of prior human exposure. As influenza A virus-specific CD8(+) T lymphocytes (CTLs) can be broadly cross-reactive, we tested whether immunogenic peptides derived from H7N9 might be recognized by memory CTLs established following infection with other influenza strains. Probing across multiple ethnicities, we identified 32 conserved epitopes derived from the nucleoprotein (NP) and matrix-1 (M1) proteins. These NP and M1 peptides are presented by HLAs prevalent in 16-57% of individuals. Remarkably, some HLA alleles (A*0201, A*0301, B*5701, B*1801, and B*0801) elicit robust CTL responses against any human influenza A virus, including H7N9, whereas ethnicities where HLA-A*0101, A*6801, B*1501, and A*2402 are prominent, show limited CTL response profiles. By this criterion, some groups, especially the Alaskan and Australian Indigenous peoples, would be particularly vulnerable to H7N9 infection. This dissection of CTL-mediated immunity to H7N9 thus suggests strategies for both vaccine delivery and development.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Influenza A Virus, H7N9 Subtype/immunology , Influenza, Human/ethnology , Influenza, Human/immunology , Australia , Cross Reactions/immunology , Crystallography, X-Ray , Epitopes, T-Lymphocyte/immunology , Ethnicity , HLA Antigens/immunology , HLA-A Antigens/immunology , Humans , Immunologic Memory , Influenza Vaccines/immunology , Leukocytes, Mononuclear/cytology , Likelihood Functions , Mutation , Peptides/immunology
15.
Nat Commun ; 4: 2663, 2013.
Article in English | MEDLINE | ID: mdl-24173108

ABSTRACT

Influenza A virus-specific CD8(+) cytotoxic T lymphocytes (CTLs) provide a degree of cross-strain protection that is potentially subverted by mutation. Here we describe the sequential emergence of such variants within CTL epitopes for a persistently infected, immunocompromised infant. Further analysis in immunodeficient and wild-type mice supports the view that CTL escape variants arise frequently in influenza, accumulate with time and revert in the absence of immune pressure under MHCI-mismatched conditions. Viral fitness, the abundance of endogenous CD8(+) T cell responses and T cell receptor repertoire diversity influence the nature of these de novo mutants. Structural characterization of dominant escape variants shows how the peptide-MHCI interaction is modified to affect variant-MHCI stability. The mechanism of influenza virus escape thus looks comparable to that recognized for chronic RNA viruses like HIV and HCV, suggesting that immunocompromised patients with prolonged viral infection could have an important part in the emergence of influenza quasispecies.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Immunocompromised Host , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/immunology , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Antigens, Viral/genetics , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Epitopes, T-Lymphocyte/genetics , Gene Expression/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immune Evasion , Infant , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/pathology , Influenza, Human/virology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Peptides/genetics , Peptides/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Cytotoxic/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...