Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 6 of 6
  • Item
    Thumbnail Image
    Robust and prototypical immune responses toward influenza vaccines in the high-risk group of Indigenous Australians
    Hensen, L ; Nguyen, THO ; Rowntree, LC ; Damelang, T ; Koutsakos, M ; Aban, M ; Hurt, A ; Harland, KL ; Auladell, M ; van de Sandt, CE ; Everitt, A ; Blacker, C ; Oyong, DA ; Loughland, JR ; Webb, JR ; Wines, BD ; Hogarth, PM ; Flanagan, KL ; Plebanski, M ; Wheatley, A ; Chung, AW ; Kent, SJ ; Miller, A ; Clemens, EB ; Doherty, PC ; Nelson, J ; Davies, J ; Tong, SYC ; Kedzierska, K (NATL ACAD SCIENCES, 2021-10-12)
    Morbidity and mortality rates from seasonal and pandemic influenza occur disproportionately in high-risk groups, including Indigenous people globally. Although vaccination against influenza is recommended for those most at risk, studies on immune responses elicited by seasonal vaccines in Indigenous populations are largely missing, with no data available for Indigenous Australians and only one report published on antibody responses in Indigenous Canadians. We recruited 78 Indigenous and 84 non-Indigenous Australians vaccinated with the quadrivalent influenza vaccine into the Looking into InFluenza T cell immunity - Vaccination cohort study and collected blood to define baseline, early (day 7), and memory (day 28) immune responses. We performed in-depth analyses of T and B cell activation, formation of memory B cells, and antibody profiles and investigated host factors that could contribute to vaccine responses. We found activation profiles of circulating T follicular helper type-1 cells at the early stage correlated strongly with the total change in antibody titers induced by vaccination. Formation of influenza-specific hemagglutinin-binding memory B cells was significantly higher in seroconverters compared with nonseroconverters. In-depth antibody characterization revealed a reduction in immunoglobulin G3 before and after vaccination in the Indigenous Australian population, potentially linked to the increased frequency of the G3m21* allotype. Overall, our data provide evidence that Indigenous populations elicit robust, broad, and prototypical immune responses following immunization with seasonal inactivated influenza vaccines. Our work strongly supports the recommendation of influenza vaccination to protect Indigenous populations from severe seasonal influenza virus infections and their subsequent complications.
  • Item
    Thumbnail Image
    Immune cellular networks underlying recovery from influenza virus infection in acute hospitalized patients
    Nguyen, THO ; Koutsakos, M ; van de Sandt, CE ; Crawford, JC ; Loh, L ; Sant, S ; Grzelak, L ; Allen, EK ; Brahm, T ; Clemens, EB ; Auladell, M ; Hensen, L ; Wang, Z ; Nussing, S ; Jia, X ; Gunther, P ; Wheatley, AK ; Kent, SJ ; Aban, M ; Deng, Y-M ; Laurie, KL ; Hurt, AC ; Gras, S ; Rossjohn, J ; Crowe, J ; Xu, J ; Jackson, D ; Brown, LE ; La Gruta, N ; Chen, W ; Doherty, PC ; Turner, SJ ; Kotsimbos, TC ; Thomas, PG ; Cheng, AC ; Kedzierska, K (NATURE PORTFOLIO, 2021-05-11)
    How innate and adaptive immune responses work in concert to resolve influenza disease is yet to be fully investigated in one single study. Here, we utilize longitudinal samples from patients hospitalized with acute influenza to understand these immune responses. We report the dynamics of 18 important immune parameters, related to clinical, genetic and virological factors, in influenza patients across different severity levels. Influenza disease correlates with increases in IL-6/IL-8/MIP-1α/β cytokines and lower antibody responses. Robust activation of circulating T follicular helper cells correlates with peak antibody-secreting cells and influenza heamaglutinin-specific memory B-cell numbers, which phenotypically differs from vaccination-induced B-cell responses. Numbers of influenza-specific CD8+ or CD4+ T cells increase early in disease and retain an activated phenotype during patient recovery. We report the characterisation of immune cellular networks underlying recovery from influenza infection which are highly relevant to other infectious diseases.
  • Item
    Thumbnail Image
    Suboptimal SARS-CoV-2-specific CD8+ T cell response associated with the prominent HLA-A*02:01 phenotype
    Habel, JR ; Nguyen, THO ; van de Sandt, CE ; Juno, JA ; Chaurasia, P ; Wragg, K ; Koutsakos, M ; Hensen, L ; Jia, X ; Chua, B ; Zhang, W ; Tan, H-X ; Flanagan, KL ; Doolan, DL ; Torresi, J ; Chen, W ; Wakim, LM ; Cheng, AC ; Doherty, PC ; Petersen, J ; Rossjohn, J ; Wheatley, AK ; Kent, SJ ; Rowntree, LC ; Kedzierska, K (NATL ACAD SCIENCES, 2020-09-29)
    An improved understanding of human T cell-mediated immunity in COVID-19 is important for optimizing therapeutic and vaccine strategies. Experience with influenza shows that infection primes CD8+ T cell memory to peptides presented by common HLA types like HLA-A2, which enhances recovery and diminishes clinical severity upon reinfection. Stimulating peripheral blood mononuclear cells from COVID-19 convalescent patients with overlapping peptides from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led to the clonal expansion of SARS-CoV-2-specific CD8+ and CD4+ T cells in vitro, with CD4+ T cells being robust. We identified two HLA-A*02:01-restricted SARS-CoV-2-specfic CD8+ T cell epitopes, A2/S269-277 and A2/Orf1ab3183-3191 Using peptide-HLA tetramer enrichment, direct ex vivo assessment of A2/S269+CD8+ and A2/Orf1ab3183+CD8+ populations indicated that A2/S269+CD8+ T cells were detected at comparable frequencies (∼1.3 × 10-5) in acute and convalescent HLA-A*02:01+ patients. These frequencies were higher than those found in uninfected HLA-A*02:01+ donors (∼2.5 × 10-6), but low when compared to frequencies for influenza-specific (A2/M158) and Epstein-Barr virus (EBV)-specific (A2/BMLF1280) (∼1.38 × 10-4) populations. Phenotyping A2/S269+CD8+ T cells from COVID-19 convalescents ex vivo showed that A2/S269+CD8+ T cells were predominantly negative for CD38, HLA-DR, PD-1, and CD71 activation markers, although the majority of total CD8+ T cells expressed granzymes and/or perforin. Furthermore, the bias toward naïve, stem cell memory and central memory A2/S269+CD8+ T cells rather than effector memory populations suggests that SARS-CoV-2 infection may be compromising CD8+ T cell activation. Priming with appropriate vaccines may thus be beneficial for optimizing CD8+ T cell immunity in COVID-19.
  • Item
    Thumbnail Image
    Human γδ T-cell receptor repertoire is shaped by influenza viruses, age and tissue compartmentalisation
    Sant, S ; Jenkins, MR ; Dash, P ; Watson, KA ; Wang, Z ; Pizzolla, A ; Koutsakos, M ; Nguyen, THO ; Lappas, M ; Crowe, J ; Loudovaris, T ; Mannering, S ; Westall, GP ; Kotsimbos, TC ; Cheng, AC ; Wakim, L ; Doherty, PC ; Thomas, PG ; Loh, L ; Kedzierska, K (WILEY, 2019)
    BACKGROUND: Although γδ T cells comprise up to 10% of human peripheral blood T cells, questions remain regarding their role in disease states and T-cell receptor (TCR) clonal expansions. We dissected anti-viral functions of human γδ T cells towards influenza viruses and defined influenza-reactive γδ TCRs in the context of γδ-TCRs across the human lifespan. METHODS: We performed 51Cr-killing assay and single-cell time-lapse live video microscopy to define mechanisms underlying γδ T-cell-mediated killing of influenza-infected targets. We assessed cytotoxic profiles of γδ T cells in influenza-infected patients and IFN-γ production towards influenza-infected lung epithelial cells. Using single-cell RT-PCR, we characterised paired TCRγδ clonotypes for influenza-reactive γδ T cells in comparison with TCRs from healthy neonates, adults, elderly donors and tissues. RESULTS: We provide the first visual evidence of γδ T-cell-mediated killing of influenza-infected targets and show distinct features to those reported for CD8+ T cells. γδ T cells displayed poly-cytotoxic profiles in influenza-infected patients and produced IFN-γ towards influenza-infected cells. These IFN-γ-producing γδ T cells were skewed towards the γ9δ2 TCRs, particularly expressing the public GV9-TCRγ, capable of pairing with numerous TCR-δ chains, suggesting their significant role in γδ T-cell immunity. Neonatal γδ T cells displayed extensive non-overlapping TCRγδ repertoires, while adults had enriched γ9δ2-pairings with diverse CDR3γδ regions. Conversely, the elderly showed distinct γδ-pairings characterised by large clonal expansions, a profile also prominent in adult tissues. CONCLUSION: Human TCRγδ repertoire is shaped by age, tissue compartmentalisation and the individual's history of infection, suggesting that these somewhat enigmatic γδ T cells indeed respond to antigen challenge.
  • Item
    No Preview Available
    Human mucosal-associated invariant T cells contribute to antiviral influenza immunity via IL-18-dependent activation
    Loh, L ; Wang, Z ; Sant, S ; Koutsakos, M ; Jegaskanda, S ; Corbett, AJ ; Liu, L ; Fairlie, DP ; Crowe, J ; Rossjohn, J ; Xu, J ; Doherty, PC ; McCluskey, J ; Kedzierska, K (NATL ACAD SCIENCES, 2016-09-06)
    Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes known to elicit potent immunity to a broad range of bacteria, mainly via the rapid production of inflammatory cytokines. Whether MAIT cells contribute to antiviral immunity is less clear. Here we asked whether MAIT cells produce cytokines/chemokines during severe human influenza virus infection. Our analysis in patients hospitalized with avian H7N9 influenza pneumonia showed that individuals who recovered had higher numbers of CD161(+)Vα7.2(+) MAIT cells in peripheral blood compared with those who succumbed, suggesting a possible protective role for this lymphocyte population. To understand the mechanism underlying MAIT cell activation during influenza, we cocultured influenza A virus (IAV)-infected human lung epithelial cells (A549) and human peripheral blood mononuclear cells in vitro, then assayed them by intracellular cytokine staining. Comparison of influenza-induced MAIT cell activation with the profile for natural killer cells (CD56(+)CD3(-)) showed robust up-regulation of IFNγ for both cell populations and granzyme B in MAIT cells, although the individual responses varied among healthy donors. However, in contrast to the requirement for cell-associated factors to promote NK cell activation, the induction of MAIT cell cytokine production was dependent on IL-18 (but not IL-12) production by IAV-exposed CD14(+) monocytes. Overall, this evidence for IAV activation via an indirect, IL-18-dependent mechanism indicates that MAIT cells are protective in influenza, and also possibly in any human disease process in which inflammation and IL-18 production occur.
  • Item
    Thumbnail Image
    Clonally diverse CD38+HLA-DR+CD8+ T cells persist during fatal H7N9 disease
    Wang, Z ; Zhu, L ; Nguyen, THO ; Wan, Y ; Sant, S ; Quinones-Parra, SM ; Crawford, JC ; Eltahla, AA ; Rizzetto, S ; Bull, RA ; Qiu, C ; Koutsakos, M ; Clemens, EB ; Loh, L ; Chen, T ; Liu, L ; Cao, P ; Ren, Y ; Kedzierski, L ; Kotsimbos, T ; McCaw, JM ; La Gruta, NL ; Turner, SJ ; Cheng, AC ; Luciani, F ; Zhang, X ; Doherty, PC ; Thomas, PG ; Xu, J ; Kedzierska, K (NATURE PORTFOLIO, 2018-02-26)
    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)-αβ analyses of activated CD38+HLA-DR+CD8+ T cells show similar TCRαβ 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 TCRαβ clonotypes with appropriate transcriptome signatures is needed for early protection against severe influenza disease.