Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 14
  • Item
    Thumbnail Image
    High expression of CD38 and MHC class II on CD8+ T cells during severe influenza disease reflects bystander activation and trogocytosis
    Jia, X ; Chua, BY ; Loh, L ; Koutsakos, M ; Kedzierski, L ; Olshansky, M ; Heath, WR ; Chang, SY ; Xu, J ; Wang, Z ; Kedzierska, K (WILEY, 2021)
    OBJECTIVES: Although co-expression of CD38 and HLA-DR reflects T-cell activation during viral infections, high and prolonged CD38+HLA-DR+ expression is associated with severe disease. To date, the mechanism underpinning expression of CD38+HLA-DR+ is poorly understood. METHODS: We used mouse models of influenza A/H9N2, A/H7N9 and A/H3N2 infection to investigate mechanisms underpinning CD38+MHC-II+ phenotype on CD8+ T cells. To further understand MHC-II trogocytosis on murine CD8+ T cells as well as the significance behind the scenario, we used adoptively transferred transgenic OT-I CD8+ T cells and A/H3N2-SIINKEKL infection. RESULTS: Analysis of influenza-specific immunodominant DbNP366 +CD8+ T-cell responses showed that CD38+MHC-II+ co-expression was detected on both virus-specific and bystander CD8+ T cells, with increased numbers of both CD38+MHC-II+CD8+ T-cell populations observed in immune organs including the site of infection during severe viral challenge. OT-I cells adoptively transferred into MHC-II-/- mice had no MHC-II after infection, suggesting that MHC-II was acquired via trogocytosis. The detection of CD19 on CD38+MHC-II+ OT-I cells supports the proposition that MHC-II was acquired by trogocytosis sourced from B cells. Co-expression of CD38+MHC-II+ on CD8+ T cells was needed for optimal recall following secondary infection. CONCLUSIONS: Overall, our study demonstrates that both virus-specific and bystander CD38+MHC-II+ CD8+ T cells are recruited to the site of infection during severe disease, and that MHC-II presence occurs via trogocytosis from antigen-presenting cells. Our findings highlight the importance of the CD38+MHC-II+ phenotype for CD8+ T-cell recall.
  • Item
    Thumbnail Image
    CD8+ T cell landscape in Indigenous and non-Indigenous people restricted by influenza mortality-associated HLA-A*24:02 allomorph
    Hensen, L ; Illing, PT ; Bridie Clemens, E ; Nguyen, THO ; Koutsakos, M ; van de Sandt, CE ; Mifsud, NA ; Nguyen, AT ; Szeto, C ; Chua, BY ; Halim, H ; Rizzetto, S ; Luciani, F ; Loh, L ; Grant, EJ ; Saunders, PM ; Brooks, AG ; Rockman, S ; Kotsimbos, TC ; Cheng, AC ; Richards, M ; Westall, GP ; Wakim, LM ; Loudovaris, T ; Mannering, SI ; Elliott, M ; Tangye, SG ; Jackson, DC ; Flanagan, KL ; Rossjohn, J ; Gras, S ; Davies, J ; Miller, A ; Tong, SYC ; Purcell, AW ; Kedzierska, K (NATURE PORTFOLIO, 2021-05-18)
    Indigenous people worldwide are at high risk of developing severe influenza disease. HLA-A*24:02 allele, highly prevalent in Indigenous populations, is associated with influenza-induced mortality, although the basis for this association is unclear. Here, we define CD8+ T-cell immune landscapes against influenza A (IAV) and B (IBV) viruses in HLA-A*24:02-expressing Indigenous and non-Indigenous individuals, human tissues, influenza-infected patients and HLA-A*24:02-transgenic mice. We identify immunodominant protective CD8+ T-cell epitopes, one towards IAV and six towards IBV, with A24/PB2550-558-specific CD8+ T cells being cross-reactive between IAV and IBV. Memory CD8+ T cells towards these specificities are present in blood (CD27+CD45RA- phenotype) and tissues (CD103+CD69+ phenotype) of healthy individuals, and effector CD27-CD45RA-PD-1+CD38+CD8+ T cells in IAV/IBV patients. Our data show influenza-specific CD8+ T-cell responses in Indigenous Australians, and advocate for T-cell-mediated vaccines that target and boost the breadth of IAV/IBV-specific CD8+ T cells to protect high-risk HLA-A*24:02-expressing Indigenous and non-Indigenous populations from severe influenza disease.
  • 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
    Influenza A Virus-Infected Lung Epithelial Cell Co-Culture with Human Peripheral Blood Mononuclear Cells
    Loh, L ; Koutsakos, M ; Kedzierska, K ; Hinks, TSC ; Kaipe, H ; Magalhaes, I (HUMANA PRESS INC, 2020)
    Sensing of influenza A virus (IAV) infection by pattern recognition receptors can occur by either direct infection of lung epithelial cells or uptake of virus-infected cells by innate cells such as dendritic cells/monocytes. This triggers a series of downstream events including activation of the inflammasome, the production of cytokines, chemokines, and the upregulation of stress-induced ligands that can lead to the activation of innate cells. These cells include innate lymphocytes such as MAIT, NKT, NK, and γδ T cells. Here we describe a method used to allow activation of human innate lymphocytes in co-culture with an IAV-infected human lung epithelial cell line (A549) to measure ex vivo effector functions (TNF and IFNγ) in a mixed culture environment. We describe (1) infection of the human lung epithelial cell line, (2) co-culture with PBMC, and (3) measurement of activation using intracellular cytokine staining.
  • Item
    Thumbnail Image
    Study of MAIT Cell Activation in Viral Infections In Vivo
    Hinks, TSC ; van Wilgenburg, B ; Wang, H ; Loh, L ; Koutsakos, M ; Kedzierska, K ; Corbett, AJ ; Chen, Z ; Kaipe, H ; Magalhaes, I (HUMANA PRESS INC, 2020)
    MAIT cells are abundant, highly evolutionarily conserved innate-like lymphocytes expressing a semi-invariant T cell receptor (TCR), which recognizes microbially derived small intermediate molecules from the riboflavin biosynthetic pathway. However, in addition to their TCR-mediated functions they can also be activated in a TCR-independent manner via cytokines including IL-12, -15, -18, and type I interferon. Emerging data suggest that they are expanded and activated by a range of viral infections, and significantly that they can contribute to a protective anti-viral response. Here we describe methods used to investigate these anti-viral functions in vivo in murine models. To overcome the technical challenge that MAIT cells are rare in specific pathogen-free laboratory mice, we describe how pulmonary MAIT cells can be expanded using intranasal bacterial infection or a combination of synthetic MAIT cell antigen and TLR agonists. We also describe protocols for adoptive transfer of MAIT cells, methods for lung homogenization for plaque assays, and surface and intracellular cytokine staining to determine MAIT cell activation.
  • Item
    Thumbnail Image
    Single-Cell Approach to Influenza Specific CD8+ T Cell Receptor Repertoires Across Different Age Groups, Tissues, and Following Influenza Virus Infection
    Sant, S ; Grzelak, L ; Wang, Z ; Pizzollal, A ; Koutsakos, M ; Crowe, J ; Loudoyaris, T ; Mannering, SI ; Westall, GP ; Wakiml, LM ; Rossjohn, J ; Gras, S ; Richards, M ; Xu, J ; Thomas, PG ; Loh, L ; Nguyen, TH ; Kedzierska, K (FRONTIERS MEDIA SA, 2018-06-27)
    CD8+ T cells recognizing antigenic peptides derived from conserved internal viral proteins confer broad protection against distinct influenza viruses. As memory CD8+ T cells change throughout the human lifetime and across tissue compartments, we investigated how T cell receptor (TCR) composition and diversity relate to memory CD8+ T cells across anatomical sites and immunological phases of human life. We used ex vivo peptide-HLA tetramer magnetic enrichment, single-cell multiplex RT-PCR for both the TCR-alpha (TCRα) and TCR-beta (TCRβ) chains, and new TCRdist and grouping of lymphocyte interactions by paratope hotspots (GLIPH) algorithms to compare TCRs directed against the most prominent human influenza epitope, HLA-A*02:01-M158-66 (A2+M158). We dissected memory TCR repertoires directed toward A2+M158 CD8+ T cells within human tissues and compared them to human peripheral blood of young and elderly adults. Furthermore, we compared these memory CD8+ T cell repertoires to A2+M158 CD8+ TCRs during acute influenza disease in patients hospitalized with avian A/H7N9 virus. Our study provides the first ex vivo comparative analysis of paired antigen-specific TCR-α/β clonotypes across different tissues and peripheral blood across different age groups. We show that human A2+M158 CD8+ T cells can be readily detected in human lungs, spleens, and lymph nodes, and that tissue A2+M158 TCRαβ repertoires reflect A2+M158 TCRαβ clonotypes derived from peripheral blood in healthy adults and influenza-infected patients. A2+M158 TCRαβ repertoires displayed distinct features only in elderly adults, with large private TCRαβ clonotypes replacing the prominent and public TRBV19/TRAV27 TCRs. Our study provides novel findings on influenza-specific TCRαβ repertoires within human tissues, raises the question of how we can prevent the loss of optimal TCRαβ signatures with aging, and provides important insights into the rational design of T cell-mediated vaccines and immunotherapies.
  • 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
    Thumbnail Image
    Monocyte apoptotic bodies are vehicles for influenza A virus propagation
    Atkin-Smith, GK ; Duan, M ; Zanker, DJ ; Loh, L ; Nguyen, THO ; Koutsakos, M ; Nguyen, T ; Jiang, X ; Carrera, J ; Phan, TK ; Liu, C ; Paone, S ; Oveissi, S ; Hodge, AL ; Baxter, AA ; Kedzierska, K ; Mackenzie, JM ; Hulett, MD ; Bilsel, P ; Chen, W ; Poon, IKH (NATURE PUBLISHING GROUP, 2020-05-08)
    The disassembly of apoptotic cells into small membrane-bound vesicles termed apoptotic bodies (ApoBDs) is a hallmark of apoptosis; however, the functional significance of this process is not well defined. We recently discovered a new membrane protrusion (termed beaded apoptopodia) generated by apoptotic monocytes which fragments to release an abundance of ApoBDs. To investigate the function of apoptotic monocyte disassembly, we used influenza A virus (IAV) infection as a proof-of-concept model, as IAV commonly infects monocytes in physiological settings. We show that ApoBDs generated from IAV-infected monocytes contained IAV mRNA, protein and virions and consequently, could facilitate viral propagation in vitro and in vivo, and induce a robust antiviral immune response. We also identified an antipsychotic, Haloperidol, as an unexpected inhibitor of monocyte cell disassembly which could impair ApoBD-mediated viral propagation under in vitro conditions. Together, this study reveals a previously unrecognised function of apoptotic monocyte disassembly in the pathogenesis of IAV infections.
  • Item
    Thumbnail Image
    HLA-B*27:05 alters immunodominance hierarchy of universal influenza-specific CD8+T cells
    Sant, S ; Quinones-Parra, SM ; Koutsakos, M ; Grant, EJ ; Loudovaris, T ; Mannering, SI ; Crowe, J ; van de Sandt, CE ; Rimmelzwaan, GF ; Rossjohn, J ; Gras, S ; Loh, L ; Nguyen, THO ; Kedzierska, K ; Schultz-Cherry, S (PUBLIC LIBRARY SCIENCE, 2020-08)
    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 TCRαβ 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 TCRαβ 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.
  • Item
    Thumbnail Image
    Circulating TFH cells, serological memory, and tissue compartmentalization shape human influenza-specific B cell immunity
    Koutsakos, M ; Wheatley, AK ; Loh, L ; Clemens, EB ; Sant, S ; Nussing, S ; Fox, A ; Chung, AW ; Laurie, KL ; Hurt, AC ; Rockman, S ; Lappas, M ; Loudovaris, T ; Mannering, SI ; Westall, GP ; Elliot, M ; Tangye, SG ; Wakim, LM ; Kent, SJ ; Nguyen, THO ; Kedzierska, K (AMER ASSOC ADVANCEMENT SCIENCE, 2018-02-14)
    Immunization with the inactivated influenza vaccine (IIV) remains the most effective strategy to combat seasonal influenza infections. IIV activates B cells and T follicular helper (TFH) cells and thus engenders antibody-secreting cells and serum antibody titers. However, the cellular events preceding generation of protective immunity in humans are inadequately understood. We undertook an in-depth analysis of B cell and T cell immune responses to IIV in 35 healthy adults. Using recombinant hemagglutinin (rHA) probes to dissect the quantity, phenotype, and isotype of influenza-specific B cells against A/California09-H1N1, A/Switzerland-H3N2, and B/Phuket, we showed that vaccination induced a three-pronged B cell response comprising a transient CXCR5-CXCR3+ antibody-secreting B cell population, CD21hiCD27+ memory B cells, and CD21loCD27+ B cells. Activation of circulating TFH cells correlated with the development of both CD21lo and CD21hi memory B cells. However, preexisting antibodies could limit increases in serum antibody titers. IIV had no marked effect on CD8+, mucosal-associated invariant T, γδ T, and natural killer cell activation. In addition, vaccine-induced B cells were not maintained in peripheral blood at 1 year after vaccination. We provide a dissection of rHA-specific B cells across seven human tissue compartments, showing that influenza-specific memory (CD21hiCD27+) B cells primarily reside within secondary lymphoid tissues and the lungs. Our study suggests that a rational design of universal vaccines needs to consider circulating TFH cells, preexisting serological memory, and tissue compartmentalization for effective B cell immunity, as well as to improve targeting cellular T cell immunity.