Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 87
  • Item
    Thumbnail Image
    Increasing HbA1c is associated with reduced CD8+ T cell functionality in response to influenza virus in a TCR-dependent manner in individuals with diabetes mellitus
    Hulme, KD ; Tong, ZWM ; Rowntree, LC ; van de Sandt, CE ; Ronacher, K ; Grant, EJ ; Dorey, ES ; Gallo, LA ; Gras, S ; Kedzierska, K ; Barrett, HL ; Short, KR (SPRINGER BASEL AG, 2024-12)
    Diabetes mellitus is on the rise globally and is a known susceptibility factor for severe influenza virus infections. However, the mechanisms by which diabetes increases the severity of an influenza virus infection are yet to be fully defined. Diabetes mellitus is hallmarked by high glucose concentrations in the blood. We hypothesized that these high glucose concentrations affect the functionality of CD8+ T cells, which play a key role eliminating virus-infected cells and have been shown to decrease influenza disease severity. To study the effect of hyperglycemia on CD8+ T cell function, we stimulated peripheral blood mononuclear cells (PBMCs) from donors with and without diabetes with influenza A virus, anti-CD3/anti-CD28-coated beads, PMA and ionomycin (PMA/I), or an influenza viral peptide pool. After stimulation, cells were assessed for functionality [as defined by expression of IFN-γ, TNF-α, macrophage inflammatory protein (MIP)-1β, and lysosomal-associated membrane protein-1 (CD107a)] using flow cytometry. Our results showed that increasing HbA1c correlated with a reduction in TNF-α production by CD8+ T cells in response to influenza stimulation in a TCR-specific manner. This was not associated with any changes to CD8+ T cell subsets. We conclude that hyperglycemia impairs CD8+ T cell function to influenza virus infection, which may be linked with the increased risk of severe influenza in patients with diabetes.
  • Item
    Thumbnail Image
    Interim results from a phase I randomized, placebo-controlled trial of novel SARS-CoV-2 beta variant receptor-binding domain recombinant protein and mRNA vaccines as a 4th dose booster
    Nolan, TM ; Deliyannis, G ; Griffith, M ; Braat, S ; Allen, LF ; Audsley, J ; Chung, AW ; Ciula, M ; Gherardin, NA ; Giles, ML ; Gordon, TP ; Grimley, SL ; Horng, L ; Jackson, DC ; Juno, JA ; Kedzierska, K ; Kent, SJ ; Lewin, SR ; Littlejohn, M ; McQuilten, HA ; Mordant, FL ; Nguyen, THO ; Soo, VP ; Price, B ; Purcell, DFJ ; Ramanathan, P ; Redmond, SJ ; Rockman, S ; Ruan, Z ; Sasadeusz, J ; Simpson, JA ; Subbarao, K ; Fabb, SA ; Payne, TJ ; Takanashi, A ; Tan, CW ; Torresi, J ; Wang, JJ ; Wang, L-F ; Al-Wassiti, H ; Wong, CY ; Zaloumis, S ; Pouton, CW ; Godfrey, DI (ELSEVIER, 2023-12)
    BACKGROUND: SARS-CoV-2 booster vaccination should ideally enhance protection against variants and minimise immune imprinting. This Phase I trial evaluated two vaccines targeting SARS-CoV-2 beta-variant receptor-binding domain (RBD): a recombinant dimeric RBD-human IgG1 Fc-fusion protein, and an mRNA encoding a membrane-anchored RBD. METHODS: 76 healthy adults aged 18-64 y, previously triple vaccinated with licensed SARS-CoV-2 vaccines, were randomised to receive a 4th dose of either an adjuvanted (MF59®, CSL Seqirus) protein vaccine (5, 15 or 45 μg, N = 32), mRNA vaccine (10, 20, or 50 μg, N = 32), or placebo (saline, N = 12) at least 90 days after a 3rd boost vaccination or SARS-CoV-2 infection. Bleeds occurred on days 1 (prior to vaccination), 8, and 29. CLINICALTRIALS: govNCT05272605. FINDINGS: No vaccine-related serious or medically-attended adverse events occurred. The protein vaccine reactogenicity was mild, whereas the mRNA vaccine was moderately reactogenic at higher dose levels. Best anti-RBD antibody responses resulted from the higher doses of each vaccine. A similar pattern was seen with live virus neutralisation and surrogate, and pseudovirus neutralisation assays. Breadth of immune response was demonstrated against BA.5 and more recent omicron subvariants (XBB, XBB.1.5 and BQ.1.1). Binding antibody titres for both vaccines were comparable to those of a licensed bivalent mRNA vaccine. Both vaccines enhanced CD4+ and CD8+ T cell activation. INTERPRETATION: There were no safety concerns and the reactogenicity profile was mild and similar to licensed SARS-CoV-2 vaccines. Both vaccines showed strong immune boosting against beta, ancestral and omicron strains. FUNDING: Australian Government Medical Research Future Fund, and philanthropies Jack Ma Foundation and IFM investors.
  • Item
    Thumbnail Image
    Evolution of Humoral and Cellular Immunity Post-Breakthrough Coronavirus Disease 2019 in Vaccinated Patients With Hematologic Malignancy Receiving Tixagevimab-Cilgavimab
    Hall, VG ; Nguyen, THO ; Allen, LF ; Rowntree, LC ; Kedzierski, L ; Chua, BY ; Lim, C ; Saunders, NR ; Klimevski, E ; Tennakoon, GS ; Seymour, JF ; Wadhwa, V ; Cain, N ; Vo, KL ; Nicholson, S ; Karapanagiotidis, T ; Williamson, DA ; Thursky, KA ; Spelman, T ; Yong, MK ; Slavin, MA ; Kedzierska, K ; Teh, BW (OXFORD UNIV PRESS INC, 2023-11-01)
    BACKGROUND: In-depth immunogenicity studies of tixagevimab-cilgavimab (T-C) are lacking, including following breakthrough coronavirus disease 2019 (COVID-19) in vaccinated patients with hematologic malignancy (HM) receiving T-C as pre-exposure prophylaxis. METHODS: We performed a prospective, observational cohort study and detailed immunological analyses of 93 patients with HM who received T-C from May 2022, with and without breakthrough infection, during a follow-up period of 6 months and dominant Omicron BA.5 variant. RESULTS: In 93 patients who received T-C, there was an increase in Omicron BA.4/5 receptor-binding domain (RBD) immunoglobulin G (IgG) antibody titers that persisted for 6 months and was equivalent to 3-dose-vaccinated uninfected healthy controls at 1 month postinjection. Omicron BA.4/5 neutralizing antibody was lower in patients receiving B-cell-depleting therapy within 12 months despite receipt of T-C. COVID-19 vaccination during T-C treatment did not incrementally improve RBD or neutralizing antibody levels. In 16 patients with predominantly mild breakthrough infection, no change in serum neutralization of Omicron BA.4/5 postinfection was detected. Activation-induced marker assay revealed an increase in CD4+ (but not CD8+) T cells post infection, comparable to previously infected healthy controls. CONCLUSIONS: Our study provides proof-of-principle for a pre-exposure prophylaxis strategy and highlights the importance of humoral and cellular immunity post-breakthrough COVID-19 in vaccinated patients with HM.
  • Item
    No Preview Available
    Broad spectrum SARS-CoV-2-specific immunity in hospitalized First Nations peoples recovering from COVID-19
    Zhang, W ; Clemens, EB ; Kedzierski, L ; Chua, BY ; Mayo, M ; Lonzi, C ; Hinchcliff, A ; Rigas, V ; Middleton, BF ; Binks, P ; Rowntree, LC ; Allen, LF ; Tan, H-X ; Petersen, J ; Chaurasia, P ; Krammer, F ; Wheatley, AK ; Kent, SJ ; Rossjohn, J ; Miller, A ; Lynar, S ; Nelson, J ; Nguyen, THO ; Davies, J ; Kedzierska, K (WILEY, 2023-11)
    Indigenous peoples globally are at increased risk of COVID-19-associated morbidity and mortality. However, data that describe immune responses to SARS-CoV-2 infection in Indigenous populations are lacking. We evaluated immune responses in Australian First Nations peoples hospitalized with COVID-19. Our work comprehensively mapped out inflammatory, humoral and adaptive immune responses following SARS-CoV-2 infection. Patients were recruited early following the lifting of strict public health measures in the Northern Territory, Australia, between November 2021 and May 2022. Australian First Nations peoples recovering from COVID-19 showed increased levels of MCP-1 and IL-8 cytokines, IgG-antibodies against Delta-RBD and memory SARS-CoV-2-specific T cell responses prior to hospital discharge in comparison with hospital admission, with resolution of hyperactivated HLA-DR+ CD38+ T cells. SARS-CoV-2 infection elicited coordinated ASC, Tfh and CD8+ T cell responses in concert with CD4+ T cell responses. Delta and Omicron RBD-IgG, as well as Ancestral N-IgG antibodies, strongly correlated with Ancestral RBD-IgG antibodies and Spike-specific memory B cells. We provide evidence of broad and robust immune responses following SARS-CoV-2 infection in Indigenous peoples, resembling those of non-Indigenous COVID-19 hospitalized patients.
  • Item
    No Preview Available
    Breakthrough COVID-19 is mild in vaccinated patients with hematological malignancy receiving tixagevimab-cilgavimab as pre-exposure prophylaxis
    Hall, VG ; Lim, C ; Saunders, NR ; Klimevski, E ; Nguyen, THO ; Kedzierski, L ; Seymour, JF ; Wadhwa, V ; Thursky, KA ; Yong, MK ; Kedzierska, K ; Slavin, MA ; Teh, BW (TAYLOR & FRANCIS LTD, 2023-09)
  • Item
    No Preview Available
    CD4+ T cell calibration of antigen-presenting cells optimizes antiviral CD8+ T cell immunity
    Gressier, E ; Schulte-Schrepping, J ; Petrov, L ; Brumhard, S ; Stubbemann, P ; Hiller, A ; Obermayer, B ; Spitzer, J ; Kostevc, T ; Whitney, PG ; Bachem, A ; Odainic, A ; van de Sandt, C ; Nguyen, THO ; Ashhurst, T ; Wilson, K ; Oates, CVL ; Gearing, LJ ; Meischel, T ; Hochheiser, K ; Greyer, M ; Clarke, M ; Kreutzenbeck, M ; Gabriel, SS ; Kastenmueller, W ; Kurts, C ; Londrigan, SL ; Kallies, A ; Kedzierska, K ; Hertzog, PJ ; Latz, E ; Chen, Y-CE ; Radford, KJ ; Chopin, M ; Schroeder, J ; Kurth, F ; Gebhardt, T ; Sander, LE ; Sawitzki, B ; Schultze, JL ; Schmidt, SV ; Bedoui, S (NATURE PORTFOLIO, 2023-06)
    Antiviral CD8+ T cell immunity depends on the integration of various contextual cues, but how antigen-presenting cells (APCs) consolidate these signals for decoding by T cells remains unclear. Here, we describe gradual interferon-α/interferon-β (IFNα/β)-induced transcriptional adaptations that endow APCs with the capacity to rapidly activate the transcriptional regulators p65, IRF1 and FOS after CD4+ T cell-mediated CD40 stimulation. While these responses operate through broadly used signaling components, they induce a unique set of co-stimulatory molecules and soluble mediators that cannot be elicited by IFNα/β or CD40 alone. These responses are critical for the acquisition of antiviral CD8+ T cell effector function, and their activity in APCs from individuals infected with severe acute respiratory syndrome coronavirus 2 correlates with milder disease. These observations uncover a sequential integration process whereby APCs rely on CD4+ T cells to select the innate circuits that guide antiviral CD8+ T cell responses.
  • Item
    No Preview Available
    Newborn and child-like molecular signatures in older adults stem from TCR shifts across human lifespan
    van de Sandt, CE ; Nguyen, THO ; Gherardin, NA ; Crawford, JC ; Samir, J ; Minervina, AA ; Pogorelyy, MV ; Rizzetto, S ; Szeto, C ; Kaur, J ; Ranson, N ; Sonda, S ; Harper, A ; Redmond, SJ ; McQuilten, HA ; Menon, T ; Sant, S ; Jia, X ; Pedrina, K ; Karapanagiotidis, T ; Cain, N ; Nicholson, S ; Chen, Z ; Lim, R ; Clemens, EB ; Eltahla, A ; La Gruta, NL ; Crowe, J ; Lappas, M ; Rossjohn, J ; Godfrey, DI ; Thomas, PG ; Gras, S ; Flanagan, KL ; Luciani, F ; Kedzierska, K (Nature Research, 2023-11)
    CD8+ T cells provide robust antiviral immunity, but how epitope-specific T cells evolve across the human lifespan is unclear. Here we defined CD8+ T cell immunity directed at the prominent influenza epitope HLA-A*02:01-M158-66 (A2/M158) across four age groups at phenotypic, transcriptomic, clonal and functional levels. We identify a linear differentiation trajectory from newborns to children then adults, followed by divergence and a clonal reset in older adults. Gene profiles in older adults closely resemble those of newborns and children, despite being clonally distinct. Only child-derived and adult-derived A2/M158+CD8+ T cells had the potential to differentiate into highly cytotoxic epitope-specific CD8+ T cells, which was linked to highly functional public T cell receptor (TCR)αβ signatures. Suboptimal TCRαβ signatures in older adults led to less proliferation, polyfunctionality, avidity and recognition of peptide mutants, although displayed no signs of exhaustion. These data suggest that priming T cells at different stages of life might greatly affect CD8+ T cell responses toward viral infections.
  • Item
    No Preview Available
    Long-term safety and immunogenicity of an MF59-adjuvanted spike glycoprotein-clamp vaccine for SARS-CoV-2 in adults aged 18–55 years or ≥56 years: 12-month results from a randomised, double-blind, placebo-controlled, phase 1 trial
    Chappell, KJ ; Mordant, FL ; Amarilla, AA ; Modhiran, N ; Liang, B ; Li, Z ; Wijesundara, DK ; Lackenby, JA ; Grif, P ; Bennet, JK ; Hensen, L ; Zhang, W ; Nguyen, THO ; Tran, MH ; Tapley, P ; Barnes, J ; Reading, PC ; Kedzierska, K ; Ranasinghe, C ; Subbarao, K ; Watterson, D ; Young, PR ; Munro, TP (Elsevier, 2023-11)
    BACKGROUND: We previously demonstrated the safety and immunogenicity of an MF59-adjuvanted COVID-19 vaccine based on the SARS-CoV-2 spike glycoprotein stabilised in a pre-fusion conformation by a molecular clamp using HIV-1 glycoprotein 41 sequences. Here, we describe 12-month results in adults aged 18-55 years and ≥56 years. METHODS: Phase 1, double-blind, placebo-controlled trial conducted in Australia (July 2020-December 2021; ClinicalTrials.govNCT04495933; active, not recruiting). Healthy adults (Part 1: 18-55 years; Part 2: ≥56 years) received two doses of placebo, 5 μg, 15 μg, or 45 μg vaccine, or one 45 μg dose of vaccine followed by placebo (Part 1 only), 28 days apart (n = 216; 24 per group). Safety, humoral immunogenicity (including against virus variants), and cellular immunogenicity were assessed to day 394 (12 months after second dose). Effects of subsequent COVID-19 vaccination on humoral responses were examined. FINDINGS: All two-dose vaccine regimens were well tolerated and elicited strong antigen-specific and neutralising humoral responses, and CD4+ T-cell responses, by day 43 in younger and older adults, although cellular responses were lower in older adults. Humoral responses waned by day 209 but were boosted in those receiving authorised vaccines. Neutralising activity against Delta and Omicron variants was present but lower than against the Wuhan strain. Cross-reactivity in HIV diagnostic tests declined over time but remained detectable in most participants. INTERPRETATION: The SARS-CoV-2 molecular clamp vaccine is well tolerated and evokes robust immune responses in adults of all ages. Although the HIV glycoprotein 41-based molecular clamp is not being progressed, the clamp concept represents a viable platform for vaccine development. FUNDING: This study was funded by the Coalition for Epidemic Preparedness Innovations, the National Health and Medical Research Council of Australia, and the Queensland Government.
  • Item
    No Preview Available
    Robust immunity to influenza vaccination in haematopoietic stem cell transplant recipients following reconstitution of humoral and adaptive immunity
    Zhang, W ; Rowntree, LC ; Muttucumaru, R ; Damelang, T ; Aban, M ; Hurt, AC ; Auladell, M ; Esterbauer, R ; Wines, B ; Hogarth, M ; Turner, SJ ; Wheatley, AK ; Kent, SJ ; Patil, S ; Avery, S ; Morrissey, O ; Chung, AW ; Koutsakos, M ; Nguyen, THO ; Cheng, AC ; Kotsimbos, TC ; Kedzierska, K (WILEY, 2023)
    OBJECTIVES: Influenza causes significant morbidity and mortality, especially in high-risk populations. Although current vaccination regimens are the best method to combat annual influenza disease, vaccine efficacy can be low in high-risk groups, such as haematopoietic stem cell transplant (HSCT) recipients. METHODS: We comprehensively assessed humoral immunity, antibody landscapes, systems serology and influenza-specific B-cell responses, together with their phenotypes and isotypes, to the inactivated influenza vaccine (IIV) in HSCT recipients in comparison to healthy controls. RESULTS: Inactivated influenza vaccine significantly increased haemagglutination inhibition (HAI) titres in HSCT recipients, similar to healthy controls. Systems serology revealed increased IgG1 and IgG3 antibody levels towards the haemagglutinin (HA) head, but not to neuraminidase, nucleoprotein or HA stem. IIV also increased frequencies of total, IgG class-switched and CD21loCD27+ influenza-specific B cells, determined by HA probes and flow cytometry. Strikingly, 40% of HSCT recipients had markedly higher antibody responses towards A/H3N2 vaccine strain than healthy controls and showed cross-reactivity to antigenically drifted A/H3N2 strains by antibody landscape analysis. These superior humoral responses were associated with a greater time interval after HSCT, while multivariant analyses revealed the importance of pre-existing immune memory. Conversely, in HSCT recipients who did not respond to the first dose, the second IIV dose did not greatly improve their humoral response, although 50% of second-dose patients reached a seroprotective HAI titre for at least one of vaccine strains. CONCLUSIONS: Our study demonstrates efficient, although time-dependent, immune responses to IIV in HSCT recipients, and provides insights into influenza vaccination strategies targeted to immunocompromised high-risk groups.
  • Item
    No Preview Available
    Dual TCR-α Expression on Mucosal-Associated Invariant T Cells as a Potential Confounder of TCR Interpretation
    Suliman, S ; Kjer-Nielsen, L ; Iwany, SK ; Tamara, KL ; Loh, L ; Grzelak, L ; Kedzierska, K ; Ocampo, TA ; Corbett, AJ ; McCluskey, J ; Rossjohn, J ; Leon, SR ; Calderon, R ; Lecca-Garcia, L ; Murray, MB ; Moody, DB ; Van Rhijn, I (AMER ASSOC IMMUNOLOGISTS, 2022-03-15)
    Mucosal-associated invariant T (MAIT) cells are innate-like T cells that are highly abundant in human blood and tissues. Most MAIT cells have an invariant TCRα-chain that uses T cell receptor α-variable 1-2 (TRAV1-2) joined to TRAJ33/20/12 and recognizes metabolites from bacterial riboflavin synthesis bound to the Ag-presenting molecule MHC class I related (MR1). Our attempts to identify alternative MR1-presented Ags led to the discovery of rare MR1-restricted T cells with non-TRAV1-2 TCRs. Because altered Ag specificity likely alters affinity for the most potent known Ag, 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU), we performed bulk TCRα- and TCRβ-chain sequencing and single-cell-based paired TCR sequencing on T cells that bound the MR1-5-OP-RU tetramer with differing intensities. Bulk sequencing showed that use of V genes other than TRAV1-2 was enriched among MR1-5-OP-RU tetramerlow cells. Although we initially interpreted these as diverse MR1-restricted TCRs, single-cell TCR sequencing revealed that cells expressing atypical TCRα-chains also coexpressed an invariant MAIT TCRα-chain. Transfection of each non-TRAV1-2 TCRα-chain with the TCRβ-chain from the same cell demonstrated that the non-TRAV1-2 TCR did not bind the MR1-5-OP-RU tetramer. Thus, dual TCRα-chain expression in human T cells and competition for the endogenous β-chain explains the existence of some MR1-5-OP-RU tetramerlow T cells. The discovery of simultaneous expression of canonical and noncanonical TCRs on the same T cell means that claims of roles for non-TRAV1-2 TCR in MR1 response must be validated by TCR transfer-based confirmation of Ag specificity.