Microbiology & Immunology - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 28
  • Item
    No Preview Available
    Durable reprogramming of neutralizing antibody responses following Omicron breakthrough infection
    Lee, WS ; Tan, H-X ; Reynaldi, A ; Esterbauer, R ; Koutsakos, M ; Nguyen, J ; Amarasena, T ; Kent, HE ; Aggarwal, A ; Turville, SG ; Taiaroa, G ; Kinsella, P ; Liew, KC ; Tran, T ; Williamson, DA ; Cromer, D ; Davenport, MP ; Kent, SJ ; Juno, JA ; Khoury, DS ; Wheatley, AK (AMER ASSOC ADVANCEMENT SCIENCE, 2023-07)
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection of vaccinated individuals is increasingly common with the circulation of highly immune evasive and transmissible Omicron variants. Here, we report the dynamics and durability of recalled spike-specific humoral immunity following Omicron BA.1 or BA.2 breakthrough infection, with longitudinal sampling up to 8 months after infection. Both BA.1 and BA.2 infections robustly boosted neutralization activity against the infecting strain while expanding breadth against BA.4, although neutralization activity was substantially reduced for the more recent XBB and BQ.1.1 strains. Cross-reactive memory B cells against both ancestral and Omicron spike were predominantly expanded by infection, with limited recruitment of de novo Omicron-specific B cells or antibodies. Modeling of neutralization titers predicts that protection from symptomatic reinfection against antigenically similar strains will be durable but is undermined by new emerging strains with further neutralization escape.
  • Item
    No Preview Available
    Cutting Edge: SARS-CoV-2 Infection Induces Robust Germinal Center Activity in the Human Tonsil
    Tan, H-X ; Wragg, KM ; Kelly, HG ; Esterbauer, R ; Dixon, BJ ; Lau, JSY ; Flanagan, KL ; van de Sandt, CE ; Kedzierska, K ; McMahon, JH ; Wheatley, AK ; Juno, JA ; Kent, SJ (AMER ASSOC IMMUNOLOGISTS, 2022-05-15)
    Understanding the generation of immunity to SARS-CoV-2 in lymphoid tissues draining the site of infection has implications for immunity to SARS-CoV-2. We performed tonsil biopsies under local anesthesia in 19 subjects who had recovered from SARS-CoV-2 infection 24-225 d previously. The biopsies yielded >3 million cells for flow cytometric analysis in 17 subjects. Total and SARS-CoV-2 spike-specific germinal center B cells, and T follicular helper cells, were readily detectable in human tonsils early after SARS-CoV-2 infection, as assessed by flow cytometry. Responses were higher in samples within 2 mo of infection but still detectable in some subjects out to 7 mo following infection. We conclude the tonsils are a secondary lymphoid organ that develop germinal center responses to SARS-CoV-2 infection and could play a role in the long-term development of immunity.
  • Item
    Thumbnail Image
    Lung-resident memory B cells established after pulmonary influenza infection display distinct transcriptional and phenotypic profiles
    Tan, H-X ; Juno, JA ; Esterbauer, R ; Kelly, HG ; Wragg, KM ; Konstandopoulos, P ; Alcantara, S ; Alvarado, C ; Jones, R ; Starkey, G ; Wang, BZ ; Yoshino, O ; Tiang, T ; Grayson, ML ; Opdam, H ; D'Costa, R ; Vago, A ; Mackay, LK ; Gordon, CL ; Masopust, D ; Groom, JR ; Kent, SJ ; Wheatley, AK (AMER ASSOC ADVANCEMENT SCIENCE, 2022-01)
    Recent studies have established that memory B cells, largely thought to be circulatory in the blood, can take up long-term residency in inflamed tissues, analogous to widely described tissue-resident T cells. The dynamics of recruitment and retention of memory B cells to tissues and their immunological purpose remains unclear. Here, we characterized tissue-resident memory B cells (BRM) that are stably maintained in the lungs of mice after pulmonary influenza infection. Influenza-specific BRM were localized within inducible bronchus-associated lymphoid tissues (iBALTs) and displayed transcriptional signatures distinct from classical memory B cells in the blood or spleen while showing partial overlap with memory B cells in lung-draining lymph nodes. We identified lung-resident markers, including elevated expression of CXCR3, CCR6, and CD69, on hemagglutinin (HA)- and nucleoprotein (NP)-specific lung BRM. We found that CCR6 facilitates increased recruitment and/or retention of BRM in lungs and differentiation into antibody-secreting cells upon recall. Although expression of CXCR3 and CCR6 was comparable in total and influenza-specific memory B cells isolated across tissues of human donors, CD69 expression was higher in memory B cells from lung and draining lymph nodes of human organ donors relative to splenic and PBMC-derived populations, indicating that mechanisms underpinning BRM localization may be evolutionarily conserved. Last, we demonstrate that human memory B cells in lungs are transcriptionally distinct to populations in lung-draining lymph nodes or PBMCs. These data suggest that BRM may constitute a discrete component of B cell immunity, positioned at the lung mucosa for rapid humoral response against respiratory viral infections.
  • Item
    No Preview Available
    Establishment and recall of SARS-CoV-2 spike epitope-specific CD4+ T cell memory
    Wragg, KM ; Lee, WS ; Koutsakos, M ; Tan, H-X ; Amarasena, T ; Reynaldi, A ; Gare, G ; Konstandopoulos, P ; Field, KR ; Esterbauer, R ; Kent, HE ; Davenport, MP ; Wheatley, AK ; Kent, SJ ; Juno, JA (NATURE PORTFOLIO, 2022-05)
    Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and vaccination elicit CD4+ T cell responses to the spike protein, including circulating follicular helper T (cTFH) cells that correlate with neutralizing antibodies. Using a novel HLA-DRB1*15:01/S751 tetramer to track spike-specific CD4+ T cells, we show that primary infection or vaccination induces robust S751-specific CXCR5- and cTFH cell memory responses. Secondary exposure induced recall of CD4+ T cells with a transitory CXCR3+ phenotype, and drove expansion of cTFH cells transiently expressing ICOS, CD38 and PD-1. In both contexts, cells exhibited a restricted T cell antigen receptor repertoire, including a highly public clonotype and considerable clonotypic overlap between CXCR5- and cTFH populations. Following a third vaccine dose, the rapid re-expansion of spike-specific CD4+ T cells contrasted with the comparatively delayed increase in antibody titers. Overall, we demonstrate that stable pools of cTFH and memory CD4+ T cells established by infection and/or vaccination are efficiently recalled upon antigen reexposure and may contribute to long-term protection against SARS-CoV-2.
  • Item
    No Preview Available
    Robust and prototypical immune responses toward COVID-19 vaccine in First Nations peoples are impacted by comorbidities
    Zhang, W ; Kedzierski, L ; Chua, BY ; Mayo, M ; Lonzi, C ; Rigas, V ; Middleton, BF ; McQuilten, HA ; Rowntree, LC ; Allen, LF ; Purcell, RA ; Tan, H-X ; Petersen, J ; Chaurasia, P ; Mordant, F ; Pogorelyy, MV ; Minervina, AA ; Crawford, JC ; Perkins, GB ; Zhang, E ; Gras, S ; Clemens, EB ; Juno, JA ; Audsley, J ; Khoury, DS ; Holmes, NE ; Thevarajan, I ; Subbarao, K ; Krammer, F ; Cheng, AC ; Davenport, MP ; Grubor-Bauk, B ; Coates, PT ; Christensen, B ; Thomas, PG ; Wheatley, AK ; Kent, SJ ; Rossjohn, J ; Chung, AW ; Boffa, J ; Miller, A ; Lynar, S ; Nelson, J ; Nguyen, THO ; Davies, J ; Kedzierska, K (NATURE PORTFOLIO, 2023-06)
    High-risk groups, including Indigenous people, are at risk of severe COVID-19. Here we found that Australian First Nations peoples elicit effective immune responses to COVID-19 BNT162b2 vaccination, including neutralizing antibodies, receptor-binding domain (RBD) antibodies, SARS-CoV-2 spike-specific B cells, and CD4+ and CD8+ T cells. In First Nations participants, RBD IgG antibody titers were correlated with body mass index and negatively correlated with age. Reduced RBD antibodies, spike-specific B cells and follicular helper T cells were found in vaccinated participants with chronic conditions (diabetes, renal disease) and were strongly associated with altered glycosylation of IgG and increased interleukin-18 levels in the plasma. These immune perturbations were also found in non-Indigenous people with comorbidities, indicating that they were related to comorbidities rather than ethnicity. However, our study is of a great importance to First Nations peoples who have disproportionate rates of chronic comorbidities and provides evidence of robust immune responses after COVID-19 vaccination in Indigenous people.
  • Item
    No Preview Available
    SARS-CoV-2 breakthrough infection induces rapid memory and de novo T cell responses
    Koutsakos, M ; Reynaldi, A ; Lee, WS ; Nguyen, J ; Amarasena, T ; Taiaroa, G ; Kinsella, P ; Liew, KC ; Tran, T ; Kent, HE ; Tan, H-X ; Rowntree, LC ; Nguyen, THO ; Thomas, PG ; Kedzierska, K ; Petersen, J ; Rossjohn, J ; Williamson, DA ; Khoury, D ; Davenport, MP ; Kent, SJ ; Wheatley, AK ; Juno, JA (CELL PRESS, 2023-04-11)
    Although the protective role of neutralizing antibodies against COVID-19 is well established, questions remain about the relative importance of cellular immunity. Using 6 pMHC multimers in a cohort with early and frequent sampling, we define the phenotype and kinetics of recalled and primary T cell responses following Delta or Omicron breakthrough infection in previously vaccinated individuals. Recall of spike-specific CD4+ T cells was rapid, with cellular proliferation and extensive activation evident as early as 1 day post symptom onset. Similarly, spike-specific CD8+ T cells were rapidly activated but showed variable degrees of expansion. The frequency of activated SARS-CoV-2-specific CD8+ T cells at baseline and peak inversely correlated with peak SARS-CoV-2 RNA levels in nasal swabs and accelerated viral clearance. Our study demonstrates that a rapid and extensive recall of memory T cell populations occurs early after breakthrough infection and suggests that CD8+ T cells contribute to the control of viral replication in breakthrough SARS-CoV-2 infections.
  • Item
    No Preview Available
    Immune profiling of SARS-CoV-2 infection during pregnancy reveals NK cell and ?? T cell perturbations
    Habel, JR ; Chua, BY ; Kedzierski, L ; Selva, KJ ; Damelang, T ; Haycroft, ER ; Nguyen, THO ; Koay, H-F ; Nicholson, S ; McQuilten, HA ; Jia, X ; Allen, LF ; Hensen, L ; Zhang, W ; Sandt, CEVD ; Neil, JA ; Pragastis, K ; Lau, JSY ; Jumarang, J ; Allen, EK ; Amanant, F ; Krammer, F ; Wragg, KM ; Juno, JA ; Wheatley, AK ; Tan, H-X ; Pell, G ; Walker, S ; Audsley, J ; Reynaldi, A ; Thevarajan, I ; Denholm, JT ; Subbarao, K ; Davenport, MP ; Hogarth, PM ; Godfrey, DI ; Cheng, AC ; Tong, SYC ; Bond, K ; Williamson, DA ; McMahon, JH ; Thomas, PG ; Pannaraj, PS ; James, F ; Holmes, NE ; Smibert, OC ; Trubiano, JA ; Gordon, CL ; Chung, AW ; Whitehead, CL ; Kent, SJ ; Lappas, M ; Rowntree, LC ; Kedzierska, K (AMER SOC CLINICAL INVESTIGATION INC, 2023-03-22)
    Pregnancy poses a greater risk for severe COVID-19; however, underlying immunological changes associated with SARS-CoV-2 during pregnancy are poorly understood. We defined immune responses to SARS-CoV-2 in unvaccinated pregnant and nonpregnant women with acute and convalescent COVID-19, quantifying 217 immunological parameters. Humoral responses to SARS-CoV-2 were similar in pregnant and nonpregnant women, although our systems serology approach revealed distinct antibody and FcγR profiles between pregnant and nonpregnant women. Cellular analyses demonstrated marked differences in NK cell and unconventional T cell activation dynamics in pregnant women. Healthy pregnant women displayed preactivated NK cells and γδ T cells when compared with healthy nonpregnant women, which remained unchanged during acute and convalescent COVID-19. Conversely, nonpregnant women had prototypical activation of NK and γδ T cells. Activation of CD4+ and CD8+ T cells and T follicular helper cells was similar in SARS-CoV-2-infected pregnant and nonpregnant women, while antibody-secreting B cells were increased in pregnant women during acute COVID-19. Elevated levels of IL-8, IL-10, and IL-18 were found in pregnant women in their healthy state, and these cytokine levels remained elevated during acute and convalescent COVID-19. Collectively, we demonstrate perturbations in NK cell and γδ T cell activation in unvaccinated pregnant women with COVID-19, which may impact disease progression and severity during pregnancy.
  • Item
    Thumbnail Image
    The magnitude and timing of recalled immunity after breakthrough infection is shaped by SARS-CoV-2 variants
    Koutsakos, M ; Lee, WS ; Reynaldi, A ; Tan, H-X ; Gare, G ; Kinsella, P ; Liew, KC ; Taiaroa, G ; Williamson, DA ; Kent, HE ; Stadler, E ; Cromer, D ; Khoury, DS ; Wheatley, AK ; Juno, JA ; Davenport, MP ; Kent, SJ (CELL PRESS, 2022-07-12)
    Vaccination against SARS-CoV-2 protects from infection and improves clinical outcomes in breakthrough infections, likely reflecting residual vaccine-elicited immunity and recall of immunological memory. Here, we define the early kinetics of spike-specific humoral and cellular immunity after vaccination of seropositive individuals and after Delta or Omicron breakthrough infection in vaccinated individuals. Early longitudinal sampling revealed the timing and magnitude of recall, with the phenotypic activation of B cells preceding an increase in neutralizing antibody titers. While vaccination of seropositive individuals resulted in robust recall of humoral and T cell immunity, recall of vaccine-elicited responses was delayed and variable in magnitude during breakthrough infections and depended on the infecting variant of concern. While the delayed kinetics of immune recall provides a potential mechanism for the lack of early control of viral replication, the recall of antibodies coincided with viral clearance and likely underpins the protective effects of vaccination against severe COVID-19.
  • Item
    Thumbnail Image
    Landscape of human antibody recognition of the SARS-CoV-2 receptor binding domain
    Wheatley, AK ; Pymm, P ; Esterbauer, R ; Dietrich, MH ; Lee, WS ; Drew, D ; Kelly, HG ; Chan, L-J ; Mordant, FL ; Black, KA ; Adair, A ; Tan, H-X ; Juno, JA ; Wragg, KM ; Amarasena, T ; Lopez, E ; Selva, KJ ; Haycroft, ER ; Cooney, JP ; Venugopal, H ; Tan, LL ; Neill, MTO ; Allison, CC ; Cromer, D ; Davenport, MP ; Bowen, RA ; Chung, AW ; Pellegrini, M ; Liddament, MT ; Glukhova, A ; Subbarao, K ; Kent, SJ ; Tham, W-H (CELL PRESS, 2021-10-12)
    Potent neutralizing monoclonal antibodies are one of the few agents currently available to treat COVID-19. SARS-CoV-2 variants of concern (VOCs) that carry multiple mutations in the viral spike protein can exhibit neutralization resistance, potentially affecting the effectiveness of some antibody-based therapeutics. Here, the generation of a diverse panel of 91 human, neutralizing monoclonal antibodies provides an in-depth structural and phenotypic definition of receptor binding domain (RBD) antigenic sites on the viral spike. These RBD antibodies ameliorate SARS-CoV-2 infection in mice and hamster models in a dose-dependent manner and in proportion to in vitro, neutralizing potency. Assessing the effect of mutations in the spike protein on antibody recognition and neutralization highlights both potent single antibodies and stereotypic classes of antibodies that are unaffected by currently circulating VOCs, such as B.1.351 and P.1. These neutralizing monoclonal antibodies and others that bind analogous epitopes represent potentially useful future anti-SARS-CoV-2 therapeutics.
  • Item
    Thumbnail Image
    Integrated immune networks in SARS-CoV-2 infected pregnant women reveal differential NK cell and unconventional T cell activation
    Kedzierska, K ; Habel, J ; Chua, B ; Kedzierski, L ; Selva, K ; Damelang, T ; Haycroft, E ; Nguyen, T ; Koay, H-F ; Nicholson, S ; McQuilten, H ; Jia, X ; Allen, L ; Hensen, L ; Zhang, W ; de Sandt, CV ; Neil, J ; Amanat, F ; Krammer, F ; Wragg, K ; Juno, J ; Wheatley, A ; Tan, H-X ; Pell, G ; Audsley, J ; Thevarajan, I ; Denholm, J ; Subbarao, K ; Godfrey, D ; Cheng, A ; Tong, S ; Bond, K ; Williamson, D ; James, F ; Holmes, N ; Smibert, O ; Trubiona, J ; Gordon, C ; Chung, A ; Whitehead, C ; Kent, S ; Lappas, M ; Rowntree, L ( 2021)
    Although pregnancy poses a greater risk for severe COVID-19, the underlying immunological changes associated with SARS-CoV-2 infection during pregnancy are poorly understood. We defined immune responses to SARS-CoV-2 in pregnant and non-pregnant women during acute and convalescent COVID-19 up to 258 days post symptom onset, quantifying 217 immunological parameters. Additionally, matched maternal and cord blood were collected from COVID-19 convalescent pregnancies. Although serological responses to SARS-CoV-2 were similar in pregnant and non-pregnant women, cellular immune analyses revealed marked differences in key NK cell and unconventional T cell responses during COVID-19 in pregnant women. While NK, γδ T cells and MAIT cells displayed pre-activated phenotypes in healthy pregnant women when compared to non-pregnant age-matched women, activation profiles of these pre-activated NK and unconventional T cells remained unchanged at acute and convalescent COVID-19 in pregnancy. Conversely, activation dynamics of NK and unconventional T cells were prototypical in non-pregnant women in COVID-19. In contrast, activation of αβ CD4 + and CD8 + T cells, T follicular helper cells and antibody-secreting cells was similar in pregnant and non-pregnant women with COVID-19. Elevated levels of IL-1β, IFN-γ, IL-8, IL-18 and IL-33 were also found in pregnant women in their healthy state, and these cytokine levels remained elevated during acute and convalescent COVID-19. Collectively, our study provides the first comprehensive map of longitudinal immunological responses to SARS-CoV-2 infection in pregnant women, providing insights into patient management and education during COVID-19 pregnancy.