Infectious Diseases - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 12
  • Item
    Thumbnail Image
    Mosquitoes provide a transmission route between possums and humans for Buruli ulcer in southeastern Australia
    Mee, PT ; Buultjens, AH ; Oliver, J ; Brown, K ; Crowder, JC ; Porter, JL ; Hobbs, EC ; Judd, LM ; Taiaroa, G ; Puttharak, N ; Williamson, DA ; Blasdell, KR ; Tay, EL ; Feldman, R ; Muzari, MO ; Sanders, C ; Larsen, S ; Crouch, SR ; Johnson, PDR ; Wallace, JR ; Price, DJ ; Hoffmann, AA ; Gibney, KB ; Stinear, TP ; Lynch, SE (NATURE PORTFOLIO, 2024-02)
    Buruli ulcer, a chronic subcutaneous infection caused by Mycobacterium ulcerans, is increasing in prevalence in southeastern Australia. Possums are a local wildlife reservoir for M. ulcerans and, although mosquitoes have been implicated in transmission, it remains unclear how humans acquire infection. We conducted extensive field survey analyses of M. ulcerans prevalence among mosquitoes in the Mornington Peninsula region of southeastern Australia. PCR screening of trapped mosquitoes revealed a significant association between M. ulcerans and Aedes notoscriptus. Spatial scanning statistics revealed overlap between clusters of M. ulcerans-positive Ae. notoscriptus, M. ulcerans-positive possum excreta and Buruli ulcer cases, and metabarcoding analyses showed individual mosquitoes had fed on humans and possums. Bacterial genomic analysis confirmed shared single-nucleotide-polymorphism profiles for M. ulcerans detected in mosquitoes, possum excreta and humans. These findings indicate Ae. notoscriptus probably transmit M. ulcerans in southeastern Australia and highlight mosquito control as a Buruli ulcer prevention measure.
  • Item
    Thumbnail Image
    Non-SARS-CoV-2 respiratory viral detection and whole genome sequencing from COVID-19 rapid antigen test devices: a laboratory evaluation study
    Moso, MA ; Taiaroa, G ; Steinig, E ; Zhanduisenov, M ; Butel-Simoes, G ; Savic, I ; Taouk, ML ; Chea, S ; Moselen, J ; Keefe, JO' ; Prestedge, J ; Pollock, GL ; Khan, M ; Soloczynskyj, K ; Fernando, J ; Martin, GE ; Caly, L ; Barr, IG ; Tran, T ; Druce, J ; Lim, CK ; Williamson, DA (ELSEVIER, 2024-04)
    BACKGROUND: There has been high uptake of rapid antigen test device use for point-of-care COVID-19 diagnosis. Individuals who are symptomatic but test negative on COVID-19 rapid antigen test devices might have a different respiratory viral infection. We aimed to detect and sequence non-SARS-CoV-2 respiratory viruses from rapid antigen test devices, which could assist in the characterisation and surveillance of circulating respiratory viruses in the community. METHODS: We applied archival clinical nose and throat swabs collected between Jan 1, 2015, and Dec 31, 2022, that previously tested positive for a common respiratory virus (adenovirus, influenza, metapneumovirus, parainfluenza, rhinovirus, respiratory syncytial virus [RSV], or seasonal coronavirus; 132 swabs and 140 viral targets) on PCR to two commercially available COVID-19 rapid antigen test devices, the Panbio COVID-19 Ag Rapid Test Device and Roche SARS-CoV-2 Antigen Self-Test. In addition, we collected 31 COVID-19 rapid antigen test devices used to test patients who were symptomatic at The Royal Melbourne Hospital emergency department in Melbourne, Australia. We extracted total nucleic acid from the device paper test strips and assessed viral recovery using multiplex real-time PCR (rtPCR) and capture-based whole genome sequencing. Sequence and genome data were analysed through custom computational pipelines, including subtyping. FINDINGS: Of the 140 respiratory viral targets from archival samples, 89 (64%) and 88 (63%) were positive on rtPCR for the relevant taxa following extraction from Panbio or Roche rapid antigen test devices, respectively. Recovery was variable across taxa: we detected influenza A in nine of 18 samples from Panbio and seven of 18 from Roche devices; parainfluenza in 11 of 20 samples from Panbio and 12 of 20 from Roche devices; human metapneumovirus in 11 of 16 from Panbio and 14 of 16 from Roche devices; seasonal coronavirus in eight of 19 from Panbio and two of 19 from Roche devices; rhinovirus in 24 of 28 from Panbio and 27 of 28 from Roche devices; influenza B in four of 15 in both devices; and RSV in 16 of 18 in both devices. Of the 31 COVID-19 devices collected from The Royal Melbourne Hospital emergency department, 11 tested positive for a respiratory virus on rtPCR, including one device positive for influenza A virus, one positive for RSV, four positive for rhinovirus, and five positive for SARS-CoV-2. Sequences of target respiratory viruses from archival samples were detected in 55 (98·2%) of 56 samples from Panbio and 48 (85·7%) of 56 from Roche rapid antigen test devices. 98 (87·5%) of 112 viral genomes were completely assembled from these data, enabling subtyping for RSV and influenza viruses. All 11 samples collected from the emergency department had viral sequences detected, with near-complete genomes assembled for influenza A and RSV. INTERPRETATION: Non-SARS-CoV-2 respiratory viruses can be detected and sequenced from COVID-19 rapid antigen devices. Recovery of near full-length viral sequences from these devices provides a valuable opportunity to expand genomic surveillance programmes for public health monitoring of circulating respiratory viruses. FUNDING: Australian Government Medical Research Future Fund and Australian National Health and Medical Research Council.
  • 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
    Intra- and interhost genomic diversity of monkeypox virus
    Taouk, ML ; Steinig, E ; Taiaroa, G ; Savic, I ; Tran, T ; Higgins, N ; Tran, S ; Lee, A ; Braddick, M ; Moso, MA ; Chow, EPF ; Fairley, CK ; Towns, J ; Chen, MY ; Caly, L ; Lim, CK ; Williamson, DA (WILEY, 2023-08)
    The impact and frequency of infectious disease outbreaks demonstrate the need for timely genomic surveillance to inform public health responses. In the largest known outbreak of mpox, genomic surveillance efforts have primarily focused on high-incidence nations in Europe and the Americas, with a paucity of data from South-East Asia and the Western Pacific. Here we analyzed 102 monkeypox virus (MPXV) genomes sampled from 56 individuals in Melbourne, Australia. All genomes fell within the 2022 MPXV outbreak lineage (B.1), with likely onward local transmission detected. We observed within-host diversity and instances of co-infection, and highlight further examples of structural variation and apolipoprotein B editing complex-driven micro-evolution in the current MPXV outbreak. Updating our understanding of MPXV emergence and diversification will inform public health measures and enable monitoring of the virus' evolutionary trajectory throughout the mpox outbreak.
  • Item
    No Preview Available
    Maintaining genomic surveillance using whole-genome sequencing of SARS-CoV-2 from rapid antigen test devices
    Martin, GE ; Taiaroa, G ; Taouk, ML ; Savic, I ; O'Keefe, J ; Quach, R ; Prestedge, J ; Krysiak, M ; Caly, L ; Williamson, DA (ELSEVIER SCI LTD, 2022-10)
  • 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
    Thumbnail Image
    Analytical Sensitivity of Lateral Flow Devices against SARS-CoV-2 Omicron Subvariants BA.4, BA.5, and BA.2.75
    Mackenzie, C ; Batty, M ; Papadakis, G ; Stevens, L ; Yoga, Y ; Taiaroa, G ; Stefanatos, H ; Savic, I ; Tran, T ; Deerain, J ; Prestedge, J ; Druce, J ; Caly, L ; Williamson, DA ; Tang, Y-W (AMER SOC MICROBIOLOGY, 2022-11-16)
  • 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
    Monkeypox infection presenting as genital rash, Australia, May 2022
    Hammerschlag, Y ; MacLeod, G ; Papadakis, G ; Sanchez, AA ; Druce, J ; Taiaroa, G ; Savic, I ; Mumford, J ; Roberts, J ; Caly, L ; Friedman, D ; Williamson, DA ; Cheng, AC ; McMahon, JH (EUR CENTRE DIS PREVENTION & CONTROL, 2022-06-02)
    Rapid diagnosis and whole genome sequencing confirmed a case of monkeypox in an HIV-positive individual receiving antiretroviral therapy. The patient had a normal CD4+ T-cell count and suppressed HIV viral load and presented with a genital rash in Melbourne, Australia after return from Europe in May 2022. He subsequently developed systemic illness and disseminated rash and 11 days after symptom onset, he was hospitalised to manage painful bacterial cellulitis of the genital area.
  • Item
    Thumbnail Image
    Characterisation of Treponema pallidum lineages within the contemporary syphilis outbreak in Australia: a genomic epidemiological analysis
    Taouk, ML ; Taiaroa, G ; Pasricha, S ; Herman, S ; Chow, EPF ; Azzatto, F ; Zhang, B ; Sia, CM ; Duchene, S ; Lee, A ; Higgins, N ; Prestedge, J ; Lee, YW ; Thomson, NR ; Graves, B ; Meumann, E ; Gunathilake, M ; Hocking, JS ; Bradshaw, CS ; Beale, MA ; Howden, BP ; Chen, MY ; Fairley, CK ; Ingle, DJ ; Williamson, DA (ELSEVIER, 2022-06)
    BACKGROUND: The incidence of syphilis has increased markedly in the past decade in high-income countries, including Australia. To date, however, genomic studies of Treponema pallidum have focused mainly on the northern hemisphere. Here, we aimed to characterise the lineages of T pallidum driving the current syphilis epidemic in Australia. METHODS: In this genomic epidemiological analysis, using phylogenomic and phylodynamic analyses, we analysed 456 high-quality T pallidum genomes collected from clinical samples in Australia between Oct 19, 2005, and Dec 31, 2020, and contextualised this information with publicly available sequence data. We also performed detailed genomic characterisation of putative antimicrobial resistance determinants, in addition to correlating single-locus typing of the TP0548 allele with the T pallidum phylogeny. FINDINGS: Phylogenomic analyses identified four major sublineages circulating in Australia and globally, two belonging to the SS14 lineage, and two belonging to the Nichols lineage. Australian sublineages were further delineated into twelve subgroups, with five of the six largest subgroups associated with men who have sex with men, and the sixth lineage was predominantly associated with heterosexual people. Most Australian T pallidum genomes (398 [87%] of 456) were genotypically macrolide resistant, and TP0548 typing correlated significantly with T pallidum genomic subgroups. INTERPRETATION: These findings show that the current syphilis epidemic in Australia is driven by multiple lineages of T pallidum, rather than one distinct outbreak. Major subgroups of T pallidum in Australia have emerged within the past 30 years, are closely related to global lineages, and circulate across different sexual networks. In conjunction with improved testing and treatment, these data could better inform the control of syphilis in Australia. FUNDING: National Health and Medical Research Council, Australian Research Council.