Paediatrics (RCH) - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 35
  • Item
    Thumbnail Image
    Unconventional T Cell Immunity in the Lungs of Young Children with Cystic Fibrosis
    McElroy, R ; Talesh, GA ; Harpur, CM ; Carzino, R ; Corbett, AJ ; Pellicci, DG ; Ranganathan, S ; Sutton, P (IMR PRESS, 2022-05)
    BACKGROUND: People with Cystic Fibrosis (CF) develop pulmonary inflammation, chronic infection and structural lung damage early in life, with these manifestations being prevalent among preschool children and infants. While early immune events are believed to play critical roles in shaping the progression, severity and disease burden later in life, T cells and their subsets are poorly studied in the CF lung, particularly during the formative early stages of disease. METHODS: Using flow cytometry, we analyzed Mucosal Associated Invariant T (MAIT) cells, γδ T cells, and Natural Killer T (NKT)-like cells in bronchoalveolar lavage (BAL) samples from seventeen children with CF, aged two to six years old. The effect of age, sex and lung infections on the frequencies of these cells in BAL samples was analysed (grouped data were tested for normality and compared by t-test or Kruskal-Wallis analysis). RESULTS: No difference was noted in the proportions of unconventional T cells related to the sex or age of the children. The frequency of γδ T cells and MAIT cells appeared unchanged by infection status. However, viral infections were associated with a significant increase in the proportion of NKT-like cells. CONCLUSIONS: By evaluating T cells in the lungs of children during the early formative stages of CF, this study identified potentially important interactions between these cells and viral pathogens.
  • Item
    Thumbnail Image
    Human pluripotent stem cell-derived macrophages host Mycobacterium abscessus infection
    Sun, S ; See, M ; Nim, HT ; Strumila, K ; Ng, ES ; Hidalgo, A ; Ramialison, M ; Sutton, P ; Elefanty, AG ; Sarkar, S ; Stanley, EG (CELL PRESS, 2022-09-13)
    Human macrophages are a natural host of many mycobacterium species, including Mycobacterium abscessus (M. abscessus), an emerging pathogen affecting immunocompromised and cystic fibrosis patients with few available treatments. The search for an effective treatment is hindered by the lack of a tractable in vitro intracellular infection model. Here, we established a reliable model for M. abscessus infection using human pluripotent stem cell-derived macrophages (hPSC-macrophages). hPSC differentiation permitted reproducible generation of functional macrophages that were highly susceptible to M. abscessus infection. Electron microscopy demonstrated that M. abscessus was present in the hPSC-macrophage vacuoles. RNA sequencing analysis revealed a time-dependent host cell response, with differing gene and protein expression patterns post-infection. Engineered tdTOMATO-expressing hPSC-macrophages with GFP-expressing mycobacteria enabled rapid image-based high-throughput analysis of intracellular infection and quantitative assessment of antibiotic efficacy. Our study describes the first to our knowledge hPSC-based model for M. abscessus infection, representing a novel and accessible system for studying pathogen-host interaction and drug discovery.
  • Item
    Thumbnail Image
    Virology and immune dynamics reveal high household transmission of ancestral SARS-CoV-2 strain
    Tosif, S ; Haycroft, ER ; Sarkar, S ; Toh, ZQ ; Lien, AHD ; Donato, CM ; Selva, KJ ; Hoq, M ; Overmars, I ; Nguyen, J ; Lee, L-Y ; Clifford, V ; Daley, A ; Mordant, FL ; McVernon, J ; Mulholland, K ; Marcato, AJ ; Smith, MZ ; Curtis, N ; McNab, S ; Saffery, R ; Kedzierska, K ; Subarrao, K ; Burgner, D ; Steer, A ; Bines, JE ; Sutton, P ; Licciardi, P ; Chung, AW ; Neeland, MR ; Crawford, NW (WILEY, 2022-07)
    BACKGROUND: Household studies are crucial for understanding the transmission of SARS-CoV-2 infection, which may be underestimated from PCR testing of respiratory samples alone. We aim to combine the assessment of household mitigation measures; nasopharyngeal, saliva, and stool PCR testing; along with mucosal and systemic SARS-CoV-2-specific antibodies, to comprehensively characterize SARS-CoV-2 infection and transmission in households. METHODS: Between March and September 2020, we obtained samples from 92 participants in 26 households in Melbourne, Australia, in a 4-week period following the onset of infection with ancestral SARS-CoV-2 variants. RESULTS: The secondary attack rate was 36% (24/66) when using nasopharyngeal swab (NPS) PCR positivity alone. However, when respiratory and nonrespiratory samples were combined with antibody responses in blood and saliva, the secondary attack rate was 76% (50/66). SARS-CoV-2 viral load of the index case and household isolation measures were key factors that determine secondary transmission. In 27% (7/26) of households, all family members tested positive by NPS for SARS-CoV-2 and were characterized by lower respiratory Ct values than low transmission families (Median 22.62 vs. 32.91; IQR 17.06-28.67 vs. 30.37-34.24). High transmission families were associated with enhanced plasma antibody responses to multiple SARS-CoV-2 antigens and the presence of neutralizing antibodies. Three distinguishing saliva SARS-CoV-2 antibody features were identified according to age (IgA1 to Spike 1, IgA1 to nucleocapsid protein (NP)), suggesting that adults and children generate distinct mucosal antibody responses during the acute phase of infection. CONCLUSION: Utilizing respiratory and nonrespiratory PCR testing, along with the measurement of SARS-CoV-2-specific local and systemic antibodies, provides a more accurate assessment of infection within households and highlights some of the immunological differences in response between children and adults.
  • Item
    Thumbnail Image
    IL-33 promotes gastric tumour growth in concert with activation and recruitment of inflammatory myeloid cells.
    Tran, CP ; Scurr, M ; O'Connor, L ; Buzzelli, JN ; Ng, GZ ; Chin, SCN ; Stamp, LA ; Minamoto, T ; Giraud, AS ; Judd, LM ; Sutton, P ; Menheniott, TR (Impact Journals, LLC, 2022)
    Interleukin-33 (IL-33) is an IL-1 family cytokine known to promote T-helper (Th) type 2 immune responses that are often deregulated in gastric cancer (GC). IL-33 is overexpressed in human gastric tumours suggesting a role in driving GC progression although a causal link has not been proven. Here, we investigated the impact of IL-33 genetic deficiency in the well-characterized gp130 F/F mouse model of GC. Expression of IL-33 (and it's cognate receptor, ST2) was increased in human and mouse GC progression. IL-33 deficient gp130 F/F /Il33 -/- mice had reduced gastric tumour growth and reduced recruitment of pro-tumorigenic myeloid cells including key mast cell subsets and type-2 (M2) macrophages. Cell sorting of gastric tumours revealed that IL-33 chiefly localized to gastric (tumour) epithelial cells and was absent from tumour-infiltrating immune cells (except modest IL-33 enrichment within CD11b+ CX3CR1+CD64+MHCII+ macrophages). By contrast, ST2 was absent from gastric epithelial cells and localized exclusively within the (non-macrophage) immune cell fraction together with mast cell markers, Mcpt1 and Mcpt2. Collectively, we show that IL-33 is required for gastric tumour growth and provide evidence of a likely mechanism by which gastric epithelial-derived IL-33 drives mobilization of tumour-promoting inflammatory myeloid cells.
  • Item
    Thumbnail Image
    Superoxide dismutase from Helicobacter pylori suppresses the production of pro-inflammatory cytokines during in vivo infection
    Stent, A ; Every, AL ; Chionh, YT ; Ng, GZ ; Sutton, P (WILEY, 2018-02)
    BACKGROUND: Helicobacter pylori has undergone considerable adaptation to allow chronic persistence within the gastric environment. While H. pylori-associated diseases are driven by an excessive inflammation, severe gastritis is detrimental to colonization by this pathogen. Hence, H. pylori has developed strategies to minimize the severity of gastritis it triggers in its host. Superoxide dismutase (SOD) is well known for its role in protecting against oxidative attack; less recognized is its ability to inhibit immunity, shown for SOD from mammalian sources and those of some bacterial species. This study examined whether H. pylori SOD (HpSOD) has the ability to inhibit the host immune response to these bacteria. MATERIALS AND METHODS: The ability of recombinant HpSOD to modify the response to LPS was measured using mouse macrophages. A monoclonal antibody against HpSOD was generated and injected into H. pylori-infected mice. RESULTS: Addition of HpSOD to cultures of mouse macrophages significantly inhibited the pro-inflammatory cytokine response to LPS stimulation. A monoclonal antibody was generated that was specific for SOD from H. pylori. When injected into mice infected with H. pylori for 3 months, this antibody was readily detected in both sera and gastric tissues 5 days later. While treatment with anti-HpSOD had no effect on H. pylori colonization at this time point, it significantly increased the levels of a range of pro-inflammatory cytokines in the gastric tissues. This did not occur with antibodies against other antioxidant enzymes. CONCLUSIONS: SOD from H. pylori can inhibit the production of pro-inflammatory cytokine during in vivo infection.
  • Item
    Thumbnail Image
    Lack of small colony variants of Staphylococcus aureus from lower respiratory tract specimens
    Carzino, R ; Hart, E ; Sutton, P ; King, L ; Ranganathan, S (WILEY, 2017-05)
    BACKGROUND: Small-colony variants (SCVs) of Staphylococcus aureus are associated with worse lung disease in children with Cystic Fibrosis (CF), exhibit a higher resistance to antibiotics and co-colonize more commonly with Pseudomonas aeruginosa compared to the normal phenotype. The prevalence of SCVs in lower airway specimens from children with CF is largely unknown. METHODS: Each visible morphotype of S. aureus was subcultured onto horse blood agar (HBA) to enable identification of SCVs. RESULTS: Sixty-one samples from 41 children (mean age 11.7 (SD 5.3) years) were identified with a positive S. aureus culture from lower respiratory tract specimens collected in 2014-2015. None of the differing morphotypes isolated were identified as S. aureus SCVs. CONCLUSION: In a center where anti staphylococcal prophylaxis is adopted, S. aureus SCVs were not isolated from the lower airways specimens in young children with CF indicating that acquisition of small colony variant S. aureus may not be a significant clinical problem in young children with CF.
  • Item
    Thumbnail Image
    No evidence of a role for mitochondrial complex I in Helicobacter pylori pathogenesis
    Ng, GZ ; Ke, B-X ; Laskowski, A ; Thorburn, DR ; Sutton, P (WILEY, 2017-06)
    BACKGROUND: Complex I is the first enzyme complex in the mitochondrial respiratory chain, responsible for generating a large fraction of energy during oxidative phosphorylation. Recently, it has been identified that complex I deficiency can result in increased inflammation due to the generation of reactive oxygen species by innate immune cells. As a reduction in complex I activity has been demonstrated in human stomachs with atrophic gastritis, we investigated whether complex I deficiency could influence Helicobacter pylori pathogenesis. MATERIALS AND METHODS: Ndufs6gt/gt mice have a partial complex I deficiency. Complex I activity was quantified in the stomachs and immune cells of Ndufs6gt/gt mice by spectrophotometric assays. Ndufs6gt/gt mice were infected with H. pylori and bacterial colonization assessed by colony-forming assay, gastritis assessed histologically, and H. pylori -specific humoral response quantified by ELISA. RESULTS: The immune cells and stomachs of Ndufs6gt/gt mice were found to have significantly decreased complex I activity, validating the model for assessing the effects of complex I deficiency in H. pylori infection. However, there was no observable effect of complex I deficiency on either H. pylori colonization, the resulting gastritis, or the humoral response. CONCLUSIONS: Although complex I activity is described to suppress innate immune responses and is decreased during atrophic gastritis in humans, our data suggest it does not affect H. pylori pathogenesis.
  • Item
    Thumbnail Image
    Immune responses to SARS-CoV-2 in children of parents with symptomatic COVID-19
    Tosif, S ; Neeland, M ; Sutton, P ; Licciardi, P ; Sarkar, S ; Selva, K ; Do, LAH ; Donato, C ; Toh, ZQ ; Higgins, R ; de Sandt, CV ; Lemke, M ; Lee, C ; Shoffner, S ; Flanagan, K ; Arnold, K ; Mordant, F ; Mulholland, K ; Bines, J ; Dohle, K ; Pellicci, D ; Curtis, N ; McNab, S ; Steer, A ; Saffery, R ; Subbarao, K ; Chung, A ; Kedzierska, K ; Burgner, D ; Crawford, N ( 2020)
    Compared to adults, children with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have mild or asymptomatic infection, but the underlying immunological differences remain unclear. We describe clinical features, virology, longitudinal cellular and cytokine immune profile, SARS-CoV-2-specific serology and salivary antibody responses in a family of two parents with PCR-confirmed symptomatic SARS-CoV-2 infection and their three children, who were repeatedly SARS-CoV-2 PCR negative. Cellular immune profiles and cytokine responses of all children were similar to their parents at all timepoints. All family members had salivary anti-SARS-CoV-2 antibodies detected, predominantly IgA, that coincided with symptom resolution in 3 of 4 symptomatic members. Plasma from both parents and one child had IgG antibody detected against the S1 protein and virus neutralising activity ranging from just detectable to robust titers. Using a systems serology approach, we show that all family members demonstrated higher levels of SARS-CoV-2-specific antibody features than healthy controls. These data indicate that children can mount an immune response to SARS-CoV-2 without virological evidence of infection. This raises the possibility that despite chronic exposure, immunity in children prevents establishment of SARS-CoV-2 infection. Relying on routine virological and serological testing may therefore not identify exposed children, with implications for epidemiological and clinical studies across the life-span.
  • Item
    Thumbnail Image
    NOD1 is required for Helicobacter pylori induction of IL-33 responses in gastric epithelial cells
    Tran, LS ; Tran, D ; De Paoli, A ; D'Costa, K ; Creed, SJ ; Ng, GZ ; Le, L ; Sutton, P ; Silke, J ; Nachbur, U ; Ferrero, RL (WILEY, 2018-05)
    Helicobacter pylori (H. pylori) causes chronic inflammation which is a key precursor to gastric carcinogenesis. It has been suggested that H. pylori may limit this immunopathology by inducing the production of interleukin 33 (IL-33) in gastric epithelial cells, thus promoting T helper 2 immune responses. The molecular mechanism underlying IL-33 production in response to H. pylori infection, however, remains unknown. In this study, we demonstrate that H. pylori activates signalling via the pathogen recognition molecule Nucleotide-Binding Oligomerisation Domain-Containing Protein 1 (NOD1) and its adaptor protein receptor-interacting serine-threonine Kinase 2, to promote production of both full-length and processed IL-33 in gastric epithelial cells. Furthermore, IL-33 responses were dependent on the actions of the H. pylori Type IV secretion system, required for activation of the NOD1 pathway, as well as on the Type IV secretion system effector protein, CagA. Importantly, Nod1+/+ mice with chronic H. pylori infection exhibited significantly increased gastric IL-33 and splenic IL-13 responses, but decreased IFN-γ responses, when compared with Nod1-/- animals. Collectively, our data identify NOD1 as an important regulator of mucosal IL-33 responses in H. pylori infection. We suggest that NOD1 may play a role in protection against excessive inflammation.
  • Item
    Thumbnail Image
    Systems serology detects functionally distinct coronavirus antibody features in children and elderly
    Selva, KJ ; van de Sandt, CE ; Lemke, MM ; Lee, CY ; Shoffner, SK ; Chua, BY ; Davis, SK ; Nguyen, THO ; Rowntree, LC ; Hensen, L ; Koutsakos, M ; Wong, CY ; Mordant, F ; Jackson, DC ; Flanagan, KL ; Crowe, J ; Tosif, S ; Neeland, MR ; Sutton, P ; Licciardi, P ; Crawford, NW ; Cheng, AC ; Doolan, DL ; Amanat, F ; Krammer, F ; Chappell, K ; Modhiran, N ; Watterson, D ; Young, P ; Lee, WS ; Wines, BD ; Hogarth, PM ; Esterbauer, R ; Kelly, HG ; Tan, H-X ; Juno, JA ; Wheatley, AK ; Kent, SJ ; Arnold, KB ; Kedzierska, K ; Chung, AW (NATURE PORTFOLIO, 2021-04-01)
    The hallmarks of COVID-19 are higher pathogenicity and mortality in the elderly compared to children. Examining baseline SARS-CoV-2 cross-reactive immunological responses, induced by circulating human coronaviruses (hCoVs), is needed to understand such divergent clinical outcomes. Here we show analysis of coronavirus antibody responses of pre-pandemic healthy children (n = 89), adults (n = 98), elderly (n = 57), and COVID-19 patients (n = 50) by systems serology. Moderate levels of cross-reactive, but non-neutralizing, SARS-CoV-2 antibodies are detected in pre-pandemic healthy individuals. SARS-CoV-2 antigen-specific Fcγ receptor binding accurately distinguishes COVID-19 patients from healthy individuals, suggesting that SARS-CoV-2 infection induces qualitative changes to antibody Fc, enhancing Fcγ receptor engagement. Higher cross-reactive SARS-CoV-2 IgA and IgG are observed in healthy elderly, while healthy children display elevated SARS-CoV-2 IgM, suggesting that children have fewer hCoV exposures, resulting in less-experienced but more polyreactive humoral immunity. Age-dependent analysis of COVID-19 patients, confirms elevated class-switched antibodies in elderly, while children have stronger Fc responses which we demonstrate are functionally different. These insights will inform COVID-19 vaccination strategies, improved serological diagnostics and therapeutics.