Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Sci Rep ; 12(1): 18090, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302796

ABSTRACT

The South Pacific Gyre (SPG) plays a vital role in regulating Southern Hemisphere climate and ecosystems. The SPG has been intensifying since the twentieth century due to changes in large scale wind forcing. These changes result from variability in the Southern Annular Mode (SAM), causing warming along the eastern SPG which affects local ecosystems. However, our understanding of SPG variability on timescales greater than several decades is poor due to limited observations. Marine sediment cores are traditionally used to determine if recent ocean trends are anomalous, but rarely capture centennial variability in the southwest Pacific and limit our understanding of SPG variability. Here we capture centennial SPG dynamics using a novel high-resolution paleocirculation archive: radiocarbon reservoir ages (R) and local reservoir corrections (∆R) in SPG deep-sea black corals. We find black coral R and ∆R correlates with SAM reconstructions over 0-1000 cal BP and 2000-3000 cal BP. We propose this correlation indicates varying transport of well-ventilated subtropical waters resulting from SPG and SAM interactions. We reconstruct several 'spin up' cycles reminiscent of the recent gyre intensification, which has been attributed to anthropogenic causes. This implies gyre strength and SAM show natural co-variability on anthropogenic timescales which should factor into future climate projections.


Subject(s)
Anthozoa , Ecosystem , Animals , Geologic Sediments , Pacific Ocean
2.
Stem Cell Reports ; 17(9): 2156-2166, 2022 09 13.
Article in English | MEDLINE | ID: mdl-35985333

ABSTRACT

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.


Subject(s)
Mycobacterium Infections, Nontuberculous , Mycobacterium abscessus , Mycobacterium , Pluripotent Stem Cells , Humans , Macrophages/metabolism , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium Infections, Nontuberculous/microbiology
3.
Article in English | MEDLINE | ID: mdl-35871459

ABSTRACT

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.


Subject(s)
COVID-19 , SARS-CoV-2 , Adult , Antibodies, Viral , COVID-19/diagnosis , Child , Humans , Immunoglobulin A
4.
Oncotarget ; 13: 785-799, 2022.
Article in English | MEDLINE | ID: mdl-35677533

ABSTRACT

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.


Subject(s)
Interleukin-33 , Stomach Neoplasms , Animals , Cytokine Receptor gp130 , Cytokines , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/metabolism , Mice , Mice, Knockout , Myeloid Cells/metabolism , Signal Transduction , Stomach Neoplasms/pathology
5.
Front Biosci (Landmark Ed) ; 27(5): 149, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35638416

ABSTRACT

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.


Subject(s)
Cystic Fibrosis , T-Lymphocytes/immunology , Virus Diseases , Child , Child, Preschool , Cystic Fibrosis/complications , Cystic Fibrosis/immunology , Cystic Fibrosis/virology , Humans , Infant , Lung/immunology , Lung/virology
6.
Nat Commun ; 12(1): 2037, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795692

ABSTRACT

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.


Subject(s)
Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibody Formation/immunology , SARS-CoV-2/immunology , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/immunology , COVID-19 Vaccines/immunology , Child , Child, Preschool , Cross Reactions/immunology , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin M/blood , Immunoglobulin M/immunology , Middle Aged , Receptors, IgG/immunology , Spike Glycoprotein, Coronavirus/immunology , Young Adult
7.
Scand J Immunol ; 94(1): e13040, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33759233

ABSTRACT

Cystic Fibrosis (CF) is primarily a progressive lung disease, characterized by chronic pulmonary infections with opportunistic pathogens. Such infections typically commence early in life, producing an inflammatory response marked by IL-8 chemokine production and neutrophilic infiltration, major contributory factors in CF progression. Studying this inflammation, especially early in life, is critical for developing new strategies for preventing or slowing disruption to the structural integrity of the CF airways. However, evaluating the immune responses of bronchoalveolar lavage (BAL) cells from children with CF faces technical challenges, including contamination carried from the lung due to pre-existing infections and low cell number availability. Here, we describe a technique for preparing BAL cells from young children with CF and using those cells in a bacterial stimulation assay. Initial antibiotic treatment proved essential for preventing resident bacteria from overgrowing BAL cell cultures, or non-specifically activating the cells. ACTB, identified as an optimal reference gene, was validated for accurate analysis of gene expression in these cells. Pseudomonas aeruginosa and Staphylococcus aureus were used as bacterial stimulants to evaluate the immune response of BAL cells from young children with CF. Addition of gentamicin prevented bacterial overgrowth, although if added after 3 hours of culture an extremely variable response resulted, with the bacteria causing a suppressive effect in some cultures. Addition of gentamicin after 1 hour of culture completely prevented this suppressive effect. This technique was then able to reproducibly measure the IL-8 response to stimulation with S. aureus and P. aeruginosa, including co-stimulation with both bacteria.


Subject(s)
Bronchoalveolar Lavage Fluid/immunology , Cystic Fibrosis/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Bronchoalveolar Lavage/methods , Bronchoalveolar Lavage Fluid/microbiology , Child , Child, Preschool , Cystic Fibrosis/microbiology , Female , Humans , Infant , Inflammation/immunology , Inflammation/microbiology , Lung/immunology , Lung/microbiology , Male , Pseudomonas Infections/microbiology , Staphylococcal Infections/microbiology
8.
Nat Commun ; 12(1): 1084, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597531

ABSTRACT

Children have mild severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) confirmed disease (COVID-19) compared to adults and the immunological mechanisms underlying this difference remain unclear. Here, we report acute and convalescent innate immune responses in 48 children and 70 adults infected with, or exposed to, SARS-CoV-2. We find clinically mild SARS-CoV-2 infection in children is characterised by reduced circulating subsets of monocytes (classical, intermediate, non-classical), dendritic cells and natural killer cells during the acute phase. In contrast, SARS-CoV-2-infected adults show reduced proportions of non-classical monocytes only. We also observe increased proportions of CD63+ activated neutrophils during the acute phase to SARS-CoV-2 in infected children. Children and adults exposed to SARS-CoV-2 but negative on PCR testing display increased proportions of low-density neutrophils that we observe up to 7 weeks post exposure. This study characterises the innate immune response during SARS-CoV-2 infection and household exposure in children.


Subject(s)
Antibodies, Viral/immunology , COVID-19/immunology , Immunity, Innate/immunology , SARS-CoV-2/immunology , Adolescent , Adult , COVID-19/blood , COVID-19/virology , Child , Child, Preschool , Dendritic Cells/immunology , Eosinophils/immunology , Humans , Infant , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Middle Aged , Neutrophils/immunology , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Young Adult
9.
Glob Chang Biol ; 27(7): 1470-1484, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33502819

ABSTRACT

Fisheries harvest has pervasive impacts on wild fish populations, including the truncation of size and age structures, altered population dynamics and density, and modified habitat and assemblage composition. Understanding the degree to which harvest-induced impacts increase the sensitivity of individuals, populations and ultimately species to environmental change is essential to ensuring sustainable fisheries management in a rapidly changing world. Here we generated multiple long-term (44-62 years), annually resolved, somatic growth chronologies of four commercially important fishes from New Zealand's coastal and shelf waters. We used these novel data to investigate how regional- and basin-scale environmental variability, in concert with fishing activity, affected individual somatic growth rates and the magnitude of spatial synchrony among stocks. Changes in somatic growth can affect individual fitness and a range of population and fishery metrics such as recruitment success, maturation schedules and stock biomass. Across all species, individual growth benefited from a fishing-induced release of density controls. For nearshore snapper and tarakihi, regional-scale wind and temperature also additively affected growth, indicating that future climate change-induced warming and potentially strengthened winds will initially promote the productivity of more poleward populations. Fishing increased the sensitivity of deep-water hoki and ling growth to the Interdecadal Pacific Oscillation (IPO). A forecast shift to a positive IPO phase, in concert with current harvest strategies, will likely promote individual hoki and ling growth. At the species level, historical fishing practices and IPO synergized to strengthen spatial synchrony in average growth between stocks separated by 400-600 nm of ocean. Increased spatial synchrony can, however, increase the vulnerability of stocks to deleterious stochastic events. Together, our individual- and species-level results show how fishing and environmental factors can conflate to initially promote individual growth but then possibly heighten the sensitivity of stocks to environmental change.


Subject(s)
Climate Change , Fisheries , Animals , Ecosystem , Fishes , Humans , New Zealand , Population Dynamics
10.
Nat Commun ; 11(1): 5703, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177504

ABSTRACT

Compared to adults, children with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have predominantly mild or asymptomatic infections, but the underlying immunological differences remain unclear. Here, 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 tested repeatedly SARS-CoV-2 PCR negative. Cellular immune profiles and cytokine responses of all children are similar to their parents at all timepoints. All family members have salivary anti-SARS-CoV-2 antibodies detected, predominantly IgA, that coincide with symptom resolution in 3 of 4 symptomatic members. Plasma from both parents and one child have IgG antibody against the S1 protein and virus-neutralizing activity detected. Using a systems serology approach, we demonstrate higher levels of SARS-CoV-2-specific antibody features of these family members compared to healthy controls. These data indicate that children can mount an immune response to SARS-CoV-2 without virological confirmation of infection, raising the possibility that immunity in children can prevent the establishment of SARS-CoV-2 infection. Relying on routine virological and serological testing may not identify exposed children, with implications for epidemiological and clinical studies across the life-span.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/immunology , Coronavirus Infections/transmission , Cytokines/blood , Pneumonia, Viral/transmission , Saliva/immunology , Adult , Antibodies, Viral/immunology , Australia , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , COVID-19 , Child , Child, Preschool , Coronavirus Infections/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin A/blood , Immunoglobulin A/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Monocytes/immunology , Pandemics , Parents , Pneumonia, Viral/immunology , SARS-CoV-2 , Serologic Tests , Spike Glycoprotein, Coronavirus/immunology
11.
Article in English | MEDLINE | ID: mdl-32793510

ABSTRACT

The cell surface mucin MUC1 is an important host factor limiting Helicobacter pylori (H. pylori) pathogenesis in both humans and mice by providing a protective barrier and modulating mucosal epithelial and leukocyte responses. The aim of this study was to establish the time-course of molecular events in MUC1-modulated gene expression profiles in response to H. pylori infection in wild type (WT) and MUC1-deficient mice using microarray-determined mRNA expression, gene network analysis and Ingenuity Pathway Analysis (IPA). A time-course over the first 72 h of infection showed significantly higher mucosal loads of bacteria at 8 h of infection in Muc1-/- mice compared with WT, confirming its importance in the early stages of infection (P = 0.0003). Microarray analysis revealed 266 differentially expressed genes at one or more time-points over 72 h in the gastric mucosa of Muc1-/- mice compared with WT control using a threshold of 2-fold change. The SPINK1 pancreatic cancer canonical pathway was strongly inhibited in Muc1-/- mice compared with WT at sham and 8 h infection (P = 6.08E-14 and P = 2.25 E-19, respectively) but potently activated at 24 and 72 h post-infection (P = 1.38E-22 and P = 5.87E-13, respectively). The changes in this pathway are reflective of higher expression of genes mediating digestion and absorption of lipids, carbohydrates, and proteins at sham and 8 h infection in the absence of MUC1, but that this transcriptional signature is highly down regulated as infection progresses in the absence of MUC1. Uninfected Muc1-/- gastric tissue was highly enriched for expression of factors involved in lipid metabolism and 8 h infection further activated this network compared with WT. As infection progressed, a network of antimicrobial and anti-inflammatory response genes was more highly activated in Muc1-/- than WT mice. Key target genes identified by time-course microarrays were independently validated using RT-qPCR. These results highlight the dynamic interplay between the host and H. pylori, and the role of MUC1 in host defense, and provide a general picture of changes in cellular gene expression modulated by MUC1 in a time-dependent manner in response to H. pylori infection.


Subject(s)
Gastric Mucosa , Helicobacter Infections , Mucin-1/genetics , Animals , Helicobacter pylori , Mice , Transcriptome
12.
Am J Physiol Gastrointest Liver Physiol ; 319(2): G175-G188, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32538140

ABSTRACT

Gastrokines (GKNs) are anti-inflammatory proteins secreted by gastric epithelial (surface mucous and pit) cells, with their aberrant loss of expression causally linked to premalignant inflammation and gastric cancer (GC). Transcriptional mechanisms accounting for GKN expression loss have not been elucidated. Using human clinical cohorts, mouse transgenics, bioinformatics, and transfection/reporter assays, we report a novel mechanism of GKN gene transcriptional regulation and its impairment in GC. GKN1/GKN2 loss is highly coordinated, with both genes showing parallel downregulation during human and mouse GC development, suggesting joint transcriptional control. In BAC transgenic studies, we defined a 152-kb genomic region surrounding the human GKN1/GKN2 genes sufficient to direct their tissue- and lineage-restricted expression. A screen of the 152-kb region for candidate regulatory elements identified a DNase I hypersensitive site (CR2) located 4 kb upstream of the GKN1 gene. CR2 showed overlapping enrichment of enhancer-related histone marks (H3K27Ac), a consensus binding site (GRE) for the glucocorticoid receptor (GR), strong GR occupancy in ChIP-seq data sets and, critically, exhibited dexamethasone-sensitive enhancer activity in reporter assays. Strikingly, GR showed progressive expression loss, paralleling that of GKN1/2, in human and mouse GC, suggesting desensitized glucocorticoid signaling as a mechanism underlying GKN loss. Finally, mouse adrenalectomy studies revealed a critical role for endogenous glucocorticoids in sustaining correct expression (and anti-inflammatory restraint) of GKNs in vivo. Together, these data link the coordinate expression of GKNs to a glucocorticoid-responsive and likely shared transcriptional enhancer mechanism, with its compromised activation contributing to dual GKN loss during GC progression.NEW & NOTEWORTHY Gastrokine 2 (GKN2) is an anti-inflammatory protein produced by the gastric epithelium. GKN2 expression is progressively lost during gastric cancer (GC), which is believed to play a casual role in GC development. Here, we use bacterial artificial chromosome transgenic studies to identify a glucocorticoid-responsive enhancer element that likely governs expression of GKN1/GKN2, which, via parallel expression loss of the anti-inflammatory glucocorticoid receptor, reveals a novel mechanism to explain the loss of GKN2 during GC pathogenesis.


Subject(s)
Carrier Proteins/metabolism , Glucocorticoids/pharmacology , Peptide Hormones/metabolism , Stomach Neoplasms/metabolism , A549 Cells , Animals , Carrier Proteins/genetics , Chromosomes, Artificial, Bacterial , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Multigene Family , Peptide Hormones/genetics
14.
Helicobacter ; 25(2): e12681, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32088936

ABSTRACT

BACKGROUND: Helicobacter pylori occupy a unique niche, located within the mucus layer lining the stomach, and attached to the apical surface of the gastric epithelium. As such, antibodies would be expected to play a major role in regulating infection and/or pathogenesis. However, experiments using antibody-deficient mice to study gastric helicobacter infection have yielded inconsistent results, although some pointed toward antibodies increasing colonization levels and decreasing gastritis severity. The variability in these studies is possibly due to their use of nonmatched wild-type controls. This current study presents the first evaluation of the role of antibodies in H pylori infection by comparing antibody-deficient mice with matched wild-type siblings. METHODS: Matched wild-type and antibody-deficient µMT mice were generated by heterozygous crossings. In two separate experiments, appropriately genotyped sibling littermates were infected with H pylori for 4 months and then sera and stomachs were collected. RESULTS: There was no difference in H pylori colonization levels between infected µMT mice and sibling wild-type controls. Similarly, there was no significant difference in the severity of gastritis between these groups of mice, although there was a trend toward less severe gastritis in µMT mice which was supported by a significantly lower IFNγ (Th1) gastric cytokine response. CONCLUSIONS: Comparing matched antibody-deficient and antibody-competent mice indicates that an antibody response does not influence H pylori colonization levels. Contrary to previous studies, these results suggest antibodies might have a minor pro-inflammatory effect by promoting gastric Th1 cytokines, although this did not translate to a significant effect on gastritis severity.


Subject(s)
Antibodies/immunology , Helicobacter Infections/immunology , Helicobacter pylori , Animals , Cytokines/metabolism , Gastric Mucosa/microbiology , Gastritis/microbiology , Gastritis/pathology , Helicobacter Infections/pathology , Helicobacter pylori/growth & development , Helicobacter pylori/pathogenicity , Immunity, Mucosal , Mice , Mice, Inbred C57BL , Stomach/microbiology , Stomach/pathology
15.
Hum Immunol ; 80(10): 878-882, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31213370

ABSTRACT

Mucin 1 is a cell-membrane associated mucin, expressed on epithelial and immune cells that helps protect against pathogenic infections. In humans, MUC1 is highly polymorphic, predominantly due to the presence of a variable number tandem repeat (VNTR) region in the extracellular domain that results in MUC1 molecules of typically either short or long length. A genetic link is known between these MUC1 polymorphisms and inflammation-driven diseases, although the mechanism is not fully understood. We previously showed that MUC1 on murine macrophages specifically restricts activation of the NLRP3 inflammasome, thereby repressing inflammation. This study evaluated the effect of MUC1 VNTR polymorphisms on activity of the NLRP3 inflammasome in human macrophages, finding that long MUC1 alleles correlated with increased IL-1ß production following NLRP3 inflammasome activation. This indicates that the length of MUC1 can influence IL-1ß production, thus providing the first evidence of an immune-modulatory role of MUC1 VNTR polymorphisms in human macrophages.


Subject(s)
Inflammasomes/immunology , Macrophages/immunology , Mucin-1/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polymorphism, Genetic/immunology , Adolescent , Alleles , Child , Gene Frequency/genetics , Genotype , Healthy Volunteers , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Male , Minisatellite Repeats/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Nigericin/pharmacology , Signal Transduction/drug effects
16.
Vaccine ; 37(50): 7295-7299, 2019 11 28.
Article in English | MEDLINE | ID: mdl-29627231

ABSTRACT

Gastric adenocarcinoma is globally the third leading cause of death due to malignancy, with the bulk of this disease burden being suffered by low and middle income countries (LMIC), especially in Asia. The majority of these cancers develop as a result of a chronic gastritis that arises in response to infection with the stomach-dwelling bacterium, Helicobacter pylori. A vaccine against this pathogen would therefore be a powerful tool for preventing gastric adenocarcinoma. However, notwithstanding a proof-of-concept that vaccination can protect children from acquisition of H. pylori infection, there are currently no advanced vaccine candidates with only a single vaccine in Phase I clinical trial. Further, the development of a vaccine against H. pylori is not a current strategic priority of major pharmaceutical companies despite the large global disease burden. Given the involvement of such companies is likely to be critical for late stage development, there is therefore a need for an increased appreciation of the burden of this disease in LMIC and more investment to reinvigorate research in H. pylori vaccine Research and Development.


Subject(s)
Adenocarcinoma/prevention & control , Bacterial Vaccines/biosynthesis , Helicobacter Infections/prevention & control , Helicobacter pylori/drug effects , Stomach Neoplasms/prevention & control , Adenocarcinoma/etiology , Adenocarcinoma/immunology , Adenocarcinoma/microbiology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Vaccines/genetics , Biomedical Research/organization & administration , Child , Clinical Trials as Topic , Developing Countries/economics , Disease Models, Animal , Drug Industry/trends , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/immunology , Helicobacter pylori/pathogenicity , Humans , Macaca mulatta , Mice , Stomach Neoplasms/etiology , Stomach Neoplasms/immunology , Stomach Neoplasms/microbiology , Vaccination/methods , Vaccines, Subunit
17.
Am J Physiol Gastrointest Liver Physiol ; 316(2): G251-G262, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30520693

ABSTRACT

Expression of the cytokine IL-11 is elevated in human Helicobacter pylori infection and progressively increases with worsening gastric pathology. Additionally, IL-11 is required for tumor development in STAT3-dependent murine models of gastric cancer (GC) and, when administered acutely, causes resolving atrophic gastritis. However, it is unclear whether locally elevated IL-11 ligand expression can, in isolation from oncogenic gp130-JAK-STAT pathway mutations, initiate GC pathogenesis. Here we developed a transgenic mouse model of stomach-specific (keratin 19 promoter) IL-11 ligand overexpression. Keratin 19 promoter-IL-11 transgenic ( K19-IL11Tg) mice showed specific IL-11 overexpression in gastric corpus and antrum but not elsewhere in the gastrointestinal tract or in other tissues. K19-IL11Tg mice developed spontaneous premalignant disease of the gastric epithelium, progressing from atrophic gastritis to TFF2-positive metaplasia and severe epithelial hyperplasia, including adenoma-like lesions in a subset of older (1 yr old) animals. Although locally advanced, the hyperplastic lesions remained noninvasive. H. pylori infection in K19-IL11Tg mice accelerated some aspects of the premalignant phenotype. Finally, K19-IL11Tg mice had splenomegaly in association with elevated serum IL-11, with spleens showing an expanded myeloid compartment. Our results provide direct in vivo functional evidence that stomach-specific overexpression of IL-11, in isolation from germline gp130-JAK-STAT3 genetic drivers, is sufficient for premalignant progression. These findings have important functional implications for human GC, in which frequent IL-11 overexpression occurs in the reported absence of somatic mutations in gp130 signaling components. NEW & NOTEWORTHY We provide direct in vivo functional evidence that stomach-specific overexpression of the cytokine IL-11, in isolation from gp130-JAK-STAT3 pathway mutations, can trigger spontaneous atrophic gastritis progressing to locally advanced epithelial hyperplasia (but not dysplasia or carcinoma), which does not require, but may be accelerated by, concomitant Helicobacter pylori infection.


Subject(s)
Cytokine Receptor gp130/metabolism , Gastric Mucosa/metabolism , Hyperplasia/metabolism , Interleukin-11/metabolism , STAT3 Transcription Factor/metabolism , Animals , Helicobacter Infections/complications , Hyperplasia/genetics , Interleukin-11/genetics , Mice, Transgenic , Precancerous Conditions/metabolism , Stomach/pathology , Stomach Neoplasms/metabolism
18.
Cell Microbiol ; 20(5): e12826, 2018 05.
Article in English | MEDLINE | ID: mdl-29392836

ABSTRACT

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.


Subject(s)
Helicobacter Infections/genetics , Helicobacter pylori/pathogenicity , Interleukin-33/genetics , Nod1 Signaling Adaptor Protein/genetics , Receptors, Interleukin-13/genetics , Animals , Cell Line , Epithelial Cells/microbiology , Epithelial Cells/pathology , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Helicobacter pylori/immunology , Humans , Immunity, Mucosal/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Interferon-gamma/genetics , Mice , Th2 Cells/immunology , Th2 Cells/microbiology
19.
Helicobacter ; 23(1)2018 Feb.
Article in English | MEDLINE | ID: mdl-29235197

ABSTRACT

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.


Subject(s)
Cytokines/immunology , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter pylori/enzymology , Host-Pathogen Interactions/immunology , Superoxide Dismutase/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Cells, Cultured , Down-Regulation/drug effects , Female , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Inflammation Mediators/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Species Specificity , Superoxide Dismutase/antagonists & inhibitors
20.
Virulence ; 8(8): 1631-1642, 2017 11 17.
Article in English | MEDLINE | ID: mdl-28605238

ABSTRACT

Streptococcus pneumoniae is a bacterial pathogen that commonly resides in the human nasopharynx, typically without causing any disease. However, in some cases these bacteria migrate from the nasopharynx to other sites of the body such as the lungs and bloodstream causing pneumonia and sepsis, respectively. This study used a mouse model of infection to investigate the potential role of Mucin 1 (MUC1), a cell membrane-associated glycoprotein known for playing a key barrier role at mucosal surfaces, in regulating this process. Wildtype (WT) and MUC1-deficient (Muc1-/-) mice were infected intranasally with an invasive strain of S. pneumoniae and bacterial loads in the nasopharynx, lungs, and blood were analyzed. Lungs were graded histologically for inflammation and cytokine profiles in the lungs analyzed by ELISA. While there was no difference in pneumococcal colonization of the nasopharynx between WT and Muc1-/- mice, infected Muc1-/- mice showed high pneumococcal loads in their lungs 16 hours post-infection, as well as bacteremia. In contrast, infected WT mice cleared the pneumococci from their lungs and remained asymptomatic. Infection in Muc1-/- mice was associated with an elevation in lung inflammation, with cellular recruitment especially of monocytes/macrophages. While MUC1-deficiency has been shown to increase phagocytosis of Pseudomonas aeruginosa, macrophages from Muc1-/- mice exhibited a reduced capacity to phagocytose S. pneumoniae indicating diverse and bacterial-specific effects. In conclusion, these findings indicate that MUC1 plays an important role in protection against severe pneumococcal disease, potentially mediated by facilitating macrophage phagocytosis.


Subject(s)
Mucin-1/immunology , Pneumococcal Infections/immunology , Streptococcus pneumoniae/physiology , Animals , Disease Models, Animal , Female , Humans , Lung/immunology , Lung/microbiology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mucin-1/genetics , Nasopharynx/immunology , Nasopharynx/microbiology , Phagocytosis , Pneumococcal Infections/genetics , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...