Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Blood ; 129(23): 3087-3099, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28325862

ABSTRACT

ß-Thalassemia is associated with several abnormalities of the innate immune system. Neutrophils in particular are defective, predisposing patients to life-threatening bacterial infections. The molecular and cellular mechanisms involved in impaired neutrophil function remain incompletely defined. We used the Hbbth3/+ ß-thalassemia mouse and hemoglobin E (HbE)/ß-thalassemia patients to investigate dysregulated neutrophil activity. Mature neutrophils from Hbbth3/+ mice displayed a significant reduction in chemotaxis, opsonophagocytosis, and production of reactive oxygen species, closely mimicking the defective immune functions observed in ß-thalassemia patients. In Hbbth3/+ mice, the expression of neutrophil CXCR2, CD11b, and reduced NAD phosphate oxidase components (p22phox, p67phox, and gp91phox) were significantly reduced. Morphological analysis of Hbbth3/+ neutrophils showed that a large percentage of mature phenotype neutrophils (Ly6GhiLy6Clow) appeared as band form cells, and a striking expansion of immature (Ly6GlowLy6Clow) hyposegmented neutrophils, consisting mainly of myelocytes and metamyelocytes, was noted. Intriguingly, expression of an essential mediator of neutrophil terminal differentiation, the ets transcription factor PU.1, was significantly decreased in Hbbth3/+ neutrophils. In addition, in vivo infection with Streptococcus pneumoniae failed to induce PU.1 expression or upregulate neutrophil effector functions in Hbbth3/+ mice. Similar changes to neutrophil morphology and PU.1 expression were observed in splenectomized and nonsplenectomized HbE/ß-thalassemia patients. This study provides a mechanistic insight into defective neutrophil maturation in ß-thalassemia patients, which contributes to deficiencies in neutrophil effector functions.


Subject(s)
Neutrophils/immunology , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , beta-Thalassemia/genetics , beta-Thalassemia/immunology , Adult , Animals , CD11b Antigen/metabolism , Case-Control Studies , Cell Differentiation/genetics , Cell Differentiation/immunology , Chemotaxis, Leukocyte , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Middle Aged , Neutrophil Activation , Neutrophils/metabolism , Neutrophils/pathology , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/immunology , Reactive Oxygen Species/metabolism , Receptors, Interleukin-8B/metabolism , Trans-Activators/deficiency , Trans-Activators/immunology , Young Adult , beta-Thalassemia/pathology
8.
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
9.
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
10.
Helicobacter ; 22(3)2017 Jun.
Article in English | MEDLINE | ID: mdl-28181350

ABSTRACT

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.


Subject(s)
Electron Transport Complex I/metabolism , Gastritis/physiopathology , Helicobacter Infections/physiopathology , Helicobacter pylori/pathogenicity , Mitochondria/enzymology , Animals , Bacterial Load , Disease Models, Animal , Electron Transport Complex I/deficiency , Female , Gastritis/pathology , Helicobacter Infections/pathology , Histocytochemistry , Mice, Inbred C57BL , Mice, Knockout
11.
Gut ; 65(7): 1087-99, 2016 07.
Article in English | MEDLINE | ID: mdl-26079943

ABSTRACT

OBJECTIVES: The mucin MUC1, best known for providing an epithelial barrier, is an important protective host factor in both humans and mice during Helicobacter pylori pathogenesis. This study aimed to identify the long-term consequences of MUC1 deficiency on H. pylori pathogenesis and the mechanism by which MUC1 protects against H. pylori gastritis. DESIGN: Wildtype and Muc1(-/-) mice were infected for up to 9 months, and the gastric pathology, immunological response and epigenetic changes assessed. The effects of MUC1 on the inflammasome, a potent inflammatory pathway, were examined in macrophages and H. pylori-infected mice deficient in both MUC1 and inflammasome components. RESULTS: Muc1(-/-) mice began to die 6 months after challenge, indicating Muc1 deficiency made H. pylori a lethal infection. Surprisingly, chimaeric mouse infections revealed MUC1 expression by haematopoietic-derived immune cells limits H. pylori-induced gastritis. Gastritis in infected Muc1(-/-) mice was associated with elevated interleukin (IL)-1ß and epigenetic changes in their gastric mucosa similar to those in transgenic mice overexpressing gastric IL-1ß, implicating MUC1 regulation of an inflammasome. In support of this, infected Muc1(-/-)Casp1(-/-) mice did not develop severe gastritis. Further, MUC1 regulated Nlrp3 expression via an nuclear factor (NF)-κB-dependent pathway and reduced NF-κB pathway activation via inhibition of IRAK4 phosphorylation. The importance of this regulation was proven using Muc1(-/-)Nlrp3(-/-) mice, which did not develop severe gastritis. CONCLUSIONS: MUC1 is an important, previously unidentified negative regulator of the NLRP3 inflammasome. H. pylori activation of the NLRP3 inflammasome is normally tightly regulated by MUC1, and loss of this critical regulation results in the development of severe pathology.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/metabolism , Helicobacter pylori/pathogenicity , Inflammasomes/metabolism , Mucin-1/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Caspase 1/genetics , DNA Methylation , Female , Gastric Mucosa/immunology , Gastric Mucosa/metabolism , Gastritis/pathology , Gene Expression , Helicobacter Infections/complications , Helicobacter Infections/immunology , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Mucin-1/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Time Factors , Trefoil Factor-2/genetics
12.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G514-20, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27469367

ABSTRACT

The bacterial pathogen Helicobacter pylori is the etiological agent of a range of gastrointestinal pathologies including peptic ulcer disease and the major killer, gastric adenocarcinoma. Infection with this bacterium induces a chronic inflammatory response in the gastric mucosa (gastritis). It is this gastritis that, over decades, eventually drives the development of H. pylori-associated disease in some individuals. The majority of studies investigating H. pylori pathogenesis have focused on factors that promote disease development in infected individuals. However, an estimated 85% of those infected with H. pylori remain completely asymptomatic, despite the presence of pathogenic bacteria that drive a chronic gastritis that lasts many decades. This indicates the presence of highly effective regulatory processes in the host that, in most cases, keeps a check on inflammation and protect against disease. In this minireview we discuss such known host factors and how they prevent the development of H. pylori-associated pathologies.


Subject(s)
Gastritis/microbiology , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Helicobacter pylori , Peptic Ulcer/microbiology , Stomach Neoplasms/microbiology , Humans
13.
Helicobacter ; 20(3): 217-22, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25683672

ABSTRACT

BACKGROUND: Helicobacter pylori pathogenesis results from the inflammation induced by chronic infection. CBA mice are nonresponsive to gastric Helicobacter infection, providing a useful model for examining host regulation of Helicobacter-induced gastritis. We examined whether gastric Helicobacter nonresponsiveness impacts upon vaccine efficacy and whether immune-mediated protection could occur in the absence of inflammation. METHODS: Mice were vaccinated prior to challenge with Helicobacter felis or H. pylori. Gastritis and H. felis colonization was evaluated histologically. H. pylori colonization was quantified by colony-forming assay. RESULTS: Immunizations protected CBA mice against challenge with either H. felis or H. pylori. Protection against H. felis was marked by a loss of nonresponsiveness and development of an atrophic gastritis with mucus metaplasia. However, vaccine-induced protection against H. pylori was only associated with cell infiltration into the gastric mucosa. CONCLUSIONS: Nonresponsiveness to gastric Helicobacter infection did not interfere with vaccination-induced protection. Vaccine-induced protective immunity against H. pylori was linked with the induction of cellular infiltration, but importantly not atrophic gastritis.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Vaccines/immunology , Gastric Mucosa/immunology , Gastritis, Atrophic/immunology , Helicobacter Infections/immunology , Helicobacter/immunology , Immunization , Stomach/immunology , Animals , Disease Models, Animal , Female , Gastritis, Atrophic/prevention & control , Helicobacter Infections/prevention & control , Helicobacter felis/immunology , Helicobacter pylori/immunology , Humans , Inflammation , Metaplasia/pathology , Mice , Mice, Inbred CBA
14.
Linacre Q ; 82(4): 351-63, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26997676

ABSTRACT

In recent years, national and international medical and mental-health associations typically have emphasized the potential harmfulness of professional care for unwanted same-sex attraction (SSA or homosexuality) and behavior. State legislatures in the US and legislative bodies in other countries either have passed or are considering passing laws which would penalize professionals who provide professional care for unwanted SSA-to minors and/or adults-including the loss of the license to practice. This paper was written as a response to the present situation in the UK. The paper reviews the universal ethics of all medical and mental-health professionals to avoid harm and do good (non-maleficence/non-malfeasance and beneficence); discusses the documented potential for harm when using every mental-health treatment for every presenting problem; clarifies steps taken by the Alliance for Therapeutic Choice and Scientific Integrity (Alliance), its clinical and research divisions, the National Association for Research and Therapy of Homosexuality Institute (NARTH Institute) and its international division, the International Federation for Therapeutic Choice (IFTC), to promote ethical professional care for unwanted SSA; clarifies the injustice and presumed ideological biases of the medical and mental-health associations' warning about the potential for harm for psychotherapy only for unwanted SSA and not all psychotherapy approaches; and documents that the research purporting to show this harmfulness, in the research authors own words, does not do so. Recommendations to promote scientific integrity in the conduct and reporting of relevant research are offered. Lay Summary: There has been a lot of controversy about the potential harmfulness of professional care for unwanted same-sex attraction and behavior (SSA or homosexuality). This paper reviews the ethics of all medical and mental health professionals to avoid harm and do good; discusses the known potential for harm when using any mental health treatment for any problem; clarifies steps taken to promote ethical professional care for unwanted SSA; notes the injustice and possible biases of those who warn about the potential for harm of psychotherapy for unwanted SSA; and documents that the research said to show this harmfulness, in the research authors own words, does not do so.

15.
Helicobacter ; 19(1): 48-54, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24165046

ABSTRACT

BACKGROUND: The development of an effective vaccine against Helicobacter pylori is impeded by the inability to reliably produce sterilizing immunity and our lack of knowledge regarding mechanisms of protective immunity against this pathogen. It has previously been described that salivary glands are essential for vaccine-mediated protection against H. pylori, but the mechanism responsible for this effect has not been identified. In this study we tested the hypothesis that vaccines reduce H. pylori colonization by inducing an immune-mediated change in salivary gland mucin secretion. MATERIALS AND METHODS: Sublingual and submandibular salivary glands were removed from untreated mice, from mice infected with H. pylori and from mice vaccinated against H. pylori then challenged with live bacteria. Cytokine levels in these salivary glands were quantified by ELISA, and salivary mucins were quantified by real-time PCR. Salivary antibody responses were determined by Western blot. RESULTS: Vaccine-mediated protection against H. pylori did not produce any evidence of a positive increase in either salivary cytokine or mucin levels. In fact, many cytokines were significantly reduced in the vaccinated/challenged mice, including IL-17A, IL-10, IL-1ß, as well as the mucin Muc10. These decreases were associated with an increase in total protein content within the salivary glands of vaccinated mice which appeared to be the result of increased IgA production. While this study showed that vaccination increased salivary IgA levels, previous studies have demonstrated that antibodies do not play a critical role in protection against H. pylori that is induced by current vaccine formulations and regimes. CONCLUSIONS: The effector mechanism of protective immunity induced by vaccination of mice did not involve immune changes within the salivary glands, nor increased production of salivary mucins.


Subject(s)
Bacterial Vaccines/immunology , Cytokines/immunology , Helicobacter pylori/immunology , Mucins/immunology , Saliva/immunology , Animals , Antibodies, Bacterial/analysis , Bacterial Vaccines/administration & dosage , Blotting, Western , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay , Female , Mice , Mice, Inbred C57BL , Mucins/analysis , Real-Time Polymerase Chain Reaction , Saliva/chemistry , Salivary Glands/chemistry , Salivary Glands/immunology
16.
Am J Physiol Gastrointest Liver Physiol ; 302(6): G579-87, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22194421

ABSTRACT

Helicobacter pylori is a microaerophilic, gram-negative pathogen of the human stomach. Despite the chronic active gastritis that develops following colonization, H. pylori is able to persist unharmed in the stomach for decades. Much of the damage caused by gastric inflammation results from the accumulation of reactive oxygen/nitrogen species within the stomach environment, which can induce oxidative damage in a wide range of biological molecules. Without appropriate defenses, this oxidative damage would be able to rapidly kill nearby H. pylori, but the organism employs a range of measures, including antioxidant enzymes, biological repair systems, and inhibitors of oxidant generation, to counter the attack. Despite the variety of measures employed to defend against oxidative injury, these processes are intimately interdependent, and any deficiency within the antioxidant system is generally sufficient to cause substantial impairment of H. pylori viability and persistence. This review provides an overview of the development of oxidative stress during H. pylori gastritis and examines the methods the organism uses to survive the resultant damage.


Subject(s)
Helicobacter pylori/physiology , Oxidative Stress/physiology , Antioxidants/metabolism , Gastric Mucosa/metabolism , Gastric Mucosa/microbiology , Gastritis/metabolism , Gastritis/microbiology , Helicobacter Infections/metabolism , Helicobacter Infections/microbiology , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
17.
Gut ; 60(12): 1661-70, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21636645

ABSTRACT

BACKGROUND AND AIMS: The MUC13 transmembrane mucin is highly and constitutively expressed in the small and large intestine. Although MUC13 polymorphisms have been associated with human inflammatory bowel diseases and susceptibility to Escherichia coli infection in pigs, the biological functions of MUC13 are unknown. This study aimed to explore whether MUC13 modulates intestinal inflammation. METHODS: Muc13(-/-) mice were generated, phenotyped and challenged with the colitis-inducing agent, dextran sodium sulphate (DSS). Colitis was assessed by clinical symptoms and intestinal histopathology. Intestinal epithelial cell apoptosis and proliferation, macrophage infiltration and cytokine production were also quantified. Apoptosis of human LS513 intestinal epithelial cells in response to apoptotic agents, including DSS, was also measured, following knockdown of MUC13 with siRNA. RESULTS: Muc13(-/-) mice were viable, fertile and developed normally, with no spontaneous intestinal pathology except mild focal neutrophilic inflammation in the small and large intestines of old mice. In response to DSS challenge, Muc13(-/-) mice developed more severe acute colitis, as reflected by increased weight loss, rectal bleeding, diarrhoea and histological colitis scores compared with wild-type mice. Increased numbers of F4/80(+) macrophages in inflamed mucosa of Muc13(-/-) mice were accompanied by increased expression of intestinal IL-1ß and TNFα mRNA. Muc13(-/-) mice had significantly increased intestinal epithelial cell apoptosis within 3 days of DSS exposure. LS513 cells were more susceptible to DSS, actinomycin-D, ultraviolet irradiation and TRAIL-induced apoptosis when MUC13 was knocked down by siRNA. CONCLUSIONS: These novel findings indicate a protective role for Muc13 in the colonic epithelium by inhibiting toxin-induced apoptosis and have important implications for intestinal infections, inflammatory diseases and the development of intestinal cancer.


Subject(s)
Antigens, Surface/physiology , Apoptosis/physiology , Colitis/physiopathology , Epidermal Growth Factor/physiology , Intestinal Mucosa/physiopathology , Mucins/physiology , Animals , Antigens, Surface/metabolism , Cell Line , Cell Proliferation , Colitis/metabolism , Colitis/pathology , Cytokines/physiology , Dextran Sulfate/pharmacology , Epidermal Growth Factor/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/physiopathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Macrophages, Peritoneal/physiology , Mice , Mice, Knockout , Mucins/metabolism , Real-Time Polymerase Chain Reaction
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL