Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Am Assoc Lab Anim Sci ; 61(4): 344-352, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35688608

ABSTRACT

Bats are known natural reservoirs of several highly pathogenic zoonotic viruses, including Hendra virus, Nipah virus, rabies virus, SARS-like coronaviruses, and suspected ancestral reservoirs of SARS-CoV-2 responsible for the ongoing COVID-19 pandemic. The capacity to survive infections of highly pathogenic agents without severe disease, together with many other unique features, makes bats an ideal animal model for studying the regulation of infection, cancer, and longevity, which is likely to translate into human health outcomes. A key factor that limits bat research is lack of breeding bat colonies. To address this need, a captive bat colony was established in Singapore from 19 wild-caught local cave nectar bats. The bats were screened for specific pathogens before the start of captive breeding. Custom-made cages and an optimized diet inclusive of Wombaroo dietary formula, liquid diet, and supplement of fruits enabled the bats to breed prolifically in our facility. Cages are washed daily and disinfected once every fortnight. Bats are observed daily to detect any sick bat or abnormal behavior. In addition, bats undergo a thorough health check once every 3 to 4 mo to check on their overall wellbeing, perform sampling, and document any potential pregnancy. The current colony houses over 80 bats that are successfully breeding, providing a valuable resource for research in Singapore and overseas.


Subject(s)
COVID-19 , Chiroptera , Animals , Breeding , Disease Reservoirs , Humans , Pandemics , Phylogeny , Plant Nectar , SARS-CoV-2 , Singapore
2.
Cell Stress Chaperones ; 24(4): 835-849, 2019 07.
Article in English | MEDLINE | ID: mdl-31230214

ABSTRACT

Bats, unique among mammals with powered flight, have many species with the longest size-proportionate lifespan of all mammals. Evolutionary adaptations would have been required to survive the elevated body temperatures during flight. Heat shock protein (HSP), highly conserved master regulators of cell stress, expression was examined across tissues and various cell lines in bats. Basal expression level of major HSPs (HSP70 and HSP90) is significantly higher in two different bat species compared to other mammals. This HSP expression could be a bat-unique, key factor to modulate cellular stress and death. Consequently, bat cells survive prolonged heat treatment, along with other stress stimuli, in a HSP-dependent manner, whereas other mammalian cells succumbed. This suggests HSP expression in bats could be an important adaption to intrinsic metabolic stresses like flight and therefore an important model to study stress resilience and longevity in general.


Subject(s)
Chiroptera/metabolism , Flight, Animal/physiology , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heat-Shock Response/physiology , Longevity/physiology , Oxidative Stress/physiology , Adaptation, Physiological/physiology , Animals , Cell Line , Humans
3.
Nat Commun ; 10(1): 2820, 2019 06 27.
Article in English | MEDLINE | ID: mdl-31249297

ABSTRACT

Bats are unusual mammals, with the ability to fly, and long lifespans. In addition, bats have a low incidence of cancer, but the mechanisms underlying this phenomenon remain elusive. Here we discovered that bat cells are more resistant than human and mouse cells to DNA damage induced by genotoxic drugs. We found that bat cells accumulate less chemical than human and mouse cells, and efficient drug efflux mediated by the ABC transporter ABCB1 underlies this improved response to genotoxic reagents. Inhibition of ABCB1 triggers an accumulation of doxorubicin, DNA damage, and cell death. ABCB1 is expressed at higher levels in several cell lines and tissues derived from bats compared to humans. Furthermore, increased drug efflux and high expression of ABCB1 are conserved across multiple bat species. Our findings suggest that enhanced efflux protects bat cells from DNA damage induced by genotoxic compounds, which may contribute to their low cancer incidence.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Chiroptera/genetics , Chiroptera/metabolism , DNA Damage/drug effects , Mutagens/toxicity , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Cell Death/drug effects , Cell Line , Doxorubicin/toxicity , Humans , Mice
4.
Gigascience ; 7(10)2018 10 01.
Article in English | MEDLINE | ID: mdl-30247613

ABSTRACT

Background: In the past two decades, bats have emerged as an important model system to study host-pathogen interactions. More recently, it has been shown that bats may also serve as a new and excellent model to study aging, inflammation, and cancer, among other important biological processes. The cave nectar bat or lesser dawn bat (Eonycteris spelaea) is known to be a reservoir for several viruses and intracellular bacteria. It is widely distributed throughout the tropics and subtropics from India to Southeast Asia and pollinates several plant species, including the culturally and economically important durian in the region. Here, we report the whole-genome and transcriptome sequencing, followed by subsequent de novo assembly, of the E. spelaea genome solely using the Pacific Biosciences (PacBio) long-read sequencing platform. Findings: The newly assembled E. spelaea genome is 1.97 Gb in length and consists of 4,470 sequences with a contig N50 of 8.0 Mb. Identified repeat elements covered 34.65% of the genome, and 20,640 unique protein-coding genes with 39,526 transcripts were annotated. Conclusions: We demonstrated that the PacBio long-read sequencing platform alone is sufficient to generate a comprehensive de novo assembled genome and transcriptome of an important bat species. These results will provide useful insights and act as a resource to expand our understanding of bat evolution, ecology, physiology, immunology, viral infection, and transmission dynamics.


Subject(s)
Chiroptera/genetics , Genome , Genomics , Transcriptome , Alternative Splicing , Animals , Chiroptera/classification , Computational Biology/methods , Evolution, Molecular , Female , Genomics/methods , High-Throughput Nucleotide Sequencing , Molecular Sequence Annotation , Phylogeny
5.
Sci Rep ; 8(1): 4726, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549333

ABSTRACT

Bats are an important animal model with long lifespans, low incidences of tumorigenesis and an ability to asymptomatically harbour pathogens. Currently, in vivo studies of bats are hampered due to their low reproduction rates. To overcome this, we transplanted bat cells from bone marrow (BM) and spleen into an immunodeficient mouse strain NOD-scid IL-2R-/- (NSG), and have successfully established stable, long-term reconstitution of bat immune cells in mice (bat-mice). Immune functionality of our bat-mouse model was demonstrated through generation of antigen-specific antibody response by bat cells following immunization. Post-engraftment of total bat BM cells and splenocytes, bat immune cells survived, expanded and repopulated the mouse without any observable clinical abnormalities. Utilizing bat's remarkable immunological functions, this novel model has a potential to be transformed into a powerful platform for basic and translational research.


Subject(s)
Bone Marrow Transplantation/methods , Graft Rejection/immunology , Graft Survival/immunology , Lymphocytes/immunology , Severe Combined Immunodeficiency/therapy , Transplantation Chimera/immunology , Animals , Chiroptera , Graft Rejection/prevention & control , Mice , Mice, Inbred NOD , Mice, SCID , Severe Combined Immunodeficiency/immunology
6.
Sci Rep ; 6: 38597, 2016 12 09.
Article in English | MEDLINE | ID: mdl-27934903

ABSTRACT

Bats carry and shed many emerging infectious disease agents including Ebola virus and SARS-like Coronaviruses, yet they rarely display clinical symptoms of infection. Bat epithelial or fibroblast cell lines were previously established to study the bat immune response against viral infection. However, the lack of professional immune cells such as dendritic cells (DC) and macrophages has greatly limited the significance of current investigations. Using Pteropus alecto (P. alecto) GM-CSF plus IL4, FLT3L and CSF-1, we successfully generated bat bone marrow-derived DC and macrophages. Cells with the phenotype, morphology and functional features of monocyte-derived DC, bona fide DC or macrophages were obtained in GM-CSF/IL4, FLT3L or CSF-1 cultures, respectively. The successful generation of the first bat bone marrow-derived immune cells paves the way to unlocking the immune mechanisms that confer host resilience to pathogens in bats.


Subject(s)
Chiroptera/immunology , Immunity , Animals , Biomarkers , Chiroptera/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunity, Innate , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Phagocytosis , Phenotype , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
J Clin Invest ; 126(4): 1383-400, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26974160

ABSTRACT

Chronic mucosal inflammation is associated with a greater risk of gastric cancer (GC) and, therefore, requires tight control by suppressive counter mechanisms. Gastrokine-2 (GKN2) belongs to a family of secreted proteins expressed within normal gastric mucosal cells. GKN2 expression is frequently lost during GC progression, suggesting an inhibitory role; however, a causal link remains unsubstantiated. Here, we developed Gkn2 knockout and transgenic overexpressing mice to investigate the functional impact of GKN2 loss in GC pathogenesis. In mouse models of GC, decreased GKN2 expression correlated with gastric pathology that paralleled human GC progression. At baseline, Gkn2 knockout mice exhibited defective gastric epithelial differentiation but not malignant progression. Conversely, Gkn2 knockout in the IL-11/STAT3-dependent gp130F/F GC model caused tumorigenesis of the proximal stomach. Additionally, gastric immunopathology was accelerated in Helicobacter pylori-infected Gkn2 knockout mice and was associated with augmented T helper cell type 1 (Th1) but not Th17 immunity. Heightened Th1 responses in Gkn2 knockout mice were linked to deregulated mucosal innate immunity and impaired myeloid-derived suppressor cell activation. Finally, transgenic overexpression of human gastrokines (GKNs) attenuated gastric tumor growth in gp130F/F mice. Together, these results reveal an antiinflammatory role for GKN2, provide in vivo evidence that links GKN2 loss to GC pathogenesis, and suggest GKN restoration as a strategy to restrain GC progression.


Subject(s)
Carrier Proteins/metabolism , Gastric Mucosa/metabolism , Neoplasm Proteins/metabolism , Precancerous Conditions/metabolism , Stomach Neoplasms/metabolism , Animals , Carrier Proteins/genetics , Gastric Mucosa/pathology , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Immunity, Innate , Immunity, Mucosal , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/metabolism , Th17 Cells/pathology
8.
Vaccine ; 34(14): 1704-11, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26876441

ABSTRACT

BACKGROUND: Neisseria meningitidis are common colonizers of the human nasopharynx. In some circumstances, N. meningitidis becomes an opportunistic pathogen that invades tissues and causes meningitis. While a vaccine against a number of serogroups has been in effective use for many years, a vaccine against N. meningitidis group B has not yet been universally adopted. Bacterial heat shock protein complex (HSPC) vaccines comprise bacterial HSPs, purified with their chaperoned protein cargo. HSPC vaccines use the intrinsic adjuvant activity of their HSP, thought to act via Toll-like receptors (TLR), to induce an immune response against their cargo antigens. This study evaluated HSPC vaccines from N. meningitidis and the closely related commensal N. lactamica. RESULTS: The protein composition of N. lactamica and N. meningitidis HSPCs were similar. Using human HEK293 cells we found that both HSPCs can induce an innate immune response via activation of TLR2. However, stimulation of TLR2 or TLR4 deficient murine splenocytes revealed that HSPCs can activate an innate immune response via multiple receptors. Vaccination of wildtype mice with the Neisseria HSPC induced a strong antibody response and a Th1-restricted T helper response. However, vaccination of mice deficient in the major TLR adaptor protein, MyD88, revealed that while the Th1 response to Neisseria HSPC requires MyD88, these vaccines unexpectedly induced an antigen-specific antibody response via a MyD88-independent mechanism. CONCLUSIONS: N. lactamica and N. meningitidis HSPC vaccines both have potential utility for immunising against neisserial meningitis without the requirement for an exogenous adjuvant. The mode of action of these vaccines is highly complex, with HSPCs inducing immune responses via both MyD88-dependent and -independent mechanisms. In particular, these HSPC vaccines induced an antibody response without detectable T cell help.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Vaccines/immunology , Heat-Shock Proteins/immunology , Immunity, Innate , Neisseria meningitidis , Animals , Bacterial Proteins/immunology , Cytokines/immunology , HEK293 Cells , Humans , Immunity, Humoral , Immunoglobulin G/blood , Meningitis, Meningococcal/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/immunology , Neisseria lactamica , Proteome , Spleen/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
9.
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
10.
Vaccine ; 32(20): 2350-8, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24625340

ABSTRACT

BACKGROUND: The development of a vaccine against the human gastric pathogen Helicobacter pylori, the main causative agent of gastric adenocarcinoma, has been hampered by a number of issues, including the lack of a mucosal adjuvant for use in humans. Heat shock proteins (Hsp), highly conserved molecules expressed by both bacteria and mammalian species, possess a range of functions, including acting as chaperones for cellular proteins and the ability to activate innate immune receptors. Hsp complex (HspC) vaccines, containing Hsp derived from pathogenic bacteria, are immunostimulatory without addition of an exogenous adjuvant and can induce immunity against their chaperoned proteins. In this study we explored in mice the potential utility of a H. pylori HspC vaccine. RESULTS: Vaccination with H. pylori HspC, by either the subcutaneous or respiratory mucosal route, induced a strong antibody response, elevated gastric cytokine levels and significant protection against subsequent live challenge with this pathogen. The level of protection induced by non-adjuvanted HspC vaccine was equivalent to that which resulted from vaccination with adjuvanted vaccines. While protection induced by immunisation with adjuvanted vaccines was associated with the development of a moderate to severe atrophic gastritis, that induced by H. pylori HspC only resulted in a mild inflammatory response, despite an increase in pro-inflammatory gastric cytokines. This reduced gastritis correlated with an increase in IL-10 and IL-13 levels in the gastric tissues of HspC vaccinated, H. pylori challenged mice. CONCLUSIONS: H. pylori HspC vaccines have the potential to overcome some of the issues preventing the development of a human vaccine against this pathogen: HspC induced protective immunity against H. pylori without addition of an adjuvant and without the induction of a severe inflammatory response. However, complete protection was not obtained so further optimisation of this technology is needed if a human vaccine is to become a reality.


Subject(s)
Adjuvants, Immunologic , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Heat-Shock Proteins/immunology , Helicobacter Infections/prevention & control , Animals , Antibodies, Bacterial/blood , Cytokines/immunology , Female , Gastric Mucosa/immunology , Helicobacter pylori , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Injections, Subcutaneous , Mice , Mice, Inbred C57BL
11.
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
12.
Expert Rev Vaccines ; 12(4): 433-41, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23560923

ABSTRACT

Helicobacter pylori is a major human pathogen that colonizes the stomach and is the lead etiological agent for several pathologies. An effective vaccine against these bacteria would be invaluable for protecting against gastric adenocarcinoma. However, the development of such a vaccine has stalled and the field has progressed little in the last decade. In this review, the authors provide an opinion on key problems that are preventing the development of a H. pylori vaccine. Primarily, this involves the inability to produce a completely protective immune response. The knock-on effects of this include a loss of industry investment. Overcoming these problems will likely involve defeating the immune-evasion defenses of H. pylori, in particular the mechanism(s) by which it evades antibody-mediated attack.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Vaccines/isolation & purification , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Drug Discovery/trends , Humans
13.
Anticancer Res ; 32(9): 3715-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22993310

ABSTRACT

BACKGROUND: PAR-1 has been involved in inflammation of the gastrointestinal tract, tumour cell growth and invasion of gastric carcinoma cells. Thus, we aimed at determining, for the first time, the association between PAR-1 - 506 ins/del and -IVSn-14 A/T and risk of Helicobacter pylori-related gastric cancer (GC) in an ethnic Chinese population. MATERIALS AND METHODS: A case-control study comprising of 225 ethnic Chinese individuals (77 non-cardia GC cases and 148 controls with functional dyspepsia) was conducted. PAR-1 IVSn-14 A/T and 506 ins/del were genotyped by means of real-time PCR and MALDI-TOF mass spectrometry, respectively. RESULTS: H. pylori infection, male gender and the PAR-1 IVSn-14 TT genotype increased GC risk (OR:3.15, 95% CI:1.54-6.45, OR:2.44, 95% CI:1.35-4.42 and OR:2.58, 95% CI:1.09-6.13, respectively). PAR-1 -506 ins/del did not provide significant results. CONCLUSION: PAR-1 IVSn-14 T allele is a risk factor for H. pylori-related GC in ethnic Chinese subjects. PAR-1 -506 ins/del polymorphism is not involved in gastric carcinogenesis.


Subject(s)
Helicobacter Infections/genetics , Helicobacter pylori/isolation & purification , Receptor, PAR-1/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology , Aged , Asian People , Case-Control Studies , Female , Helicobacter Infections/complications , Helicobacter Infections/ethnology , Humans , Male , Middle Aged , Polymorphism, Genetic , Stomach Neoplasms/ethnology
14.
Gut Microbes ; 1(1): 42-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-21327115

ABSTRACT

Recently, we demonstrated that oral delivery of whole killed bacteria, when agglutinated by an M-cell targeting lectin, resulted in an enhanced systemic and mucosal antibody response, as well as a protective immunity, against the gut pathogens Helicobacter pylori and Campylobacter jejuni. Importantly, this protection was achieved without the addition of exogenous adjuvant. Here, in this addendum, we extend this initial study by reporting on the vaginal antibody response induced by these vaccinations. These data show that the targeting of M-cells within the gastrointestinal tract also induces the secretion of antigen-specific antibodies (IgG and IgA) at a distal mucosal site, namely the vaginal mucosa. This observation raises the possibility that oral delivery of a whole, killed bacteria vaccine that target intestinal M-cells could potentially provide a strategy for inducing protective immunity against pathogenic bacteria that infect mucosal sites outside the gastrointestinal tract.

15.
Gastroenterology ; 138(2): 573-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19706295

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori infection results in a diversity of pathologies, from asymptomatic gastritis to adenocarcinoma. The reason for these diverse outcomes is multifactorial and includes host factors that regulate severity of Helicobacter-induced gastritis. Protease-activated receptors (PAR) are environmental sensors that can detect tissue damage and pathogens. Whereas PAR-2 has proinflammatory activity and PAR-1 can protect the gastric mucosa against chemical damage, neither has previously been examined for their potential roles in regulating Helicobacter pathogenesis. METHODS: PAR-1(-/-), PAR-2(-/-), and wild-type mice were infected with H pylori for up to 2 months then colonization levels determined by colony-forming assay, gastritis by histology, and serum antibody levels by enzyme-linked immunosorbent assay. Responsiveness of primary epithelial cells to PAR-1 activation was assessed by calcium mobilization assay. Primary epithelial cells, macrophages, and dendritic cells were cocultured with H pylori and nuclear factor (NF)-kappaB, and cytokine secretion was determined by enzyme-linked immunosorbent assay. RESULTS: Two months postinfection, H pylori levels were significantly reduced in PAR-1(-/-) and increased in PAR-2(-/-) mice. This effect on colonization was inversely correlated with inflammation severity. Infection of PAR-1(-/-) mice induced an increased serum antibody response. Primary epithelial cells were activated by a PAR-1-activating peptide. H pylori stimulation of primary epithelial cells, but not macrophages or dendritic cells, from PAR-1(-/-) mice induced increased levels of NF-kappaB and the proinflammatory cytokine macrophage-inflammatory protein (MIP)-2. PAR-1 also down-regulated MIP-2 secretion in response to cag pathogenicity island activity. CONCLUSIONS: PAR-1 protects the host against severe Helicobacter-induced gastritis. This may be mediated by suppressing the production of proinflammatory cytokines such as MIP-2.


Subject(s)
Gastritis/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori/physiology , Immunity, Humoral/physiology , Inflammation/physiopathology , Receptor, PAR-1/metabolism , Animals , Cells, Cultured , Chemokine CXCL2/metabolism , Disease Models, Animal , Down-Regulation/physiology , Epithelium/metabolism , Epithelium/pathology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/physiopathology , Helicobacter Infections/physiopathology , Helicobacter pylori/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Receptor, PAR-2/metabolism
16.
FEMS Immunol Med Microbiol ; 57(1): 25-31, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19624738

ABSTRACT

Adherence to the mucosal surface, essential for successful colonization of the gastric mucosa by the human pathogen Helicobacter pylori, is predominantly mediated by lectin-like molecules on the Helicobacter binding carbohydrates expressed by host epithelial cells. While clinical isolates of H. pylori do not normally infect mice, some strains have been adapted to colonize this host. We hypothesized that adaptation of H. pylori for colonization of mice may involve alterations in either bacterial surface glycan expression or their glycan-binding properties. Using a panel of lectins, we compared glycan expression on lipopolysaccharides from five mouse-colonizing strains with that of four fresh clinical isolates, but found no association with their ability to infect mice. To compare their adherence to carbohydrates expressed on host epithelial cells, we examined the ability of monosaccharides to block the attachment of mouse-adapted and clinical isolates of H. pylori to human epithelial cell lines. Mannose, N-acetylgalactosamine, N-acetylglucosamine, fucose and sialic acid were all significantly more potent at inhibiting the attachment of mouse-adapted strains to these cell lines than clinical isolates. This might suggest that colonization of the murine mucosa by H. pylori is less dependent on adhesion to host glycans than is the case during human infection.


Subject(s)
Bacterial Adhesion , Helicobacter Infections/microbiology , Helicobacter pylori/physiology , Polysaccharides/biosynthesis , Animals , Cell Line , Colony Count, Microbial , Epithelial Cells/microbiology , Female , Helicobacter pylori/isolation & purification , Helicobacter pylori/metabolism , Humans , Lectins/metabolism , Mice , Mice, Inbred C57BL , Protein Binding , Stomach/microbiology
17.
Infect Immun ; 77(7): 2962-70, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19380476

ABSTRACT

As the majority of human pathogens infect via a mucosal surface, delivery of killed vaccines by mucosal routes could potentially improve protection against many such organisms. Our ability to develop effective killed mucosal vaccines is inhibited by a lack of adjuvants that are safe and effective in humans. The Ulex europaeus agglutinin I (UEA-I) lectin specifically binds M cells lining the murine gastrointestinal tract. We explored the potential for M-cell-targeted vaccination of whole, killed Helicobacter pylori, the main causative agent of peptic ulcer disease and gastric cancer, and Campylobacter jejuni, the most common cause of diarrhea. Oral delivery of UEA-I-agglutinated H. pylori or C. jejuni induced a significant increase in both serum and intestinal antibody levels. This elevated response (i) required the use of whole bacteria, as it did not occur with lysate; (ii) was not mediated by formation of particulate clumps, as agglutination with a lectin with a different glycan specificity had no effect; and (iii) was not due to lectin-mediated, nonspecific immunostimulatory activity, as UEA-I codelivery with nonagglutinated bacteria did not enhance the response. Vaccination with UEA-I-agglutinated, killed whole H. pylori induced a protective response against subsequent live challenge that was as effective as that induced by cholera toxin adjuvant. Moreover, vaccination against C. jejuni by this approach resulted in complete protection against challenge in almost all animals. We believe that this is the first demonstration that targeting of whole killed bacteria to mucosal M cells can induce protective immunity without the addition of an immunostimulatory adjuvant.


Subject(s)
Adjuvants, Immunologic/pharmacology , Bacterial Vaccines/immunology , Campylobacter Infections/prevention & control , Helicobacter Infections/prevention & control , Plant Lectins/pharmacology , Adjuvants, Immunologic/administration & dosage , Administration, Oral , Animals , Antibodies, Bacterial/analysis , Antibodies, Bacterial/blood , Bacterial Vaccines/administration & dosage , Campylobacter Infections/pathology , Colony Count, Microbial , Female , Helicobacter Infections/pathology , Immunoglobulin A/analysis , Immunoglobulin A/blood , Immunoglobulin G/analysis , Immunoglobulin G/blood , Intestinal Mucosa/chemistry , Intestinal Mucosa/immunology , Mice , Mice, Inbred BALB C , Plant Lectins/administration & dosage , Severity of Illness Index , Stomach/microbiology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
18.
Helicobacter ; 13(6): 489-93, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19166413

ABSTRACT

BACKGROUND: The mucin Muc1 is constitutively expressed by the gastric mucosa and is likely the first point of direct contact between the host stomach and the adherent pathogens. The expression of Muc1 has been shown to limit colonization of mice by Helicobacter pylori, known to adhere to the gastric epithelium, as well as associated pathology. However, the potential role of this mucin against nonadherent Helicobacter has not been previously studied. We therefore examined the importance of Muc1 on the pathogenesis of Helicobacter felis, believed not to adhere to the murine mucosa. METHODS AND RESULTS: Using primary cell cultures, we found that H. felis can bind gastric epithelial cells in vitro, and adherence to epithelial cells deficient in Muc1 was increased compared to controls that expressed the mucin. However, following infection of deficient mice, we found that Muc1 did not impact on H. felis colonization or pathogenesis in vivo, in contrast to previous observations with H. pylori. CONCLUSIONS: This demonstrates a variable effect of Muc1 on protection against closely related adherent and nonadherent Helicobacter species, and supports a key role for Muc1 in limiting attachment of adherent bacteria to the gastric mucosal surface.


Subject(s)
Bacterial Adhesion/immunology , Epithelial Cells/microbiology , Helicobacter Infections/immunology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Mucin-1/immunology , Animals , Cells, Cultured , Helicobacter Infections/microbiology , Mice , Mice, Knockout
19.
Gastroenterology ; 133(4): 1210-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17919495

ABSTRACT

BACKGROUND & AIMS: The MUC1 mucin is expressed on the cell surface of epithelial cells lining the gastric mucosa. Epidemiologic studies suggest that functional allelic variations in the MUC1 gene may play a role in human susceptibility to Helicobacter pylori-associated pathologies, including gastric adenocarcinoma. We have evaluated the impact of Muc1 expression on the colonization and pathogenesis of gastric Helicobacter infections. METHODS: Wild-type and Muc1-deficient mice were infected with H pylori and colonization and gastritis levels determined. Primary gastric cells were used to examine the impact of Muc1 expression on bacterial adherence. RESULTS: Mice lacking Muc1 were colonized by 5-fold more H pylori within 1 day of infection, and this difference was maintained for at least 2 months postinfection. Mice heterozygous for the null Muc1 allele developed intermediate bacterial colonization. Although wild-type mice developed only a mild gastritis when infected for 2 months with H pylori, Muc1(-/-) mice developed an atrophic gastritis marked by loss of parietal cells. We demonstrate H pylori adhesion to purified MUC1 and significantly increased adhesion to cultured murine Muc1 null gastric epithelial cells, suggesting that Muc1 acts as a decoy limiting binding to the cell surface. CONCLUSIONS: Muc1 provides a protective barrier, which limits both acute and chronic colonization by H pylori, as well as playing a major role in limiting the inflammation induced by Helicobacter infection. We propose that Muc1 restricts access of H pylori to the epithelial surface, hence reducing exposure of the host to proinflammatory bacterial products.


Subject(s)
Gastric Mucosa/microbiology , Gastritis/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/pathogenicity , Mucin-1/metabolism , Animals , Antibody Formation , Bacterial Adhesion , Cell Line, Tumor , Cells, Cultured , Colony Count, Microbial , Disease Models, Animal , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Gastritis/metabolism , Gastritis/pathology , Gastritis/prevention & control , Helicobacter Infections/complications , Helicobacter Infections/immunology , Helicobacter Infections/metabolism , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Immunity, Cellular , Mice , Mice, Knockout , Mucin-1/genetics , Parietal Cells, Gastric/metabolism , Parietal Cells, Gastric/microbiology , Protein Binding , Severity of Illness Index , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...