Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 12(4)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38675791

ABSTRACT

Nanoparticles show great promise as a platform for developing vaccines for the prevention of infectious disease. We have been investigating a method whereby nanocapsules can be formulated from protein, such that the final capsules contain only the cross-linked protein itself. Such nanocapsules are made using a silica templating system and can be customised in terms of size and porosity. Here we compare the construction and characteristics of nanocapsules from four different proteins: one a model protein (ovalbumin) and three from infectious disease pathogens, namely the influenza virus, Helicobacter pylori and HIV. Two of the nanocapsules were assessed further. We confirm that nanocapsules constructed from the urease A subunit of H. pylori can reduce subsequent infection in a vaccinated mouse model. Further, we show that capsules constructed from the HIV gp120 protein can be taken up by dendritic cells in tissue culture and can be recognised by antibodies raised against the virus. These results point to the utility of this method in constructing protein-only nanocapsules from proteins of varying sizes and isoelectric points.

2.
Sci Rep ; 14(1): 8587, 2024 04 13.
Article in English | MEDLINE | ID: mdl-38615147

ABSTRACT

Helicobacter pylori infects approximately half the human population and has an unusual infective niche of the human stomach. Helicobacter pylori is a major cause of gastritis and has been classified as a group 1 carcinogen by the WHO. Treatment involves triple or quadruple antibiotic therapy, but antibiotic resistance is becoming increasingly prevalent. Helicobacter pylori expresses certain blood group related antigens (Lewis system) as a part of its lipopolysaccharide (LPS), which is thought to assist in immune evasion. Additionally, H. pylori LPS participates in adhesion to host cells alongside several adhesion proteins. This study profiled the carbohydrates of H. pylori reference strains (SS1 and 26695) using monoclonal antibodies (mAbs) and lectins, identifying interactions between two carbohydrate-targeting mAbs and multiple lectins. Atomic force microscopy (AFM) scans were used to probe lectin and antibody interactions with the bacterial surfaces. The selected mAb and lectins displayed an increased adhesive force over the surface of the curved H. pylori rods. Furthermore, this study demonstrates the ability of anti-carbohydrate antibodies to reduce the adhesion of H. pylori 26695 to human gastric adenocarcinoma cells via AFM. Targeting bacterial carbohydrates to disrupt crucial adhesion and immune evasion mechanisms represents a promising strategy for combating H. pylori infection.


Subject(s)
Blood Group Antigens , Helicobacter Infections , Helicobacter pylori , Humans , Lipopolysaccharides , Polysaccharides , Antibodies, Monoclonal , Lectins
3.
Biology (Basel) ; 13(1)2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38248478

ABSTRACT

As research on parasitic helminths has entered the post-genomic era, research efforts have turned to deciphering the function of genes in the public databases of genome sequences. It is hoped that, by understanding the role of parasite genes in maintaining their parasitic lifestyle, critical insights can be gained to develop new intervention and control strategies. Methods to manipulate and transform parasitic worms are now developed to a point where it has become possible to gain a comprehensive understanding of the molecular mechanisms underlying host-parasite interplay, and here, we summarise and discuss the advances that have been made in schistosome transgenesis over the past 25 years. The ability to genetically manipulate schistosomes holds promise in finding new ways to control schistosomiasis, which ultimately may lead to the eradication of this debilitating disease.

5.
Nanoscale ; 14(48): 17940-17954, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36349848

ABSTRACT

Carbon nanodots (C-dots) have attracted much attention for their use in the fields of bioimaging, drug delivery, and sensing due to their excellent fluorescent and photoluminescent properties, photostability, biocompatibility, and amenability to surface modification. Herein, we report a nanocomposite formulation of C-dots (<5 nm) encapsulated in lipid-based lyotropic liquid crystalline nanoparticles (∼250 nm) via either passive diffusion or electrostatic mechanisms. The physicochemical properties of the nanocomposite formulation including particle size, surface charge, internal cubic nanostructures, and pH-dependent fluorescent properties were characterised. Upon loading of C-dots into lipid nanoparticles, the highly ordered inverse bicontinuous cubic mesophase existed in the internal phase of the nanoparticles, demonstrated by synchrotron small angle X-ray scattering, molecular dynamic simulation and cryogenic transmission electron microscopy. The pH-dependent fluorescent property of the C-dots was modified via electrostatic interaction between the C-dots and cationic lipid nanoparticles, which further enhanced the brightness of C-dots through self-quenching prevention. The cytotoxicity and cellular uptake efficiency of the developed nanocomposites were also examined in an epithelial gastric adenocarcinoma cell line (AGS) and a macrophage cell line (stimulated THP-1). Compared to free C-dots, the uptake and cell imaging potential of the C-dot nanocomposites was significantly improved, by several orders of magnitude as demonstrated by cytoplasmic fluorescent intensities using confocal microscopy. Loading C-dots into mesoporous lipid nanocarriers presents a new way of modifying C-dot physicochemical and fluorescent properties, alternative to direct chemical surface modification, and advances the bioimaging potential of C-dots by enhancing cellular uptake efficiency and converging C-dot light emission.


Subject(s)
Carbon , Nanocomposites , Carbon/chemistry , Drug Delivery Systems/methods , Particle Size , Lipids
6.
Vaccine ; 40(40): 5769-5780, 2022 09 22.
Article in English | MEDLINE | ID: mdl-36064671

ABSTRACT

Vaccines are very effective in providing protection against many infectious diseases. However, it has proven difficult to develop highly efficacious vaccines against some pathogens and so there is a continuing need to improve vaccine technologies. The first successful and widely used vaccines were based on attenuated pathogens (e.g., laboratory passaged Pasteurella multocida to vaccinate against fowl cholera) or closely related non-pathogenic organisms (e.g., cowpox to vaccinate against smallpox). Subsequently, live vaccines, either attenuated pathogens or non-pathogenic microorganisms modified to deliver heterologous antigens, have been successfully used to induce protective immune responses against many pathogens. Unlike conventional killed and subunit vaccines, live vaccines can deliver antigens to mucosal surfaces in a similar manner and context as the natural infection and hence can often produce a more appropriate and protective immune response. Despite these advantages, there is still a need to improve the immunogenicity of some live vaccines. The efficacy of injectable killed and subunit vaccines is usually enhanced using adjuvants such mineral salts, oils, and saponin, but such adjuvants cannot be used with live vaccines. Instead, live vaccines can be engineered to produce immunomodulatory molecules that can stimulate the immune system to induce more robust and long-lasting adaptive immune responses. This review focuses on research that has been undertaken to engineer live vaccines to produce immunomodulatory molecules that act as adjuvants to increase immunogenicity. Adjuvant strategies with varying mechanisms of action (inflammatory, antibody-mediated, cell-mediated) and delivery modes (oral, intramuscular, intranasal) have been investigated, with varying degrees of success. The goal of such research is to define adjuvant strategies that can be adapted to enhance live vaccine efficacy by triggering strong innate and adaptive immune responses and produce vaccines against a wider range of pathogens.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Vaccines , Adjuvants, Immunologic , Humans , Vaccines, Attenuated , Vaccines, Subunit
7.
Front Immunol ; 13: 868225, 2022.
Article in English | MEDLINE | ID: mdl-35634347

ABSTRACT

Helicobacter pylori is an important human pathogen that infects half the human population and can lead to significant clinical outcomes such as acute and chronic gastritis, duodenal ulcer, and gastric adenocarcinoma. To establish infection, H. pylori employs several mechanisms to overcome the innate and adaptive immune systems. H. pylori can modulate interleukin (IL) secretion and innate immune cell function by the action of several virulence factors such as VacA, CagA and the type IV secretion system. Additionally, H. pylori can modulate local dendritic cells (DC) negatively impacting the function of these cells, reducing the secretion of immune signaling molecules, and influencing the differentiation of CD4+ T helper cells causing a bias to Th1 type cells. Furthermore, the lipopolysaccharide (LPS) of H. pylori displays a high degree of phase variation and contains human blood group carbohydrate determinants such as the Lewis system antigens, which are proposed to be involved in molecular mimicry of the host. Lastly, the H. pylori group of outer membrane proteins such as BabA play an important role in attachment and interaction with host Lewis and other carbohydrate antigens. This review examines the various mechanisms that H. pylori utilises to evade the innate immune system as well as discussing how the structure of the H. pylori LPS plays a role in immune evasion.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Humans , Immune Evasion , Lipopolysaccharides , Virulence Factors/metabolism
8.
Helicobacter ; 27(3): e12891, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35384141

ABSTRACT

For decades, traditional in vitro and in vivo models used for the study of Helicobacter pylori infection have relied heavily on the use of gastric cancer cell lines and rodents. Major challenges faced by these methods have been the inability to study cancer initiation in already cancerous cell lines, and the difficulty in translating results obtained in animal models due to genetic differences. These challenges have prevented a thorough understanding of the pathogenesis of disease and slowed the development of cancer therapies and a suitable vaccine against the pathogen. In recent years, the development of gastric organoids has provided great advantages over the traditional in vivo and in vitro models due to their similarities to the human stomach in vivo, their ease of use, and the capacity for long-term culture. This review discusses the advantages and limitations of existing in vivo and in vitro models of H. pylori infection, and how gastric organoids have been applied to study H. pylori pathogenesis, with a focus on how the pathogen interacts with the gastric epithelium, inflammatory processes, epithelial repair, and cancer initiation. The potential applications of organoids to address more complex questions on the role of hormones, vaccine-induced immunity are also discussed.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Animals , Gastric Mucosa/pathology , Helicobacter Infections/pathology , Helicobacter pylori/metabolism , Humans , Inflammation , Organoids , Stomach/pathology , Stomach Neoplasms/metabolism
9.
Int J Mol Sci ; 23(5)2022 Mar 03.
Article in English | MEDLINE | ID: mdl-35269931

ABSTRACT

Gastric cancer represents a significant disease burden worldwide. The factors that initiate cancer are not well understood. Chronic inflammation such as that triggered by H. pylori infection is the most significant cause of gastric cancer. In recent years, organoid cultures developed from human and animal adult stem cells have facilitated great advances in our understanding of gastric homeostasis. Organoid models are now being exploited to investigate the role of host genetics and bacterial factors on proliferation and DNA damage in gastric stem cells. The impact of a chronic inflammatory state on gastric stem cells and the stroma has been less well addressed. This review discusses what we have learned from the use of organoid models to investigate cancer initiation, and highlights questions on the contribution of the microbiota, chronic inflammatory milieu, and stromal cells that can now be addressed by more complex coculture models.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Stomach Neoplasms , Animals , Gastric Mucosa/microbiology , Helicobacter Infections/microbiology , Homeostasis , Inflammation/complications , Organoids , Stomach Neoplasms/genetics
10.
Adv Exp Med Biol ; 1149: 257-275, 2019.
Article in English | MEDLINE | ID: mdl-31016627

ABSTRACT

Helicobacter pylori is a highly-adapted gastrointestinal pathogen of humans and the immunology of this chronic infection is extremely complex. Despite the availability of antibiotic therapy, the global incidence of H. pylori infection remains high, particularly in low to middle-income nations. Failure of therapy and the spread of antibiotic resistance among the bacteria are significant problems and provide impetus for the development of new therapies and vaccines to treat or prevent gastric ulcer, and gastric carcinoma. The expansion of knowledge on gastric conventional and regulatory T cell responses, and the role of TH17 in chronic gastritis from studies in mouse models and patients have provided valuable insights into how gastritis is initiated and maintained. The development of human challenge models for testing candidate vaccines has meant a unique opportunity to study acute infection, but the field of vaccine development has not progressed as rapidly as anticipated. One clear lesson learned from previous studies is that we need a better understanding of the immune suppressive mechanisms in vivo to be able to design vaccine strategies. There is still an urgent need to identify practical surrogate markers of protection that could be deployed in future field vaccine trials. Important developments in our understanding of the chronic inflammatory response, progress and problems arising from human studies, and an outlook for the future of clinical vaccine trials will be discussed.


Subject(s)
Bacterial Vaccines , Gastritis , Helicobacter Infections , Helicobacter pylori , Animals , Bacterial Vaccines/immunology , Gastritis/microbiology , Gastritis/prevention & control , Helicobacter Infections/immunology , Helicobacter Infections/prevention & control , Helicobacter pylori/immunology , Humans
11.
J Biol Chem ; 293(14): 5079-5089, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29449370

ABSTRACT

In response to the widespread emergence of antibiotic-resistant microbes, new therapeutic agents are required for many human pathogens. A non-mammalian polysaccharide, poly-N-acetyl-d-glucosamine (PNAG), is produced by bacteria, fungi, and protozoan parasites. Antibodies that bind to PNAG and its deacetylated form (dPNAG) exhibit promising in vitro and in vivo activities against many microbes. A human IgG1 mAb (F598) that binds both PNAG and dPNAG has opsonic and protective activities against multiple microbial pathogens and is undergoing preclinical and clinical assessments as a broad-spectrum antimicrobial therapy. Here, to understand how F598 targets PNAG, we determined crystal structures of the unliganded F598 antigen-binding fragment (Fab) and its complexes with N-acetyl-d-glucosamine (GlcNAc) and a PNAG oligosaccharide. We found that F598 recognizes PNAG through a large groove-shaped binding site that traverses the entire light- and heavy-chain interface and accommodates at least five GlcNAc residues. The Fab-GlcNAc complex revealed a deep binding pocket in which the monosaccharide and a core GlcNAc of the oligosaccharide were almost identically positioned, suggesting an anchored binding mechanism of PNAG by F598. The Fab used in our structural analyses retained binding to PNAG on the surface of an antibiotic-resistant, biofilm-forming strain of Staphylococcus aureus Additionally, a model of intact F598 binding to two pentasaccharide epitopes indicates that the Fab arms can span at least 40 GlcNAc residues on an extended PNAG chain. Our findings unravel the structural basis for F598 binding to PNAG on microbial surfaces and biofilms.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Polysaccharides, Bacterial/immunology , Antibodies, Monoclonal/chemistry , Biofilms , Carbohydrate Conformation , Crystallography, X-Ray , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/immunology , Immunoglobulin G/chemistry , Models, Molecular , Polysaccharides, Bacterial/chemistry , Protein Conformation , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology
12.
Immunology ; 150(2): 172-183, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27676456

ABSTRACT

There is a current lack of effective mucosal vaccines against major gastroenteric pathogens and particularly against Helicobacter pylori, which causes a chronic infection that can lead to peptic ulcers and gastric cancer in a subpopulation of infected individuals. Mucosal CD4+ T-cell responses have been shown to be essential for vaccine-induced protection against H. pylori infection. The current study addresses the influence of the adjuvant and site of mucosal immunization on early CD4+ T-cell priming to H. pylori antigens. The vaccine formulation consisted of H. pylori lysate antigens and mucosal adjuvants, cholera toxin (CT) or a detoxified double-mutant heat-labile enterotoxin from Escherichia coli (dmLT), which were administered by either the sublingual or intragastric route. We report that in vitro, adjuvants CT and dmLT induce up-regulation of pro-inflammatory gene expression in purified dendritic cells and enhance the H. pylori-specific CD4+ T-cell response including interleukin-17A (IL-17A), interferon-γ (IFN-γ) and tumour necrosis factor-α (TNF-α) secretion. In vivo, sublingual immunization led to an increased frequency of IL-17A+ , IFN-γ+ and TNF-α+ secreting CD4+ T cells in the cervical lymph nodes compared with in the mesenteric lymph nodes after intragastric immunization. Subsequently, IL-17A+ cells were visualized in the stomach of sublingually immunized and challenged mice. In summary, our results suggest that addition of an adjuvant to the vaccine clearly activated dendritic cells, which in turn, enhanced CD4+ T-cell cytokines IL-17A, IFN-γ and TNF-α responses, particularly in the cervical lymph nodes after sublingual vaccination.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Helicobacter Infections/immunology , Helicobacter pylori/immunology , Immunity, Mucosal , Adjuvants, Immunologic/administration & dosage , Administration, Sublingual , Animals , Bacterial Toxins/administration & dosage , Cells, Cultured , Cholera Toxin/administration & dosage , Cytokines/metabolism , Enterotoxins/administration & dosage , Escherichia coli Proteins/administration & dosage , Female , Inflammation Mediators/metabolism , Intubation, Gastrointestinal , Lymphocyte Activation , Mice , Mice, Inbred C57BL
14.
PLoS One ; 10(7): e0131444, 2015.
Article in English | MEDLINE | ID: mdl-26168305

ABSTRACT

CD4+ T cells have been shown to be essential for vaccine-induced protection against Helicobacter pylori infection. However, the effector mechanisms leading to reductions in the gastric bacterial loads of vaccinated mice remain unclear. We have investigated the function of IFN-γ and IL-17A for vaccine-induced protection and inflammation (gastritis) using IFN-γ-gene-knockout (IFN-γ-/-) mice, after sublingual or intragastric immunization with H. pylori lysate antigens and cholera toxin. Bacteria were enumerated in the stomachs of mice and related to the gastritis score and cellular immune responses. We report that sublingually and intragastrically immunized IFN-γ-/- mice had significantly reduced bacterial loads similar to immunized wild-type mice compared to respective unimmunized infection controls. The reduction in bacterial loads in sublingually and intragastrically immunized IFN-γ-/- mice was associated with significantly higher levels of IL-17A in stomach extracts and lower gastritis scores compared with immunized wild-type mice. To study the role of IL-17A for vaccine-induced protection in sublingually immunized IFN-γ-/- mice, IL-17A was neutralized in vivo at the time of infection. Remarkably, the neutralization of IL-17A in sublingually immunized IFN-γ-/- mice completely abolished protection against H. pylori infection and the mild gastritis. In summary, our results suggest that IFN-γ responses in the stomach of sublingually immunized mice promote vaccine-induced gastritis, after infection with H. pylori but that IL-17A primarily functions to reduce the bacterial load.


Subject(s)
Helicobacter Infections/immunology , Helicobacter Infections/prevention & control , Inflammation/complications , Inflammation/pathology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Animals , Antigens, Bacterial/immunology , Cell Proliferation , Gene Expression Regulation , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori , Immunization , Inflammation/immunology , Inflammation/microbiology , Interferon-gamma/deficiency , Lymph Nodes/pathology , Mice, Inbred C57BL , Neutralization Tests , Spleen/pathology , Stomach/microbiology , Stomach/pathology
15.
Infect Immun ; 81(10): 3880-93, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23918779

ABSTRACT

The natural immune response to Helicobacter pylori neither clears infection nor prevents reinfection. However, the ability of secretory antibodies to influence the course of H. pylori infection has not been determined. We compared the natural progression of H. pylori infection in wild-type C57BL/6 mice with that in mice lacking the polymeric immunoglobulin receptor (pIgR) that is essential for the secretion of polymeric antibody across mucosal surfaces. H. pylori SS1-infected wild-type and pIgR knockout (KO) mice were sampled longitudinally for gastrointestinal bacterial load, antibody response, and histological changes. The gastric bacterial loads of wild-type and pIgR KO mice remained constant and comparable at up to 3 months postinfection (mpi) despite SS1-reactive secretory IgA in the intestinal contents of wild-type mice at that time. Conversely, abundant duodenal colonization of pIgR KO animals contrasted with the near-total eradication of H. pylori from the intestine of wild-type animals by 3 mpi. H. pylori was cultured only from the duodenum of those animals in which colonization in the distal gastric antrum was of sufficient density for immunohistological detection. By 6 mpi, the gastric load of H. pylori in wild-type mice was significantly lower than in pIgR KO animals. While there was no corresponding difference between the two mouse strains in gastric pathology results at 6 mpi, reductions in gastric bacterial load correlated with increased gastric inflammation together with an intestinal secretory antibody response in wild-type mice. Together, these results suggest that naturally produced secretory antibodies can modulate the progress of H. pylori infection, particularly in the duodenum.


Subject(s)
Antibodies, Bacterial/metabolism , Helicobacter Infections/immunology , Helicobacter pylori , Immunity, Mucosal/physiology , Intestinal Mucosa/metabolism , Animals , Blotting, Western , Gene Expression Regulation/immunology , Immunoglobulin A/blood , Immunoglobulin A/metabolism , Immunoglobulin G/blood , Immunoglobulin G/metabolism , Intestinal Mucosa/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/metabolism
16.
Naunyn Schmiedebergs Arch Pharmacol ; 386(12): 1081-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23989929

ABSTRACT

Vascular smooth muscle cells (VSMC) are present in arterial intima before atherosclerotic plaques develop and are likely to be exposed to unmodified serum lipids as they enter the vessel wall. We examined the effects of sera from mice on the morphology and function of mouse VSMC. Incubation of a mouse VSMC line (MOVAS) with sera from normocholesterolemic (C57BL/6J) or hypercholesterolemic (APOE(-/-)) mice caused concentration-dependent increases in lipid accumulation as measured by AdipoRed, with the extent of lipid uptake significantly greater with the latter sera type. Inhibition of c-Jun N-terminal kinases (SP600125), Src kinases (AG1879), and clathrin-dependent endocytosis (monodansylcadaverine) to disrupt scavenger receptor-mediated uptake of lipids had no effect on serum-induced lipid accumulation by VSMC. By contrast, inhibition of macropinocytosis with antagonists of PI-3 kinase (LY294002) and actin (cytochalasin D) markedly reduced lipid accumulation. Serum exposure reduced the expression of the ATP-binding cassette transporter A1, consistent with impaired cholesterol efflux, but had no effect on the expression of markers of VSMC differentiation. Moreover, the expression of several inflammation and foam cell markers was unchanged (CCL2, CCL5, and CD68) by mouse sera. The accumulation of unmodified serum lipids by VSMC involves a macropinocytosis-like uptake pathway and is associated with the downregulation of the ATP-binding cassette transporter. We speculate that VSMC may play an atheroprotective role in arterial intima by acting as a "sink" for unmodified lipids.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Hypercholesterolemia/metabolism , Lipids/blood , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pinocytosis , Actins/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Biomarkers/blood , Cell Line , Disease Models, Animal , Down-Regulation , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Inflammation Mediators/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Phosphoinositide-3 Kinase Inhibitors , Pinocytosis/drug effects , Protein Kinase Inhibitors/pharmacology , Time Factors
17.
Infect Immun ; 81(2): 470-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23208607

ABSTRACT

Bacterial infections are a common and serious complication of type 2 diabetes (T2D). The prevalence of melioidosis, an emerging tropical infection caused by the Gram-negative bacterium Burkholderia pseudomallei, is increased in people with T2D. This is the first study to compare murine models of T2D and melioidosis. Susceptibility and disease progression following infection with B. pseudomallei were compared in our diet-induced polygenic mouse model and a leptin receptor-deficient monogenic model of T2D. The metabolic profile of mice with diet-induced diabetes, including body weight, blood glucose, cholesterol, triglycerides, insulin resistance, and baseline levels of inflammation, closely resembled that of clinical T2D. Following subcutaneous infection with B. pseudomallei, bacterial loads at 24 and 72 h postinfection in the blood, spleen, liver, lungs, and subcutaneous adipose tissue (SAT) at the site of infection were compared in parallel with the expression of inflammatory cytokines and tissue histology. As early as 24 h postinfection, the expression of inflammatory (interleukin-1ß [IL-1ß], tumor necrosis factor alpha [TNF-α], and IL-6) and T(H)1 (IL-12 and gamma interferon [IFN-γ]) cytokines was impaired in diabetic mice compared to nondiabetic littermates. Early differences in cytokine expression were associated with excessive infiltration of polymorphonuclear neutrophils (PMN) in diabetic mice compared to nondiabetic littermates. This was accompanied by bacteremia, hematogenous dissemination of bacteria to the lungs, and uncontrolled bacterial growth in the spleens of diabetic mice by 72 h postinfection. The findings from our novel model of T2D and melioidosis comorbidity support the role of impaired early immune pathways in the increased susceptibility of individuals with T2D to bacterial infections.


Subject(s)
Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/microbiology , Melioidosis/immunology , Melioidosis/microbiology , Animals , Burkholderia pseudomallei/immunology , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Inflammation/immunology , Inflammation/microbiology , Interferon-gamma/immunology , Interleukin-12/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Male , Melioidosis/metabolism , Metabolome/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/microbiology , Tumor Necrosis Factor-alpha/immunology
18.
Front Immunol ; 3: 316, 2012.
Article in English | MEDLINE | ID: mdl-23087691

ABSTRACT

Helicobacter pylori infection induces chronic inflammation which is characterized not only by infiltrations of inflammatory cells such as neutrophils and CD4(+) T cells, but also significant populations of regulatory T cells (T(reg)). These cells are important for disease pathogenesis because they are believed to contribute to the persistence of the infection. Despite encouraging results in animal models, the prospects for an effective H. pylori vaccine are currently poor because of generally disappointing results in preclinical and phase 1 trials. As a result, a current major focus of basic research on vaccination is to better understand the mechanisms regulating the inflammatory response with the view it can inform future vaccine design. Our studies in this area have focused on gastric CD4(+) T(reg) in vaccinated mice, and raised the hypothesis that adipokines in particular leptin are involved the establishment of a protective gastric immune response. Here we discuss the hypothesis that vaccination deregulates T(reg) responses in the gastric mucosa, and that this process is mediated by leptin. We propose that reduced suppression permits a protective sub population of H. pylori-specific CD4(+) T cells to exert protective effects, presumably via the gastric epithelium. Evidence from the literature and experimental approaches will be discussed.

19.
Clin Exp Pharmacol Physiol ; 39(3): 218-26, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22211630

ABSTRACT

  NADPH oxidases (Nox) are reactive oxygen species (ROS)-generating enzymes that play important physiological roles in host defence and redox signalling. However, Nox activity is upregulated in the vascular wall during atherosclerosis and contributes to plaque formation by promoting oxidative stress and inflammation.   The bacterium Chlamydia pneumoniae has been detected in vascular smooth muscle cells (VSMC) of human atheroma. We hypothesized that C. pneumoniae infection of VSMC causes Nox activation, which initially limits infection but ultimately causes oxidative stress, activation of pro-inflammatory pathways and an atherogenic phenotype.   Chlamydia pneumoniae infection of mouse cultured VSMC significantly increased ROS production by twofold but did not upregulate mRNA expression of Nox1 or Nox4. Chlamydia pneumoniae did increase Nox2 mRNA levels significantly by threefold, but this did not translate to elevated Nox2 protein expression.   The Nox inhibitor gp91ds-tat had no effect on C. pneumoniae-induced ROS production. In contrast, apocynin significantly reduced ROS levels by 75% in C. pneumoniae-infected VSMC, an effect most likely attributable to its direct anti-oxidant action.   Although apocynin had no effect on C. pneumoniae-induced expression of inflammatory markers, bacteria recovered from apocynin-treated VSMC displayed a higher degree of infectivity in HEp-2 cells.   In conclusion, C. pneumoniae infection increases ROS production in VSMC independently of Nox activity. Although elevated ROS production appears to serve a protective role by limiting the spread of infection, we speculate that this response will be detrimental over the long term by causing oxidative stress and a smouldering inflammatory response by maintaining C. pneumoniae persistence within the cell.


Subject(s)
Chlamydophila Infections/genetics , Chlamydophila pneumoniae/pathogenicity , Immunophenotyping , Inflammation Mediators/physiology , Myocytes, Smooth Muscle/metabolism , Reactive Oxygen Species/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , Chlamydophila Infections/metabolism , Chlamydophila Infections/pathology , Humans , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/microbiology , Myocytes, Smooth Muscle/microbiology
20.
Infect Immun ; 79(10): 4186-92, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807907

ABSTRACT

While gastric adenocarcinoma is the most serious consequence of Helicobacter pylori infection, not all infected persons develop this pathology. Individuals most at risk of this cancer are those in whom the bacteria colonize the acid-secreting region of the stomach and subsequently develop severe inflammation in the gastric corpus. It has been reported anecdotally that male mice become infected with greater numbers of H. pylori bacteria than female mice. While investigating this phenomenon, we found that increased H. pylori infection densities in male mice were not related to antibody production, and this phenomenon was not normalized by gonadectomy. However, the gastric pH in male 129/Sv mice was significantly elevated compared with that in female mice. Differences in colonization were evident within 1 day postinfection and significantly arose due to colonization of the gastric corpus region in male mice. This provided a potential model for comparing the effect of corpus colonization on the development of gastritis. This was explored using two models of H. pylori-induced inflammation, namely, 2-month infections of Muc1(-/-) mice and 6-month infections of wild-type 129/Sv mice. While H. pylori infection of female mice induced a severe, corpus-predominant atrophic gastritis, to our surprise, male mice developed minimal inflammation despite being colonized with significantly more H. pylori bacteria than female controls. Thus, colonization of the gastric corpus in male mice was associated with a loss of inflammation in that region. The suppression of inflammation concomitant with infection of the gastric corpus in male mice demonstrates a powerful localized suppression of inflammation induced at sites of H. pylori colonization.


Subject(s)
Gastric Mucosa/pathology , Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Helicobacter pylori/pathogenicity , Inflammation/pathology , Stomach/pathology , Animals , Disease Models, Animal , Female , Gastric Mucosa/microbiology , Gastritis, Atrophic/immunology , Gastritis, Atrophic/microbiology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/isolation & purification , Humans , Hydrogen-Ion Concentration , Inflammation/microbiology , Male , Mice , Mice, Inbred C57BL , Stomach/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...