Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38750967

ABSTRACT

The immune response against Legionella longbeachae, a causative agent of the often-fatal Legionnaires' pneumonia, is poorly understood. Here we investigated the specific roles of tissue-resident alveolar macrophages (AM) and infiltrating phagocytes during infection with this pathogen. AM were the predominant cell type that internalized bacteria one day after infection. Three and five days after infection, AM numbers were greatly reduced while there was an influx of neutrophils and later monocyte-derived cells (MC) into lung tissue. AM carried greater numbers of viable L.longbeachae than neutrophils and MC, which correlated with a higher capacity of L.longbeachae to translocate bacterial effector proteins required for bacterial replication into the AM cytosol. Cell ablation experiments demonstrated that AM promoted infection whereas neutrophils and MC were required for efficient bacterial clearance. IL-18 was important for IFN-γ production by IL-18R+ NK cells and T cells which, in turn, stimulated ROS-mediated bactericidal activity in neutrophils resulting in restriction of L.longbeachae infection. Ciliated bronchiolar epithelial cells also expressed IL-18R but did not play a role in IL-18-mediated L.longbeachae clearance. Our results have identified opposing innate functions of tissue-resident and infiltrating immune cells during L.longbeachae infection that may be manipulated to improve protective responses.

2.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L373-L384, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36719079

ABSTRACT

Legionella pneumophila is the main etiological agent of Legionnaires' disease, a severe bacterial pneumonia. L. pneumophila is initially engulfed by alveolar macrophages (AMs) and subvert normal cellular functions to establish a replicative vacuole. Cigarette smokers are particularly susceptible to developing Legionnaires' disease and other pulmonary infections; however, little is known about the cellular mechanisms underlying this susceptibility. To investigate this, we used a mouse model of acute cigarette smoke exposure to examine the immune response to cigarette smoke and subsequent L. pneumophila infection. Contrary to previous reports, we show that cigarette smoke exposure alone causes a significant depletion of AMs using enzymatic digestion to extract cells, or via imaging intact lung lobes by light-sheet microscopy. Furthermore, treatment of mice deficient in specific types of cell death with smoke suggests that NLRP3-driven pyroptosis is a contributor to smoke-induced death of AMs. After infection, smoke-exposed mice displayed increased pulmonary L. pneumophila loads and developed more severe disease compared with air-exposed controls. We tested if depletion of AMs was related to this phenotype by directly depleting them with clodronate liposomes and found that this also resulted in increased L. pneumophila loads. In summary, our results showed that cigarette smoke depleted AMs from the lung and that this likely contributed to more severe Legionnaires' disease. Furthermore, the role of AMs in L. pneumophila infection is more nuanced than simply providing a replicative niche, and our studies suggest they play a major role in bacterial clearance.


Subject(s)
Cigarette Smoking , Legionella pneumophila , Legionnaires' Disease , Mice , Animals , Macrophages, Alveolar/metabolism , Legionnaires' Disease/metabolism , Legionnaires' Disease/microbiology , Lung/microbiology
3.
Biochem Soc Trans ; 49(3): 1287-1297, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34003245

ABSTRACT

Interferon (IFN)-induced guanosine triphosphate hydrolysing enzymes (GTPases) have been identified as cornerstones of IFN-mediated cell-autonomous defence. Upon IFN stimulation, these GTPases are highly expressed in various host cells, where they orchestrate anti-microbial activities against a diverse range of pathogens such as bacteria, protozoan and viruses. IFN-induced GTPases have been shown to interact with various host pathways and proteins mediating pathogen control via inflammasome activation, destabilising pathogen compartments and membranes, orchestrating destruction via autophagy and the production of reactive oxygen species as well as inhibiting pathogen mobility. In this mini-review, we provide an update on how the IFN-induced GTPases target pathogens and mediate host defence, emphasising findings on protection against bacterial pathogens.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , GTP Phosphohydrolases/immunology , Immunity, Innate/immunology , Interferons/immunology , Animals , Bacteria/pathogenicity , Bacterial Infections/metabolism , Bacterial Infections/microbiology , GTP Phosphohydrolases/metabolism , Host-Pathogen Interactions/immunology , Humans , Interferons/metabolism , Signal Transduction/immunology , Virulence/immunology
4.
J Leukoc Biol ; 107(2): 273-284, 2020 02.
Article in English | MEDLINE | ID: mdl-31793076

ABSTRACT

Legionella pneumophila is an opportunistic human pathogen and causative agent of the acute pneumonia known as Legionnaire's disease. Upon inhalation, the bacteria replicate in alveolar macrophages (AM), within an intracellular vacuole termed the Legionella-containing vacuole. We recently found that, in vivo, IFNγ was required for optimal clearance of intracellular L. pneumophila by monocyte-derived cells (MC), but the cytokine did not appear to influence clearance by AM. Here, we report that during L. pneumophila lung infection, expression of the IFNγ receptor subunit 1 (IFNGR1) is down-regulated in AM and neutrophils, but not MC, offering a possible explanation for why AM are unable to effectively restrict L. pneumophila replication in vivo. To test this, we used mice that constitutively express IFNGR1 in AM and found that prevention of IFNGR1 down-regulation enhanced the ability of AM to restrict L. pneumophila intracellular replication. IFNGR1 down-regulation was independent of the type IV Dot/Icm secretion system of L. pneumophila indicating that bacterial effector proteins were not involved. In contrast to previous work, we found that signaling via type I IFN receptors was not required for IFNGR1 down-regulation in macrophages but rather that MyD88- or Trif- mediated NF-κB activation was required. This work has uncovered an alternative signaling pathway responsible for IFNGR1 down-regulation in macrophages during bacterial infection.


Subject(s)
Legionella pneumophila/growth & development , Legionnaires' Disease/microbiology , Lung/microbiology , Macrophages, Alveolar/microbiology , NF-kappa B/metabolism , Receptors, Interferon/antagonists & inhibitors , Animals , Down-Regulation , Interferon Type I/metabolism , Legionella pneumophila/metabolism , Legionnaires' Disease/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Mice , Mice, Transgenic , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction , Interferon gamma Receptor
5.
Sci Rep ; 9(1): 13295, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527638

ABSTRACT

Neutrophil elastase is a serine protease that has been implicated in the pathogenesis of inflammatory bowel disease. Due to post-translational control of its activation and high expression of its inhibitors in the gut, measurements of total expression poorly reflect the pool of active, functional neutrophil elastase. Fluorogenic substrate probes have been used to measure neutrophil elastase activity, though these tools lack specificity and traceability. PK105 is a recently described fluorescent activity-based probe, which binds to neutrophil elastase in an activity-dependent manner. The irreversible nature of this probe allows for accurate identification of its targets in complex protein mixtures. We describe the reactivity profile of PK105b, a new analogue of PK105, against recombinant serine proteases and in tissue extracts from healthy mice and from models of inflammation induced by oral cancer and Legionella pneumophila infection. We apply PK105b to measure neutrophil elastase activation in an acute model of experimental colitis. Neutrophil elastase activity is detected in inflamed, but not healthy, colons. We corroborate this finding in mucosal biopsies from patients with ulcerative colitis. Thus, PK105b facilitates detection of neutrophil elastase activity in tissue lysates, and we have applied it to demonstrate that this protease is unequivocally activated during colitis.


Subject(s)
Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Leukocyte Elastase/metabolism , Neutrophil Activation/immunology , Neutrophils/immunology , Animals , Cells, Cultured , Female , Humans , Legionella pneumophila/immunology , Legionnaires' Disease/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mouth Neoplasms/pathology
6.
Front Immunol ; 10: 608, 2019.
Article in English | MEDLINE | ID: mdl-31024525

ABSTRACT

We have examined the influence of depleting plasmacytoid dendritic cells (pDC) in mice on the immune response to the gut pathogen Citrobacter rodentium, an organism that is a model for human attaching effacing pathogens such as enterohaemorraghic E. coli. A significantly higher number of C. rodentium were found in mice depleted of pDC from 7 days after infection and pDC depleted mice showed increased gut pathology and higher levels of mRNA encoding inflammatory cytokines in the colon upon infection. pDC-depletion led to a compromising of the gut mucosal barrier that may have contributed to increased numbers of C. rodentium in systemic organs. pDC-depleted mice infected with C. rodentium suffered substantial weight loss necessitating euthanasia. A number of observations suggested that this was not simply the result of dysregulation of immunity in the colon as pDC-depleted mice infected intravenously with C. rodentium also exhibited exacerbated weight loss, arguing that pDC influence systemic immune responses. Overall, these data indicate that pDC contribute at multiple levels to immunity to C. rodentium including control of bacterial numbers in the colon, maintenance of colon barrier function and regulation of immune responses to disseminated bacteria.


Subject(s)
Citrobacter rodentium/immunology , Colitis/immunology , Dendritic Cells/immunology , Enterobacteriaceae Infections/immunology , Animals , Colitis/microbiology , Colon/immunology , Colon/microbiology , Cytokines/immunology , Enterobacteriaceae Infections/microbiology , Inflammation/immunology , Inflammation/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL
7.
Methods Mol Biol ; 1921: 399-417, 2019.
Article in English | MEDLINE | ID: mdl-30694506

ABSTRACT

Infection of C57BL/6 mice with wild-type Legionella pneumophila typically results in very mild disease. However, in mice where the cytosolic recognition of flagellin is impaired by mutation, L. pneumophila infection results in more severe lung inflammation that is reminiscent of Legionnaires' disease. This can be replicated in wild-type mice by using aflagellated mutants of L. pneumophila. These models greatly facilitate the investigation of L. pneumophila virulence factors and the complex pulmonary immune system that is triggered by infection. Here we describe methods for infecting C57BL/6 mice with aflagellated L. pneumophila, the quantification of bacterial load in the lungs and isolation and analysis of invading immune cells. These assays enable the identification of phagocyte subsets and can determine whether phagocytic cells act as a replicative niche for L. pneumophila replication.


Subject(s)
Host-Pathogen Interactions , Legionella pneumophila/physiology , Legionnaires' Disease/microbiology , Animals , Bacterial Load , Biomarkers , Disease Models, Animal , Female , Flow Cytometry , Legionnaires' Disease/immunology , Legionnaires' Disease/pathology , Lung/metabolism , Lung/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/pathology , Male , Mice , Phagocytes/immunology , Phagocytes/metabolism , Phagocytes/microbiology
8.
Eur J Immunol ; 47(1): 155-167, 2017 01.
Article in English | MEDLINE | ID: mdl-27759162

ABSTRACT

How the immune system maintains peripheral tolerance under inflammatory conditions is poorly understood. Here we assessed the fate of gastritogenic T cells following inflammatory activation in vivo. Self-reactive T cells (A23 T cells) specific for the gastric H+ /K+ ATPase α subunit (HKα) were transferred into immunosufficient recipient mice and immunised at a site distant to the stomach with adjuvant containing the cognate HKα peptide antigen. Activation of A23 T cells by immunisation did not impact on either immune tolerance or protection from gastric autoimmunity in wild-type BALB/c mice. However, increased presentation of endogenously derived HKα epitopes by dendritic cells (DCs) in the gastric lymph node of IE-H+ /K+ ß transgenic mice (IEß) reduces A23 T-cell tolerance to gastric antigens after inflammatory activation, with subsequent development of gastritis. While HKα-specific A23 T cells from immunised wild-type mice were poorly responsive to in vitro antigen specific activation, A23 T cells from immunised IEß transgenic mice were readily re-activated, indicating loss of T-cell anergy. These findings show that DCs of gastric lymph nodes can maintain tolerance of pathogenic T cells following inflammatory stimulation and that the density of endogenous antigen presented to self-reactive T cells is critical in the balance between tolerance and autoimmunity.


Subject(s)
Antigen Presentation , Autoantigens/immunology , Autoimmunity , Disease Susceptibility , Gastritis/immunology , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival/genetics , Cell Survival/immunology , Clonal Anergy/genetics , Clonal Anergy/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Gastritis/metabolism , Gastritis/pathology , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, Transgenic , Phenotype , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
9.
Int Immunol ; 28(10): 513-518, 2016 10.
Article in English | MEDLINE | ID: mdl-27371614

ABSTRACT

Thymus-derived regulatory T cells (Tregs) are essential for the maintenance of immunological tolerance. Diverse signalling pathways contribute to thymic Treg cells (tTregs) development; however, the role of mammalian target of rapamycin (mTOR) remains unclear. Rapamycin is a well-characterized inhibitor of mTOR complex 1 signalling and a potent inducer of Treg cells in the periphery. However, the effect of rapamycin on the development of tTregs is poorly defined. Here we have used thymic organ culture to investigate the effect of rapamycin on tTreg development. We show that, contrary to its effect in the periphery, rapamycin inhibits the development of tTregs in wild-type thymi. The inhibition of tTregs by rapamycin could be rescued by a deficiency of Bim. However, rapamycin did not inhibit the development of antigen-specific TCR transgenic tTregs in response to exogenous peptide antigen, indicating that the development of thymic Foxp3+CD4+ cells was not intrinsically inhibited by rapamycin. Collectively our data demonstrate that rapamycin results in a reduction of tTregs because of Bim-mediated apoptosis of immature tTregs via a cell extrinsic mechanism. These findings are important not only for understanding the mechanism of tTreg induction but also for an appreciation of the impact of the clinical application of rapamycin.


Subject(s)
Bcl-2-Like Protein 11/metabolism , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Thymus Gland/drug effects , Thymus Gland/immunology , Animals , Bcl-2-Like Protein 11/deficiency , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Culture Techniques , Thymus Gland/cytology
10.
PLoS Pathog ; 12(6): e1005691, 2016 06.
Article in English | MEDLINE | ID: mdl-27300652

ABSTRACT

Legionella pneumophila is the causative agent of Legionnaires' disease, a potentially fatal lung infection. Alveolar macrophages support intracellular replication of L. pneumophila, however the contributions of other immune cell types to bacterial killing during infection are unclear. Here, we used recently described methods to characterise the major inflammatory cells in lung after acute respiratory infection of mice with L. pneumophila. We observed that the numbers of alveolar macrophages rapidly decreased after infection coincident with a rapid infiltration of the lung by monocyte-derived cells (MC), which, together with neutrophils, became the dominant inflammatory cells associated with the bacteria. Using mice in which the ability of MC to infiltrate tissues is impaired it was found that MC were required for bacterial clearance and were the major source of IL12. IL12 was needed to induce IFNγ production by lymphoid cells including NK cells, memory T cells, NKT cells and γδ T cells. Memory T cells that produced IFNγ appeared to be circulating effector/memory T cells that infiltrated the lung after infection. IFNγ production by memory T cells was stimulated in an antigen-independent fashion and could effectively clear bacteria from the lung indicating that memory T cells are an important contributor to innate bacterial defence. We also determined that a major function of IFNγ was to stimulate bactericidal activity of MC. On the other hand, neutrophils did not require IFNγ to kill bacteria and alveolar macrophages remained poorly bactericidal even in the presence of IFNγ. This work has revealed a cooperative innate immune circuit between lymphoid cells and MC that combats acute L. pneumophila infection and defines a specific role for IFNγ in anti-bacterial immunity.


Subject(s)
Interferon-gamma/immunology , Legionnaires' Disease/immunology , Lymphocytes/immunology , Monocytes/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Flow Cytometry , Immunity, Innate/immunology , Legionella pneumophila/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Polymerase Chain Reaction
11.
Eur J Immunol ; 44(12): 3621-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25231532

ABSTRACT

Depletion of Foxp3(+) CD4(+) regulatory T cells (Treg) in adults results in chronic inflammation and autoimmune disease. However, the impact of transient Treg-cell depletion on self-reactive responses is poorly defined. Here, we studied the effect of transient depletion of Treg cells on CD4(+) T-cell responses to endogenous self-antigens. Short-term ablation of Treg cells in mice resulted in rapid activation of CD4(+) T cells, increased percentage of IFN-γ(+) and Th17 cells in lymphoid organs, and development of autoimmune gastritis. To track self-reactive responses, we analyzed the activation of naïve gastric-specific CD4(+) T cells. There was a dramatic increase in proliferation and acquisition of effector function of gastric-specific T cells in the stomach draining LNs of Treg-cell-depleted mice, compared with untreated mice, either during Treg-cell depletion or after Treg-cell reconstitution. Moreover, the hyperproliferation of gastric-specific T cells in the Treg-cell-ablated mice was predominantly antigen-dependent. Transient depletion of Treg cells resulted in a shift in the ratio of peripheral:thymic Treg cells in the reemerged Treg-cell population, indicating an altered composition of Treg cells. These findings indicate that transient Treg-cell depletion results in ongoing antigen-driven self-reactive T-cell responses and emphasize the continual requirement for an intact Treg-cell population.


Subject(s)
Autoimmunity , Cell Proliferation , Lymphocyte Depletion , T-Lymphocytes, Regulatory/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Stomach/immunology , Stomach/pathology , T-Lymphocytes, Regulatory/pathology
12.
PLoS One ; 9(8): e104753, 2014.
Article in English | MEDLINE | ID: mdl-25111504

ABSTRACT

Breast cancer resistance protein (ABCG2), a member of the ATP-binding cassette transporters has been identified as a major determinant of multidrug resistance (MDR) in cancer cells, but ABC transporter inhibition has limited therapeutic value in vivo. In this research, we demonstrated that inhibition of efflux transporters ABCG2 induced the generation of tolerogenic DCs from human peripheral blood myeloid DCs (mDCs). ABCG2 expression was present in mDCs and was further increased by LPS stimulation. Treatment of CD1c+ mDCs with an ABCG2 inhibitor, Ko143, during LPS stimulation caused increased production of IL-10 and decreased production of pro-inflammatory cytokines and decreased expression of CD83 and CD86. Moreover, inhibition of ABCG2 in monocyte-derived DCs (MDDCs) abrogated the up-regulation of co-stimulatory molecules and production of pro-inflammatory cytokines in these cells in response to LPS. Furthermore, CD1c+ mDCs stimulated with LPS plus Ko143 inhibited the proliferation of allogeneic and superantigen-specific syngenic CD4+ T cells and promoted expansion of CD25+FOXP3+ regulatory T (Treg) cells in an IL-10-dependent fashion. These tolerogenic effects of ABCG2 inhibition could be abolished by ERK inhibition. Thus, we demonstrated that inhibition of ABCG2 in LPS-stimulated mDCs can potently induce tolerogenic potentials in these cells, providing crucial new information that could lead to development of better strategies to combat MDR cancer.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Adenosine/analogs & derivatives , Dendritic Cells/drug effects , Dendritic Cells/immunology , Lipopolysaccharides/pharmacology , Myeloid Cells/cytology , Neoplasm Proteins/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adenosine/pharmacology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Diketopiperazines , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Heterocyclic Compounds, 4 or More Rings , Humans , Interleukin-10/metabolism , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Phosphorylation/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Up-Regulation/drug effects , Up-Regulation/immunology
13.
J Immunol ; 192(11): 5023-30, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24760154

ABSTRACT

It has been proposed that activation of dendritic cells (DCs) presenting self-antigens during inflammation may lead to activation of autoreactive T cells and the development of autoimmunity. To test this hypothesis, we examined the presentation of the autoantigen recognized in autoimmune gastritis, gastric H(+)/K(+) ATPase, which is naturally expressed in the stomach and is constitutively presented in the stomach-draining lymph nodes. Systemic administration to mice of the TLR9 agonist CpG DNA, agonist anti-CD40 Ab, or TLR4 agonist LPS all failed to abrogate the process of peripheral clonal deletion of H(+)/K(+) ATPase-specific CD4 T cells or promote the development of autoimmune gastritis. We demonstrated that migratory DCs from the stomach-draining lymph nodes are the only DC subset capable of constitutively presenting the endogenous gastric H(+)/K(+) ATPase autoantigen in its normal physiological context. Analysis of costimulatory molecules indicated that, relative to resident DCs, migratory DCs displayed a partially activated phenotype in the steady state. Furthermore, migratory DCs were refractory to stimulation by transient exposure to TLR agonists, as they failed to upregulate costimulatory molecules, secrete significant amounts of inflammatory cytokines, or induce differentiation of effector T cells. Together, these data show that transient systemic inflammation failed to break tolerance to the gastric autoantigen, as migratory DCs presenting the gastric autoantigen remain tolerogenic under such conditions, demonstrating the robust nature of peripheral tolerance.


Subject(s)
Autoantigens/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Immune Tolerance , Stomach/immunology , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacology , Animals , Autoantigens/genetics , Cell Movement/drug effects , Cell Movement/genetics , Dendritic Cells/pathology , Inflammation/chemically induced , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Oligodeoxyribonucleotides/adverse effects , Oligodeoxyribonucleotides/pharmacology , Stomach/pathology , Toll-Like Receptor 9/agonists , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology
14.
Eur J Immunol ; 44(7): 2048-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24740292

ABSTRACT

The expression of the Ikaros transcription factor family member, Helios, has been shown to be associated with T-cell tolerance in both the thymus and the periphery. To better understand the importance of Helios in tolerance pathways, we have examined the expression of Helios in TCR-transgenic T cells specific for the gastric H(+) /K(+) ATPase, the autoantigen target in autoimmune gastritis. Analysis of H(+) /K(+) ATPase-specific T cells in mice with different patterns of H(+) /K(+) ATPase expression revealed that, in addition to the expression of Helios in CD4(+) Foxp3(+) regulatory T (Treg) cells, Helios is expressed by a large proportion of CD4(+) Foxp3(-) T cells in both the thymus and the paragastric lymph node (PgLN), which drains the stomach. In the thymus, Helios was expressed by H(+) /K(+) ATPase-specific thymocytes that were undergoing negative selection. In the periphery, Helios was expressed in H(+) /K(+) ATPase-specific CD4(+) T cells following H(+) /K(+) ATPase presentation and was more highly expressed when T-cell activation occurred in the absence of inflammation. Analysis of purified H(+) /K(+) ATPase-specific CD4(+) Foxp3(-) Helios(+) T cells demonstrated that they were functionally anergic. These results demonstrate that Helios is expressed by thymic and peripheral T cells that are being driven to tolerance in response to a genuine autoantigen.


Subject(s)
DNA-Binding Proteins/physiology , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Transcription Factors/physiology , Animals , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Forkhead Transcription Factors/analysis , Gastritis/immunology , H(+)-K(+)-Exchanging ATPase/physiology , Mice , Mice, Inbred BALB C
15.
Curr Top Microbiol Immunol ; 376: 271-91, 2013.
Article in English | MEDLINE | ID: mdl-23918179

ABSTRACT

Legionella pneumophila is an accidental respiratory pathogen of humans that provokes a robust inflammatory response upon infection. While most people exposed to L. pneumophila will clear the infection, certain groups with underlying susceptibility will develop Legionnaires' disease. Mice, like most humans, are inherently resistant to L. pneumophila and infection of most inbred strains reflects the response of immune competent people to L. pneumophila exposure. Hence, the use of mouse models of L. pneumophila infection has taught us a great deal about the innate and adaptive factors that lead to successful clearance of the pathogen and avoidance of Legionnaires' disease. At the same time, L. pneumophila has provided new insight into innate immunity in general and is now a model pathogen with which to study acute lung inflammation and inflammasome activation. This chapter will explore the history and use of the mouse model of L. pneumophila infection and examine what we know about the innate and adaptive factors that contribute to the control of L. pneumophila in the mouse lung.


Subject(s)
Disease Models, Animal , Legionnaires' Disease/immunology , Adaptive Immunity , Animals , Cytokines/biosynthesis , Guinea Pigs , Immunity, Innate , Lung/immunology , Mice , Toll-Like Receptors/physiology
16.
Eur J Immunol ; 43(5): 1286-96, 2013 May.
Article in English | MEDLINE | ID: mdl-23420509

ABSTRACT

Autoimmune disease can be prevented with immunosuppressive agents; however, the effectiveness of these treatments in advanced stage of disease and the fate of pathogenic T cells following such treatments are not clear. In this study we demonstrate that a single dose of in vitro-induced Treg cells (iTreg cells) resulted in the functional repair and restitution of stomach tissue that had been severely damaged in advanced autoimmune gastritis. iTreg cells caused depletion or inactivation of autoreactive naïve T cells that were antigen inexperienced, however, autoreactive effector/memory T cells persisted in treated mice, resulting in residual cellular infiltrates within the repaired stomach tissue. The persisting autoreactive T cells were able to rapidly cause autoimmune disease if iTreg cells were removed. Similar data were obtained from mice treated continuously with corticosteroid, in that there was substantial restitution of the gastric mucosa; however, effector T cells persisted and rapidly caused pathology following drug removal. Therefore, iTreg cells or corticosteroid can suppress pathogenic autoreactive cells in advanced autoimmune disease, reversing tissue damage and improving tissue function. However, the persistence of pathogenic T cells represents a disease risk.


Subject(s)
Autoimmune Diseases/pathology , Gastritis/pathology , Immunotherapy, Adoptive , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Adrenal Cortex Hormones/pharmacology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Cell Communication , Disease Models, Animal , Gastritis/immunology , Gastritis/therapy , Immunosuppression Therapy , Lymphocyte Activation , Mice , Prednisolone/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation
17.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23154976

ABSTRACT

Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.


Subject(s)
Gastric Acid/metabolism , H(+)-K(+)-Exchanging ATPase/metabolism , KCNQ1 Potassium Channel/metabolism , Parietal Cells, Gastric/enzymology , Animals , Cell Membrane/enzymology , Chromans/pharmacology , Cytoplasm/enzymology , Endosomes/enzymology , Fluorescence Resonance Energy Transfer , Histamine/metabolism , KCNQ1 Potassium Channel/drug effects , Mice , Mice, Transgenic , Microscopy, Fluorescence , Parietal Cells, Gastric/drug effects , Parietal Cells, Gastric/metabolism , Potassium Channel Blockers/pharmacology , Protein Transport , Sulfonamides/pharmacology , Time Factors , rab GTP-Binding Proteins/metabolism
18.
Eur J Immunol ; 42(10): 2574-83, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22777705

ABSTRACT

IL-17, produced by a distinct lineage of CD4(+) helper T (Th) cells termed Th17 cells, induces the production of pro-inflammatory cytokines from resident cells and it has been demonstrated that over-expression of IL-17 plays a crucial role in the onset of several auto-immune diseases. Here we examined the role of IL-17 in the pathogenesis of autoimmune gastritis, a disease that was previously believed to be mediated by IFN-γ. Significantly higher levels of IL-17 and IFN-γ were found in the stomachs and stomach-draining lymph nodes of mice with severe autoimmune gastritis. Unlike IL-17, which was produced solely by CD4(+) T cells in gastritic mice, the majority of IFN-γ-producing cells were CD8(+) T cells. However, CD8(+) T cells alone were not able to induce autoimmune gastritis. T cells that were deficient in IL-17 or IFN-γ production were able to induce autoimmune gastritis but to a much lower extent compared with the disease induced by wild-type T cells. These data demonstrate that production of neither IL-17 nor IFN-γ by effector T cells is essential for the initiation of autoimmune gastritis, but suggest that both are required for the disease to progress to the late pathogenic stage that includes significant tissue disruption.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gastritis/immunology , Interferon-gamma/metabolism , Interleukin-17/metabolism , Th17 Cells/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/transplantation , Cells, Cultured , Disease Progression , Interferon-gamma/genetics , Interleukin-17/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Th17 Cells/transplantation
19.
Gut ; 61(10): 1398-409, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22180059

ABSTRACT

BACKGROUND AND AIMS: IL-is important in gastric damage, mucosal repair and gastric cancer progression. We analysed IL-11 expression in H.pylori infected mouse stomach, the site of gastric IL-11 expression in mice and humans, and the effect of exogenous IL-11 on gastric mucosal homeostasis. METHODS: IL-11 protein was localised in mouse and human stomach. The impact of chronic, exogenous IL-11 on normal mouse stomach was examined histologically and transcriptionally by microarray, confirmed by mRNA and protein analysis. Functional impact of IL-11 on gastric acid secretion was determined. RESULTS: In mice infected with H.pylori, IL-11 was increased in fundic mucosa with temporal expression similar to IL-1b. IL-11 protein was localised predominantly to parietal cells in mouse and human stomach. Application of exogenous IL-11 to resulted in fundic parietal and chief cell loss, hyperplasia, mucous cell metaplasia and inflammation. Coincident with cellular changes were an increased gastric pH, altered parietal cell ultrastructure and altered gene expression, particularly genes involved in immune response and ion transport which could result in compromised acid secretion. We confirmed that a single dose of IL-11 effectively ablated the gastric response to histamine. CONCLUSIONS: IL-11 is a parietal cell cytokine that blocks gastric acid secretion, likely via reducing expression of parietal cell ion transport genes, CCKb and histamine H2 receptors. IL-11 expression is increased in H. pylori infected mouse stomach and treatment of wild type mice with IL-11 induced changes in the gastric fundic mucosa reminiscent of chronic atrophic gastritis, a precursor to gastric cancer.


Subject(s)
Gastritis, Atrophic/metabolism , Helicobacter Infections/metabolism , Helicobacter pylori , Interleukin-11/metabolism , Parietal Cells, Gastric/metabolism , Animals , Biomarkers/metabolism , Gastric Acid/metabolism , Gastritis, Atrophic/microbiology , Gastritis, Atrophic/pathology , Helicobacter Infections/complications , Helicobacter Infections/pathology , Humans , Immunoblotting , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Parietal Cells, Gastric/pathology , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism
20.
PLoS One ; 6(11): e27153, 2011.
Article in English | MEDLINE | ID: mdl-22096532

ABSTRACT

Autoimmune gastritis results from the breakdown of T cell tolerance to the gastric H(+)/K(+) ATPase. The gastric H(+)/K(+) ATPase is responsible for the acidification of gastric juice and consists of an α subunit (H/Kα) and a ß subunit (H/Kß). Here we show that CD4(+) T cells from H/Kα-deficient mice (H/Kα(-/-)) are highly pathogenic and autoimmune gastritis can be induced in sublethally irradiated wildtype mice by adoptive transfer of unfractionated CD4(+) T cells from H/Kα(-/-) mice. All recipient mice consistently developed the most severe form of autoimmune gastritis 8 weeks after the transfer, featuring hypertrophy of the gastric mucosa, complete depletion of the parietal and zymogenic cells, and presence of autoantibodies to H(+)/K(+) ATPase in the serum. Furthermore, we demonstrated that the disease significantly affected stomach weight and stomach pH of recipient mice. Depletion of parietal cells in this disease model required the presence of both H/Kα and H/Kß since transfer of H/Kα(-/-) CD4(+) T cells did not result in depletion of parietal cells in H/Kα(-/-) or H/Kß(-/-) recipient mice. The consistency of disease severity, the use of polyclonal T cells and a specific T cell response to the gastric autoantigen make this an ideal disease model for the study of many aspects of organ-specific autoimmunity including prevention and treatment of the disease.


Subject(s)
Autoimmune Diseases/immunology , Gastritis/immunology , Animals , Autoimmune Diseases/etiology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Gastric Mucosa/metabolism , Gastritis/etiology , H(+)-K(+)-Exchanging ATPase/genetics , H(+)-K(+)-Exchanging ATPase/metabolism , Mice , Mice, Mutant Strains , Stomach/pathology , T-Lymphocytes, Regulatory
SELECTION OF CITATIONS
SEARCH DETAIL
...