Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Am J Physiol Lung Cell Mol Physiol ; 324(3): L373-L384, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36719079

RESUMO

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.


Assuntos
Fumar Cigarros , Legionella pneumophila , Doença dos Legionários , Camundongos , Animais , Macrófagos Alveolares/metabolismo , Doença dos Legionários/metabolismo , Doença dos Legionários/microbiologia , Pulmão/microbiologia
2.
Biochem Soc Trans ; 49(3): 1287-1297, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34003245

RESUMO

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.


Assuntos
Bactérias/imunologia , Infecções Bacterianas/imunologia , GTP Fosfo-Hidrolases/imunologia , Imunidade Inata/imunologia , Interferons/imunologia , Animais , Bactérias/patogenicidade , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , GTP Fosfo-Hidrolases/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferons/metabolismo , Transdução de Sinais/imunologia , Virulência/imunologia
3.
Eur J Immunol ; 47(1): 155-167, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27759162

RESUMO

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.


Assuntos
Apresentação de Antígeno , Autoantígenos/imunologia , Autoimunidade , Suscetibilidade a Doenças , Gastrite/imunologia , Animais , Biomarcadores , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Anergia Clonal/genética , Anergia Clonal/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Gastrite/metabolismo , Gastrite/patologia , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Imunofenotipagem , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fenótipo , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
4.
PLoS Pathog ; 12(6): e1005691, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27300652

RESUMO

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.


Assuntos
Interferon gama/imunologia , Doença dos Legionários/imunologia , Linfócitos/imunologia , Monócitos/imunologia , Transferência Adotiva , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Imunidade Inata/imunologia , Legionella pneumophila/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Reação em Cadeia da Polimerase
6.
Int Immunol ; 28(10): 513-518, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27371614

RESUMO

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.


Assuntos
Proteína 11 Semelhante a Bcl-2/metabolismo , Sirolimo/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Timo/efeitos dos fármacos , Timo/imunologia , Animais , Proteína 11 Semelhante a Bcl-2/deficiência , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Técnicas de Cultura de Órgãos , Timo/citologia
7.
J Immunol ; 192(11): 5023-30, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24760154

RESUMO

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.


Assuntos
Autoantígenos/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica , Estômago/imunologia , Adjuvantes Imunológicos/efeitos adversos , Adjuvantes Imunológicos/farmacologia , Animais , Autoantígenos/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Dendríticas/patologia , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Oligodesoxirribonucleotídeos/efeitos adversos , Oligodesoxirribonucleotídeos/farmacologia , Estômago/patologia , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/imunologia
8.
Eur J Immunol ; 44(12): 3621-31, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25231532

RESUMO

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.


Assuntos
Autoimunidade , Proliferação de Células , Depleção Linfocítica , Linfócitos T Reguladores/imunologia , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Interferon gama/genética , Interferon gama/imunologia , Linfonodos/imunologia , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Estômago/imunologia , Estômago/patologia , Linfócitos T Reguladores/patologia
9.
Eur J Immunol ; 44(7): 2048-58, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24740292

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Tolerância Imunológica , Linfócitos T Reguladores/imunologia , Timo/imunologia , Fatores de Transcrição/fisiologia , Animais , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/análise , Gastrite/imunologia , ATPase Trocadora de Hidrogênio-Potássio/fisiologia , Camundongos , Camundongos Endogâmicos BALB C
10.
Eur J Immunol ; 43(5): 1286-96, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23420509

RESUMO

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.


Assuntos
Doenças Autoimunes/patologia , Gastrite/patologia , Imunoterapia Adotiva , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Corticosteroides/farmacologia , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Comunicação Celular , Modelos Animais de Doenças , Gastrite/imunologia , Gastrite/terapia , Terapia de Imunossupressão , Ativação Linfocitária , Camundongos , Prednisolona/farmacologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/transplante
11.
Curr Top Microbiol Immunol ; 376: 271-91, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23918179

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Doença dos Legionários/imunologia , Imunidade Adaptativa , Animais , Citocinas/biossíntese , Cobaias , Imunidade Inata , Pulmão/imunologia , Camundongos , Receptores Toll-Like/fisiologia
12.
Mucosal Immunol ; 2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38750967

RESUMO

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 (AMs) and infiltrating phagocytes during infection with this pathogen. AMs were the predominant cell type that internalized bacteria 1 day after infection. A total of 3 and 5 days after infection, AM numbers were greatly reduced, whereas there was an influx of neutrophils and, later, monocyte-derived cells (MCs) into lung tissue. AMs carried greater numbers of viable L. longbeachae than neutrophils and MCs, 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. Interleukin (IL)-18 was important for interferon-γ production by IL-18R+ natural killer cells and T cells, which, in turn, stimulated reactive oxygen species-mediated bactericidal activity in neutrophils, resulting in the 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.

13.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23154976

RESUMO

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.


Assuntos
Ácido Gástrico/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Canal de Potássio KCNQ1/metabolismo , Células Parietais Gástricas/enzimologia , Animais , Membrana Celular/enzimologia , Cromanos/farmacologia , Citoplasma/enzimologia , Endossomos/enzimologia , Transferência Ressonante de Energia de Fluorescência , Histamina/metabolismo , Canal de Potássio KCNQ1/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Transporte Proteico , Sulfonamidas/farmacologia , Fatores de Tempo , Proteínas rab de Ligação ao GTP/metabolismo
14.
Eur J Immunol ; 42(10): 2574-83, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22777705

RESUMO

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.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Gastrite/imunologia , Interferon gama/metabolismo , Interleucina-17/metabolismo , Células Th17/imunologia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Células Cultivadas , Progressão da Doença , Interferon gama/genética , Interleucina-17/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Células Th17/transplante
15.
Gut ; 61(10): 1398-409, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22180059

RESUMO

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.


Assuntos
Gastrite Atrófica/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori , Interleucina-11/metabolismo , Células Parietais Gástricas/metabolismo , Animais , Biomarcadores/metabolismo , Ácido Gástrico/metabolismo , Gastrite Atrófica/microbiologia , Gastrite Atrófica/patologia , Infecções por Helicobacter/complicações , Infecções por Helicobacter/patologia , Humanos , Immunoblotting , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Células Parietais Gástricas/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo
16.
J Immunol ; 184(10): 5429-33, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20400697

RESUMO

Plasmacytoid dendritic cells (pDCs) are well known as the major cell type that secretes type I IFN in response to viral infections. Their role in combating other classes of infectious organisms, including bacteria, and their mechanisms of action are poorly understood. We have found that pDCs play a significant role in the acute response to the intracellular bacterial pathogen Legionella pneumophila. pDCs were rapidly recruited to the lungs of L. pneumophila-infected mice, and depletion of pDCs resulted in increased bacterial load. The ability of pDCs to combat infection did not require type I IFN. This study points to an unappreciated role for pDCs in combating bacterial infections and indicates a novel mechanism of action for this cell type.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Interferon Tipo I/fisiologia , Legionella pneumophila/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Animais , Células Dendríticas/metabolismo , Interferon Tipo I/deficiência , Interferon Tipo I/genética , Legionella pneumophila/crescimento & desenvolvimento , Pulmão/citologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética
17.
Biol Cell ; 103(12): 559-72, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21899517

RESUMO

BACKGROUND INFORMATION: Acid-secreting gastric parietal cells are polarized epithelial cells that harbour highly abundant and specialized, H+,K+ ATPase-containing, tubulovesicular membranes in the apical cytoplasm. The Golgi apparatus has been implicated in the biogenesis of the tubulovesicular membranes; however, an unanswered question is how a typical Golgi organization could regulate normal membrane transport within the membrane-dense cytoplasm of parietal cells. RESULTS: Here, we demonstrate that the Golgi apparatus of parietal cells is not the typical juxta-nuclear ribbon of stacks, but rather individual Golgi units are scattered throughout the cytoplasm. The Golgi membrane structures labelled with markers of both cis- and trans-Golgi membrane, indicating the presence of intact Golgi stacks. The parietal cell Golgi stacks were closely aligned with the microtubule network and were shown to participate in both anterograde and retrograde transport pathways. Dispersed Golgi stacks were also observed in parietal cells from H+,K+ ATPase-deficient mice that lack tubulovesicular membranes. CONCLUSIONS: These results indicate that the unusual organization of individual Golgi stacks dispersed throughout the cytoplasm of these terminally differentiated cells is likely to be a developmentally regulated event.


Assuntos
Citoplasma/metabolismo , Complexo de Golgi/química , Complexo de Golgi/metabolismo , Membranas Intracelulares/metabolismo , Células Parietais Gástricas/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Células Cultivadas , Citoplasma/química , Humanos , Membranas Intracelulares/química , Camundongos , Camundongos Endogâmicos BALB C , Células Parietais Gástricas/química , Proteínas/metabolismo
18.
Clin Microbiol Rev ; 23(2): 274-98, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20375353

RESUMO

The genus Legionella contains more than 50 species, of which at least 24 have been associated with human infection. The best-characterized member of the genus, Legionella pneumophila, is the major causative agent of Legionnaires' disease, a severe form of acute pneumonia. L. pneumophila is an intracellular pathogen, and as part of its pathogenesis, the bacteria avoid phagolysosome fusion and replicate within alveolar macrophages and epithelial cells in a vacuole that exhibits many characteristics of the endoplasmic reticulum (ER). The formation of the unusual L. pneumophila vacuole is a feature of its interaction with the host, yet the mechanisms by which the bacteria avoid classical endosome fusion and recruit markers of the ER are incompletely understood. Here we review the factors that contribute to the ability of L. pneumophila to infect and replicate in human cells and amoebae with an emphasis on proteins that are secreted by the bacteria into the Legionella vacuole and/or the host cell. Many of these factors undermine eukaryotic trafficking and signaling pathways by acting as functional and, in some cases, structural mimics of eukaryotic proteins. We discuss the consequences of this mimicry for the biology of the infected cell and also for immune responses to L. pneumophila infection.


Assuntos
Legionella pneumophila/fisiologia , Doença dos Legionários/microbiologia , Animais , Genoma Bacteriano , Humanos , Legionella pneumophila/genética , Legionella pneumophila/imunologia , Legionella pneumophila/patogenicidade , Doença dos Legionários/imunologia , Camundongos , Virulência
19.
Biochim Biophys Acta ; 1800(9): 906-11, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20594946

RESUMO

BACKGROUND: The H,K-ATPase, consisting of α and ß subunits, belongs to the P-type ATPase family. There are two isoforms of the α subunit, HKα1 and HKα2 encoded by different genes. The ouabain-resistant gastric HKα1-H,K-ATPase is Sch28080-sensitive. However, the colonic HKα2-H,K-ATPase from different species shows poor primary structure conservation of the HKα2 subunit between species and diverse pharmacological sensitivity to ouabain and Sch28080. This study sought to determine the contribution of each gene to functional activity and its pharmacological profile using mouse models with targeted disruption of HKα1, HKα2, or HKbeta genes. METHODS: Membrane vesicles from gastric mucosa and distal colon in wild-type (WT), HKα1, HKα2, or HKß knockout (KO) mice were extracted. K-ATPase activity and pharmacological profiles were examined. RESULTS: The colonic H,K-ATPase demonstrated slightly greater affinity for K(+) than the gastric H,K-ATPase. This K-ATPase activity was not detected in the colon of HKα2 KO but was observed in HKß KO with properties indistinguishable from WT. Neither ouabain nor Sch28080 had a significant effect on the WT colonic K-ATPase activity, but orthovanadate abolished this activity. Amiloride and its analogs benzamil and 5-N-ethyl-N-isopropylamiloride inhibited K-ATPase activity of HKα1-containing H,K-ATPase; the dose dependence of inhibition was similar for all three inhibitors. In contrast, the colonic HKα2-H,K-ATPase was not inhibited by these compounds. CONCLUSIONS: These data demonstrate that the mouse colonic H,K-ATPase exhibits a ouabain- and Sch28080-insensitive, orthovanadate-sensitive K-ATPase activity. Interestingly, pharmacological studies suggested that the mouse gastric H,K-ATPase is sensitive to amiloride. GENERAL SIGNIFICANCE: Characterization of the pharmacological profiles of the H,K-ATPases is important for understanding the relevant knockout animals and for considering the specificity of the inhibitors.


Assuntos
Colo/enzimologia , Resistência a Medicamentos/fisiologia , Inibidores Enzimáticos/farmacologia , Mucosa Gástrica/enzimologia , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Inibidores da Bomba de Prótons , Amilorida/farmacologia , Animais , Domínio Catalítico/genética , Resistência a Medicamentos/efeitos dos fármacos , ATPase Trocadora de Hidrogênio-Potássio/genética , Imidazóis/farmacologia , Camundongos , Camundongos Knockout , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/fisiologia , Ouabaína/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Vanadatos/farmacologia
20.
Artigo em Inglês | MEDLINE | ID: mdl-20947703

RESUMO

Mutagenesis of mice with N-ethyl-N-nitrosourea (ENU) is a phenotype-driven approach to unravel gene function and discover new biological pathways. Phenotype-driven approaches have the advantage of making no assumptions about the function of genes and their products and have been successfully applied to the discovery of novel gene-phenotype relationships in many physiological systems. ENU mutagenesis of mice is used in many large-scale and more focused projects to generate and identify novel mouse models for the study of gene functions and human disease. This review examines the strategies and tools used in ENU mutagenesis screens to efficiently generate and identify functional mutations.


Assuntos
Etilnitrosoureia , Genes/fisiologia , Genoma/efeitos dos fármacos , Mutagênese , Animais , Cruzamento/métodos , Modelos Animais de Doenças , Etilnitrosoureia/efeitos adversos , Feminino , Estudo de Associação Genômica Ampla , Doenças Inflamatórias Intestinais/genética , Masculino , Camundongos , Fenótipo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa