Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Immunity ; 39(5): 874-84, 2013 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-24184056

RESUMO

Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.


Assuntos
Antraciclinas/farmacologia , Antibacterianos/farmacologia , Reparo do DNA/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Peritonite/tratamento farmacológico , Sepse/prevenção & controle , Infecções por Adenoviridae/imunologia , Animais , Antraciclinas/uso terapêutico , Antibacterianos/uso terapêutico , Proteínas Mutadas de Ataxia Telangiectasia/deficiência , Proteínas Mutadas de Ataxia Telangiectasia/fisiologia , Proteína 7 Relacionada à Autofagia , Ceco/lesões , Dano ao DNA , Epirubicina/administração & dosagem , Epirubicina/farmacologia , Epirubicina/uso terapêutico , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/fisiologia , Inflamação , Mediadores da Inflamação/análise , Injeções Intraperitoneais , Pulmão/metabolismo , Meropeném , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/fisiologia , Especificidade de Órgãos , Peritonite/etiologia , Peritonite/genética , Peritonite/imunologia , Peritonite/fisiopatologia , Infecções Respiratórias/imunologia , Choque Séptico/prevenção & controle , Tienamicinas/uso terapêutico , Irradiação Corporal Total
2.
Environ Microbiol ; 18(10): 3390-3402, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26743546

RESUMO

Pseudomonas aeruginosa produces increased levels of alginate in response to oxygen-deprived conditions. The regulatory pathway(s) that links oxygen limitation to increased synthesis of alginate has remained elusive. In the present study, using immunofluorescence microscopy, we show that anaerobiosis-induced alginate production by planktonic PAO1 requires the diguanylate cyclase (DGC) SadC, previously identified as a regulator of surface-associated lifestyles. Furthermore, we found that the gene products of PA4330 and PA4331, located in a predicted operon with sadC, have a major impact on alginate production: deletion of PA4330 (odaA, for oxygen-dependent alginate synthesis activator) caused an alginate production defect under anaerobic conditions, whereas a PA4331 (odaI, for oxygen-dependent alginate synthesis inhibitor) deletion mutant produced alginate also in the presence of oxygen, which would normally inhibit alginate synthesis. Based on their sequence, OdaA and OdaI have predicted hydratase and dioxygenase reductase activities, respectively. Enzymatic assays using purified protein showed that unlike OdaA, which did not significantly affect DGC activity of SadC, OdaI inhibited c-di-GMP production by SadC. Our data indicate that SadC, OdaA and OdaI are components of a novel response pathway of P. aeruginosa that regulates alginate synthesis in an oxygen-dependent manner.


Assuntos
Proteínas de Bactérias/metabolismo , GMP Cíclico/análogos & derivados , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Oxigênio/metabolismo , Fósforo-Oxigênio Liases/metabolismo , Pseudomonas aeruginosa/metabolismo , Alginatos , Proteínas de Bactérias/genética , GMP Cíclico/metabolismo , Proteínas de Escherichia coli/genética , Ácido Glucurônico/biossíntese , Ácidos Hexurônicos , Óperon , Fósforo-Oxigênio Liases/genética , Pseudomonas aeruginosa/genética
3.
Environ Microbiol ; 17(11): 4379-93, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25845292

RESUMO

Pseudomonas aeruginosa is a multi-host opportunistic pathogen causing a wide range of diseases because of the armoury of virulence factors it produces, and it is difficult to eradicate because of its intrinsic resistance to antibiotics. Using an integrated whole-genome approach, we searched for P. aeruginosa virulence genes with multi-host relevance. We constructed a random library of 57 360 Tn5 mutants in P. aeruginosa PAO1-L and screened it in vitro for those showing pleiotropic effects in virulence phenotypes (reduced swarming, exo-protease and pyocyanin production). A set of these pleiotropic mutants were assayed for reduced toxicity in Drosophila melanogaster, Caenorhabditis elegans, human cell lines and mice. Surprisingly, the screening revealed that the virulence of the majority of P. aeruginosa mutants varied between disease models, suggesting that virulence is dependent on the disease model used and hence the host environment. Genomic analysis revealed that these virulence-related genes encoded proteins from almost all functional classes, which were conserved among P. aeruginosa strains. Thus, we provide strong evidence that although P. aeruginosa is capable of infecting a wide range of hosts, many of its virulence determinants are host specific. These findings have important implication when searching for novel anti-virulence targets to develop new treatments against P. aeruginosa.


Assuntos
Caenorhabditis elegans/microbiologia , Drosophila melanogaster/microbiologia , Especificidade de Hospedeiro/genética , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Fatores de Virulência/genética , Animais , Antibacterianos/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Biblioteca Gênica , Genômica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Virulência/genética
4.
J Immunol ; 190(3): 1276-84, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23277486

RESUMO

Pseudomonas aeruginosa persists in patients with cystic fibrosis (CF) and drives CF lung disease progression. P. aeruginosa potently activates the innate immune system, mainly mediated through pathogen-associated molecular patterns, such as flagellin. However, the host is unable to eradicate this flagellated bacterium efficiently. The underlying immunological mechanisms are incompletely understood. Myeloid-derived suppressor cells (MDSCs) are innate immune cells generated in cancer and proinflammatory microenvironments and are capable of suppressing T cell responses. We hypothesized that P. aeruginosa induces MDSCs to escape T cell immunity. In this article, we demonstrate that granulocytic MDSCs accumulate in CF patients chronically infected with P. aeruginosa and correlate with CF lung disease activity. Flagellated P. aeruginosa culture supernatants induced the generation of MDSCs, an effect that was 1) dose-dependently mimicked by purified flagellin protein, 2) significantly reduced using flagellin-deficient P. aeruginosa bacteria, and 3) corresponded to TLR5 expression on MDSCs in vitro and in vivo. Both purified flagellin and flagellated P. aeruginosa induced an MDSC phenotype distinct from that of the previously described MDSC-inducing cytokine GM-CSF, characterized by an upregulation of the chemokine receptor CXCR4 on the surface of MDSCs. Functionally, P. aeruginosa-infected CF patient ex vivo-isolated as well as flagellin or P. aeruginosa in vitro-generated MDSCs efficiently suppressed polyclonal T cell proliferation in a dose-dependent manner and modulated Th17 responses. These studies demonstrate that flagellin induces the generation of MDSCs and suggest that P. aeruginosa uses this mechanism to undermine T cell-mediated host defense in CF and other P. aeruginosa-associated chronic lung diseases.


Assuntos
Fibrose Cística/complicações , Flagelina/imunologia , Evasão da Resposta Imune/imunologia , Tolerância Imunológica/imunologia , Células Mieloides/imunologia , Pneumonia Bacteriana/imunologia , Infecções por Pseudomonas/imunologia , Pseudomonas aeruginosa/patogenicidade , Adolescente , Adulto , Proteínas de Bactérias/genética , Células Cultivadas/imunologia , Meios de Cultivo Condicionados/farmacologia , Fibrose Cística/microbiologia , Suscetibilidade a Doenças , Feminino , Flagelos/imunologia , Flagelos/fisiologia , Flagelina/genética , Flagelina/farmacologia , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Inata , Pulmão/microbiologia , Masculino , Células Mieloides/efeitos dos fármacos , Mielopoese/imunologia , Pneumonia Bacteriana/etiologia , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/etiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/isolamento & purificação , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Receptores CXCR4/imunologia , Subpopulações de Linfócitos T/imunologia , Células Th17/imunologia , Receptor 5 Toll-Like/imunologia , Regulação para Cima/imunologia , Adulto Jovem
5.
Mediators Inflamm ; 2015: 545417, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26185363

RESUMO

PURPOSE: Neutrophil-dominated airway inflammation is a key feature of progressive lung damage in cystic fibrosis (CF). Thus, reducing airway inflammation is a major goal to prevent lung damage in CF. However, current anti-inflammatory drugs have shown several limits. PI3Kγ plays a pivotal role in leukocyte recruitment and activation; in the present study we determined the effects of genetic deletion and pharmacologic inhibition of PI3Kγ on airway inflammation and structural lung damage in a mouse model of CF lung disease. METHODS: ßENaC overexpressing mice (ßENaC-Tg) were backcrossed with PI3Kγ-deficient (PI3Kγ (KO)) mice. Tissue damage was assessed by histology and morphometry and inflammatory cell number was evaluated in bronchoalveolar lavage fluid (BALF). Furthermore, we assessed the effect of a specific PI3Kγ inhibitor (AS-605240) on inflammatory cell number in BALF. RESULTS: Genetic deletion of PI3Kγ decreased neutrophil numbers in BALF of PI3Kγ (KO)/ßENaC-Tg mice, and this was associated with reduced emphysematous changes. Treatment with the PI3Kγ inhibitor AS-605240 decreased the number of neutrophils in BALF of ßENaC-Tg mice, reproducing the effect observed with genetic deletion of the enzyme. CONCLUSIONS: These results demonstrate the biological efficacy of both genetic deletion and pharmacological inhibition of PI3Kγ in reducing chronic neutrophilic inflammation in CF-like lung disease in vivo.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/fisiologia , Fibrose Cística/terapia , Inflamação/prevenção & controle , Pulmão/patologia , Infiltração de Neutrófilos , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Fibrose Cística/complicações , Fibrose Cística/patologia , Canais Epiteliais de Sódio/fisiologia , Deleção de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores de Fosfoinositídeo-3 Quinase
6.
Cell Physiol Biochem ; 34(1): 56-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24977481

RESUMO

BACKGROUND/AIMS: Inflammation is a major and critical component of the lung pathology in the hereditary disease cystic fibrosis. The molecular mechanisms of chronic inflammation in cystic fibrosis require definition. METHODS: We used several genetic mouse models to test a role of iNKT cells and ceramide in pulmonary inflammation of cystic fibrosis mice. Inflammation was determined by the pulmonary cytokine profil and the abundance of inflammatory cells in the lung. RESULTS: Here we provide a new concept how inflammation in the lung of individuals with cystic fibrosis is initiated. We show that in cystic fibrosis mice the mutation in the Cftr gene provokes a significant up-regulation of iNKT cells in the lung. Accumulation of iNKT cells serves to control autoimmune disease, which is triggered by a ceramide-mediated induction of cell death in CF organs. Autoimmunity becomes in particular overt in cystic fibrosis mice lacking iNKT cells and although suppression of the autoimmune response by iNKT cells is beneficial, IL-17(+) iNKT cells attract macrophages and neutrophils to CF lungs resulting in chronic inflammation. Genetic deletion of iNKT cells in cystic fibrosis mice prevents inflammation in CF lungs. CONCLUSION: Our data demonstrate an important function of iNKT cells in the chronic inflammation affecting cystic fibrosis lungs. iNKT cells suppress the auto-immune response induced by ceramide-mediated death of epithelial cells in CF lungs, but also induce a chronic pulmonary inflammation.


Assuntos
Células Matadoras Naturais/imunologia , Animais , Autoanticorpos/metabolismo , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/metabolismo , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Modelos Animais de Doenças , Interleucina-17/metabolismo , Células Matadoras Naturais/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CFTR , Pneumonia/metabolismo , Pneumonia/patologia
7.
Am J Respir Crit Care Med ; 187(6): 621-9, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23306544

RESUMO

RATIONALE: Cystic fibrosis (CF) is characterized by progressive pulmonary inflammation that is infection-triggered. Pseudomonas aeruginosa represents a risk factor for deterioration of lung function and reduced life expectancy. OBJECTIVES: To assess T-cell cytokine/chemokine production in clinically stable children with CF and evaluate the association between T-cell subtypes and susceptibility for infection with P. aeruginosa. METHODS: T-cell cytokine/chemokine profiles were measured in bronchoalveolar lavage fluid (BALF) from children with CF (n = 57; 6.1 ± 5.9 yr) and non-CF control subjects (n = 18; 5.9 ± 4.3 yr). Memory responses to Aspergillus fumigatus and P. aeruginosa were monitored. High-resolution computed tomography-based Helbich score was assessed. In a prospective observational trial the association between BALF cytokine/chemokine profiles and subsequent infection with P. aeruginosa was studied. MEASUREMENTS AND MAIN RESULTS: Th1- (INF-γ), Th2- (IL-5, IL-13), Th17- (IL-17A), and Th17-related cytokines (IL-1ß, IL-6) were significantly up-regulated in airways of patients with CF. IL-17A, IL-13, and IL-5 were significantly higher in BALF of symptomatic as compared with clinically asymptomatic patients with CF. IL-17A and IL-5 correlated with the percentage of neutrophils in BALF (r = 0.41, P < 0.05 and r = 0.46, P < 0.05, respectively). Th17- (IL-17A, IL-6, IL-1ß, IL-8) and Th2-associated cytokines and chemokines (IL-5, IL-13, TARC/CCL17), but not IFN-γ levels, significantly correlated with high-resolution computed tomography changes (Helbich score; P < 0.05). P. aeruginosa- and A. fumigatus-specific T cells from patients with CF displayed significantly higher IL-5 and IL-17A mRNA expression. IL-17A and TARC/CCL17 were significantly augmented in patients that developed P. aeruginosa infection within 24 months. CONCLUSIONS: We propose a role for Th17 and Th2 T cells in chronic inflammation in lungs of patients with CF. High concentrations of these cytokines/chemokines in CF airways precede infection with P. aeruginosa.


Assuntos
Fibrose Cística/metabolismo , Citocinas/metabolismo , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Células Th17/metabolismo , Células Th2/metabolismo , Adolescente , Líquido da Lavagem Broncoalveolar , Criança , Pré-Escolar , Feminino , Humanos , Interferon gama/metabolismo , Masculino , Prognóstico , Estudos Prospectivos
8.
Am J Respir Cell Mol Biol ; 48(1): 71-7, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23043087

RESUMO

A genome-wide association study identified interferon-related development regulator-1 (IFRD1), a protein expressed by neutrophils, as a key modifier gene in cystic fibrosis (CF) lung disease. Here, we investigated the expression and regulation of IFRD1 in CF neutrophils. IFRD1 expression was quantified in peripheral blood and airway neutrophils from patients with CF, patients with non-CF lung disease, and healthy control subjects. The regulation of IFRD1 expression was analyzed using isolated neutrophils and ex vivo stimulation assays with CF airway fluids. IFRD1 single-nucleotide polymorphisms (SNPs) were analyzed in a CF cohort (n = 572) and correlated with longitudinal lung function and IFRD1 expression. Patients with CF expressed higher protein levels of IFRD1 in peripheral blood neutrophils compared with healthy or non-CF disease control subjects. Within patients with CF, IFRD1 protein expression levels in neutrophils were lower in airway fluids compared with peripheral blood. High IFRD1 expression was positively associated with the production of reactive oxygen species (ROS) in CF neutrophils. In vitro regulation studies showed that CF airway fluid and the CF-characteristic chemokines CXCL8 and CXCL2 down-regulated IFRD1 expression in neutrophils, an effect that was mediated through CXCR2. Genetic analyses showed that three IFRD1 SNPs were associated with longitudinal declines in lung function, and modulated IFRD1 expression. These studies demonstrate that IFRD1 expression is systemically up-regulated in human CF neutrophils, is linked to the production of ROS, and is modulated by chemokines in CF airway fluids, depending on the IFRD1 genotype. Understanding the regulation of IFRD1 may pave the way for novel therapeutic approaches to target neutrophilic inflammation in CF.


Assuntos
Fibrose Cística/genética , Fibrose Cística/fisiopatologia , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Estudos de Casos e Controles , Quimiocina CXCL2/metabolismo , Estudos de Coortes , Fibrose Cística/imunologia , Humanos , Imunidade Inata , Interleucina-8/metabolismo , Pulmão/imunologia , Pulmão/fisiopatologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Polimorfismo de Nucleotídeo Único , Espécies Reativas de Oxigênio/metabolismo
9.
Thorax ; 68(4): 344-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23135343

RESUMO

PURPOSE: To assess efficacy and safety of a new dry powder formulation of inhaled colistimethate sodium in patients with cystic fibrosis (CF) aged ≥6 years with chronic Pseudomonas aeruginosa lung infection. STUDY DESIGN AND METHODS: A prospective, centrally randomised, phase III, open-label study in patients with stable CF aged ≥6 years with chronic P aeruginosa lung infection. Patients were randomised to Colobreathe dry powder for inhalation (CDPI, one capsule containing colistimethate sodium 1 662 500 IU, twice daily) or three 28-day cycles with twice-daily 300 mg/5 ml tobramycin inhaler solution (TIS). Study duration was 24 weeks. RESULTS: 380 patients were randomised. After logarithmic transformation of data due to a non-normal distribution, adjusted mean difference between treatment groups (CDPI vs TIS) in change in forced expiratory volume in 1 s (FEV1% predicted) at week 24 was -0.98% (95% CI -2.74% to 0.86%) in the intention-to-treat population (n=373) and -0.56% (95% CI -2.71% to 1.70%) in the per protocol population (n=261). The proportion of colistin-resistant isolates in both groups was ≤1.1%. The number of adverse events was similar in both groups. Significantly more patients receiving CDPI rated their device as 'very easy or easy to use' (90.7% vs 53.9% respectively; p<0.001). CONCLUSION: CDPI demonstrated efficacy by virtue of non-inferiority to TIS in lung function after 24 weeks of treatment. There was no emergence of resistance of P aeruginosa to colistin. Overall, CDPI was well tolerated. TRIAL REG NO: EudraCT 2004-003675-36.


Assuntos
Colistina/análogos & derivados , Fibrose Cística/tratamento farmacológico , Administração por Inalação , Adulto , Antibacterianos , Ensaios Clínicos Fase III como Assunto , Colistina/administração & dosagem , Feminino , Volume Expiratório Forçado , Humanos , Masculino , Pós , Estudos Prospectivos , Infecções por Pseudomonas/complicações , Pseudomonas aeruginosa , Tobramicina/administração & dosagem , Resultado do Tratamento , Adulto Jovem
10.
Cell Physiol Biochem ; 31(4-5): 505-12, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23572075

RESUMO

BACKGROUND/AIMS: Several recent studies revealed an accumulation of ceramide in bronchial, tracheal and intestinal epithelial cells of mice and patients with cystic fibrosis (CF). Normalization of ceramide concentrations in lungs of CF mice employing the functional acid sphingomyelinase inhibitor amitriptyline also normalized mucociliary clearance, chronic inflammation and infection susceptibility to pulmonary P. aeruginosa in these mice. METHODS: To test for a beneficial effect of amitriptyline in vivo, we performed a phase IIb randomised, double-blind, placebo-controlled study. Twenty-one CF patients were treated with 25 mg/d amitriptyline twice daily for 28 days. The placebo consisted of 19 patients and was also treated twice per day. The primary endpoint was the change in lung function in the intention-to-treat (ITT) population. Secondary endpoints were ceramide levels in epithelial cells and safety. RESULTS: After treatment, forced expiratory volume in 1 sec predicted (FEV1) increased 6.3 ± 11.5% (p=0.08) in the ITT population (36 of 40 CF patients) and 8.5 ± 10% (p=0.013) in the per protocol (PP) population (29 of 40 patients). Ceramide levels decreased in nasal epithelial cells after amitriptyline treatment. Amitriptyline had no severe and only mild and mostly transient adverse effects, i.e. xerostomia and tiredness. CONCLUSION: Amitriptyline is safe in CF-patients, increases FEV1 and reduces ceramide in lung cells of CF patients.


Assuntos
Inibidores da Captação Adrenérgica/uso terapêutico , Amitriptilina/uso terapêutico , Fibrose Cística/tratamento farmacológico , Adolescente , Adulto , Ceramidas/análise , Estudos de Coortes , Método Duplo-Cego , Feminino , Volume Expiratório Forçado , Humanos , Masculino , Efeito Placebo , Resultado do Tratamento , Adulto Jovem
11.
J Immunol ; 184(9): 5104-11, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20351190

RESUMO

We recently demonstrated that the accumulation of ceramide in Cftr-deficient epithelial cells is important for the pathophysiology of CF. However, the role of ceramide in other lung cells, particularly lung macrophages, requires definition. In this study, we report that ceramide is accumulated in Cftr-deficient lung macrophages. Alveolar macrophages contain a vesicle population, which is stained with LysoSensor probes but not by tetramethylrhodamine dextran. These vesicles, presumably secretory lysosomes, exhibit a higher pH in Cftr-deficient macrophages than the corresponding vesicles in lung macrophages isolated from wild-type (WT) mice. Alkalinization of these vesicles in Cftr-deficient macrophages correlates with a failure of the macrophages to respond to infection with various Pseudomonas aeruginosa strains by acutely activating acid sphingomyelinase, releasing ceramide, forming ceramide-enriched membrane platforms that serve to cluster gp91(phox), and, most importantly, releasing reactive oxygen species (ROS). In contrast, these events occur rapidly in WT lung macrophages postinfection. Inhibiting ROS in WT macrophages prevents the killing of P. aeruginosa. These findings provide evidence for a novel pH-controlled pathway from acid sphingomyelinase activation via ceramide and clustering of gp91(phox) to the release of ROS in lung macrophages.


Assuntos
Ceramidas/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/deficiência , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Ceramidas/química , Ceramidas/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Dextranos/química , Dextranos/metabolismo , Corantes Fluorescentes/química , Corantes Fluorescentes/metabolismo , Concentração de Íons de Hidrogênio , Macrófagos Alveolares/enzimologia , Macrófagos Alveolares/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CFTR , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/imunologia , Espécies Reativas de Oxigênio/química , Rodaminas/química , Rodaminas/metabolismo , Teste Bactericida do Soro , Esfingomielina Fosfodiesterase/metabolismo , Coloração e Rotulagem
12.
J Infect Dis ; 202(10): 1585-92, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20942647

RESUMO

BACKGROUND: Antibiotic combination therapy might be more efficient than single antibiotics to combat Pseudomonas aeruginosa biofilms in the airways of patients with cystic fibrosis. We tested the ability of colistin sulphate-tobramycin combinations and single antibiotics to kill P. aeruginosa biofilms. METHODS: P. aeruginosa biofilms were generated in vitro and in rat lungs. In a pilot study, 5 patients with cystic fibrosis inhaled colistin and then tobramycin for 4 weeks. The changes in P. aeruginosa counts and lung function were assessed before and after therapy. RESULTS: Antibiotic combination therapy significantly reduced the number of P. aeruginosa cells in P. aeruginosa biofilm models in vitro. When rats were challenged with 1 x 10(7) cfu of P. aeruginosa, which was embedded in alginate beads, mortality rates, lung pathologic findings, and bacterial colony-forming unit counts were significantly lower after 7 days in animals receiving antibiotic combination than in animals receiving single antibiotics. In patients with cystic fibrosis, inhaled colistin-tobramycin was well tolerated and resulted in a mean decrease of 2.52 + /- 2.5 log(10) cfu of P. aeruginosa per milliliter of sputum (P = .027). Measurements of forced expiratory volume in 1 s, obtained both before and after the study, did not differ significantly. CONCLUSION: Colistin-tobramycin combinations are more efficient than respective single antibiotics for killing P. aeruginosa in biofilms in vitro, and they significantly reduced P. aeruginosa cell counts in a rat lung infection model and in patients with cystic fibrosis.


Assuntos
Antibacterianos/uso terapêutico , Biofilmes/efeitos dos fármacos , Colistina/uso terapêutico , Fibrose Cística/tratamento farmacológico , Infecções por Pseudomonas/tratamento farmacológico , Pseudomonas aeruginosa/efeitos dos fármacos , Tobramicina/uso terapêutico , Administração por Inalação , Adulto , Animais , Antibacterianos/administração & dosagem , Colistina/administração & dosagem , Fibrose Cística/microbiologia , Quimioterapia Combinada , Feminino , Humanos , Pulmão/microbiologia , Masculino , Testes de Sensibilidade Microbiana , Projetos Piloto , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/fisiologia , Ratos , Ratos Endogâmicos Lew , Tobramicina/administração & dosagem , Resultado do Tratamento
13.
mBio ; 12(2)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33879591

RESUMO

The opportunistic pathogen Pseudomonas aeruginosa produces an arsenal of virulence factors causing a wide range of diseases in multiple hosts and is difficult to eradicate due to its intrinsic resistance to antibiotics. With the antibacterial pipeline drying up, antivirulence therapy has become an attractive alternative strategy to the traditional use of antibiotics to treat P. aeruginosa infections. To identify P. aeruginosa genes required for virulence in multiple hosts, a random library of Tn5 mutants in strain PAO1-L was previously screened in vitro for those showing pleiotropic effects in the production of virulence phenotypes. Using this strategy, we identified a Tn5 mutant with an insertion in PA4130 showing reduced levels of a number of virulence traits in vitro Construction of an isogenic mutant in this gene presented results similar to those for the Tn5 mutant. Furthermore, the PA4130 isogenic mutant showed substantial attenuation in disease models of Drosophila melanogaster and Caenorhabditis elegans as well as reduced toxicity in human cell lines. Mice infected with this mutant demonstrated an 80% increased survival rate in acute and agar bead lung infection models. PA4130 codes for a protein with homology to nitrite and sulfite reductases. Overexpression of PA4130 in the presence of the siroheme synthase CysG enabled its purification as a soluble protein. Methyl viologen oxidation assays with purified PA4130 showed that this enzyme is a nitrite reductase operating in a ferredoxin-dependent manner. The preference for nitrite and production of ammonium revealed that PA4130 is an ammonia:ferredoxin nitrite reductase and hence was named NirA.IMPORTANCE The emergence of widespread antimicrobial resistance has led to the need for development of novel therapeutic interventions. Antivirulence strategies are an attractive alternative to classic antimicrobial therapy; however, they require identification of new specific targets which can be exploited in drug discovery programs. The host-specific nature of P. aeruginosa virulence adds complexity to the discovery of these types of targets. Using a sequence of in vitro assays and phylogenetically diverse in vivo disease models, we have identified a PA4130 mutant with reduced production in a number of virulence traits and severe attenuation across all infection models tested. Characterization of PA4130 revealed that it is a ferredoxin-nitrite reductase and hence was named NirA. These results, together with attenuation of nirA mutants in different clinical isolates, high level conservation of its gene product in P. aeruginosa genomes, and the lack of orthologues in human genomes, make NirA an attractive antivirulence target.


Assuntos
Nitrito Redutases/genética , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Fatores de Virulência/genética , Amônia/metabolismo , Animais , Proteínas de Bactérias/metabolismo , Caenorhabditis elegans , Linhagem Celular , Modelos Animais de Doenças , Drosophila melanogaster , Ferredoxinas/metabolismo , Biblioteca Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Nitrito Redutases/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/patogenicidade
14.
Am J Respir Cell Mol Biol ; 42(6): 716-24, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19635928

RESUMO

Employing genetic mouse models we have recently shown that ceramide accumulation is critically involved in the pathogenesis of cystic fibrosis (CF) lung disease. Genetic or systemic inhibition of the acid sphingomyelinase (Asm) is not feasible for treatment of patients or might cause adverse effects. Thus, a manipulation of ceramide specifically in lungs of CF mice must be developed. We tested whether inhalation of different acid sphingomyelinase inhibitors does reduce Asm activity and ceramide accumulation in lungs of CF mice. The efficacy and specificity of the drugs was determined. Ceramide was determined by mass spectrometry, DAG-kinase assays, and fluorescence microscopy. We determined pulmonary and systemic Asm activity, neutral sphingomyelinase (Nsm), ceramide, cytokines, and infection susceptibility. Mass spectroscopy, DAG-kinase assays, and semiquantitative immune fluorescence microscopy revealed that a standard diet did not influence ceramide in bronchial respiratory epithelial cells, while a diet with Peptamen severely affected the concentration of sphingolipids in CF lungs. Inhalation of the Asm inhibitors amitriptyline, trimipramine, desipramine, chlorprothixene, fluoxetine, amlodipine, or sertraline restored normal ceramide concentrations in murine bronchial epithelial cells, reduced inflammation in the lung of CF mice and prevented infection with Pseudomonas aeruginosa. All drugs showed very similar efficacy. Inhalation of the drugs was without systemic effects and did not inhibit Nsm. These findings employing several structurally different Asm inhibitors identify Asm as primary target in the lung to reduce ceramide concentrations. Inhaling an Asm inhibitor may be a beneficial treatment for CF, with minimal adverse systemic effects.


Assuntos
Anti-Inflamatórios/farmacologia , Ceramidas/metabolismo , Fibrose Cística/tratamento farmacológico , Pulmão/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Pneumonia/tratamento farmacológico , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Administração por Inalação , Administração Oral , Envelhecimento , Animais , Anti-Inflamatórios/administração & dosagem , Fibrose Cística/complicações , Fibrose Cística/enzimologia , Fibrose Cística/imunologia , Citocinas/metabolismo , Diacilglicerol Quinase/metabolismo , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Pulmão/enzimologia , Pulmão/imunologia , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos CFTR , Microscopia de Fluorescência , Oligopeptídeos/administração & dosagem , Inibidores de Fosfodiesterase/administração & dosagem , Pneumonia/complicações , Pneumonia/enzimologia , Pneumonia/imunologia , Infecções por Pseudomonas/enzimologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/prevenção & controle , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/enzimologia , Mucosa Respiratória/imunologia , Infecções Respiratórias/enzimologia , Infecções Respiratórias/imunologia , Infecções Respiratórias/prevenção & controle , Esfingomielina Fosfodiesterase/metabolismo , Resultado do Tratamento
15.
Infect Immun ; 78(2): 746-55, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19995892

RESUMO

Pseudomonas aeruginosa is a serious pathogen in hospitalized, immunocompromised, and cystic fibrosis (CF) patients. P. aeruginosa is motile via a single polar flagellum made of polymerized flagellin proteins differentiated into two major serotypes: a and b. Antibodies to flagella delay onset of infection in CF patients, but whether immunity to polymeric flagella and that to monomeric flagellin are comparable has not been addressed, nor has the question of whether such antibodies might negatively impact Toll-like receptor 5 (TLR5) activation, an important component of innate immunity to P. aeruginosa. We compared immunization with flagella and that with flagellin for in vitro effects on motility, opsonic killing, and protective efficacy using a mouse pneumonia model. Antibodies to flagella were superior to antibodies to flagellin at inhibiting motility, promoting opsonic killing, and mediating protection against P. aeruginosa pneumonia in mice. Protection against the flagellar type strains PAK and PA01 was maximal, but it was only marginal against motile clinical isolates from flagellum-immunized CF patients who nonetheless became colonized with P. aeruginosa. Purified flagellin was a more potent activator of TLR5 than were flagella and also elicited higher TLR5-neutralizing antibodies than did immunization with flagella. Antibody to type a but not type b flagella or flagellin inhibited TLR5 activation by whole bacterial cells. Overall, intact flagella appear to be superior for generating immunity to P. aeruginosa, and flagellin monomers might induce antibodies capable of neutralizing innate immunity due to TLR5 activation, but solid immunity to P. aeruginosa based on flagellar antigens may require additional components beyond type a and type b proteins from prototype strains.


Assuntos
Flagelos/imunologia , Flagelina/imunologia , Infecções por Pseudomonas/prevenção & controle , Vacinas contra Pseudomonas/imunologia , Pseudomonas aeruginosa/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/imunologia , Ensaio de Imunoadsorção Enzimática , Camundongos , Camundongos Endogâmicos C57BL , Receptor 5 Toll-Like/imunologia , Receptor 5 Toll-Like/metabolismo
16.
Infect Immun ; 78(7): 3036-46, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20421387

RESUMO

Probiotics are viable microorganisms that are increasingly used for treatment of a variety of diseases. Occasionally, however, probiotics may have adverse clinical effects, including septicemia. Here we examined the role of the intestinal microbiota and the adaptive immune system in preventing translocation of probiotics (e.g., Escherichia coli Nissle). We challenged C57BL/6J mice raised under germfree conditions (GF-raised C57BL/6J mice) and Rag1(-/-) mice raised under germfree conditions (GF-raised Rag1(-/-) mice) and under specific-pathogen-free conditions (SPF-raised Rag1(-/-) mice) with probiotic E. coli strain Nissle 1917, strain Nissle 1917 mutants, the commensal strain E. coli mpk, or Bacteroides vulgatus mpk. Additionally, we reconstituted Rag1(-/-) mice with CD4(+) T cells. E. coli translocation and dissemination and the mortality of mice were assessed. In GF-raised Rag1(-/-) mice, but not in SPF-raised Rag1(-/-) mice or GF-raised C57BL/6J mice, oral challenge with E. coli strain Nissle 1917, but not oral challenge with E. coli mpk, resulted in translocation and dissemination. The mortality rate was significantly higher for E. coli strain Nissle 1917-challenged GF-raised Rag1(-/-) mice (100%; P < 0.001) than for E. coli strain Nissle 1917-challenged SPF-raised Rag1(-/-) mice (0%) and GF-raised C57BL/6J mice (0%). Translocation of and mortality due to strain E. coli Nissle 1917 in GF-raised Rag1(-/-) mice were prevented when mice were reconstituted with T cells prior to strain E. coli Nissle 1917 challenge, but not when mice were reconstituted with T cells after E. coli strain Nissle 1917 challenge. Cocolonization experiments revealed that E. coli mpk could not prevent translocation of strain E. coli Nissle 1917. Moreover, we demonstrated that neither lipopolysaccharide structure nor flagella play a role in E. coli strain Nissle 1917 translocation and dissemination. Our results suggest that if both the microbiota and adaptive immunity are defective, translocation across the intestinal epithelium and dissemination of the probiotic E. coli strain Nissle 1917 may occur and have potentially severe adverse effects. Future work should define the possibly related molecular factors that promote probiotic functions, fitness, and facultative pathogenicity.


Assuntos
Imunidade Adaptativa/imunologia , Escherichia coli/imunologia , Intestinos/microbiologia , Probióticos/efeitos adversos , Animais , Citocinas/sangue , Citocinas/imunologia , Escherichia coli/genética , Escherichia coli/patogenicidade , Genes Bacterianos/genética , Genes Bacterianos/imunologia , Genes RAG-1/imunologia , Metagenoma/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Reação em Cadeia da Polimerase , Linfócitos T/imunologia , Translocação Genética/imunologia
17.
Thorax ; 65(11): 978-84, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20880875

RESUMO

BACKGROUND: Patients with cystic fibrosis (CF) with Pseudomonas aeruginosa lung infections produce endobronchial mucus plugs allowing growth of obligate anaerobes including Prevotella spp. Whether obligate anaerobes contribute to the pathophysiology of CF lung disease is unknown. METHODS: The virulence of Prevotella intermedia and Ps aeruginosa was investigated in vitro and in mice, antibodies against P intermedia in CF sera were assessed and a culture-independent detection method for P intermedia/P nigrescens in CF sputum was tested. RESULTS: P intermedia reached cell numbers of >10(5)->10(7) colony-forming units (CFU)/ml sputum. The majority of patients with CF (16/17; 94.1%) produced antibodies against two immunoreactive antigens of P intermedia. Culture supernatant fluids, collected from 10(9) P intermedia cells, were more cytotoxic to respiratory epithelial cells in vitro and inflammatory in mouse lungs than respective fluids from anaerobically grown Ps aeruginosa, while fluids from aerobically grown Ps aeruginosa had the highest cytotoxicity and inflammation. Both pathological effects were largely reduced when culture supernatant fluids from 10(7) cells of either species were used. P intermedia cells (∼10(6)CFU/lung) did not induce mortality in the agar beads lung infection mouse model, while Ps aeruginosa cells caused death in 30% of mice due to rapid multiplication. A P intermedia/P nigrescens-specific PNA probe was significantly more sensitive than culture-dependent diagnostic assays to detect these strict anaerobes. CONCLUSION: Ps aeruginosa and P intermedia become significantly virulent in vitro and in vivo when cell numbers exceed 10(8) CFU/lung.


Assuntos
Infecções por Bacteroidaceae/complicações , Fibrose Cística/complicações , Prevotella intermedia/patogenicidade , Infecções por Pseudomonas/complicações , Pseudomonas aeruginosa/patogenicidade , Animais , Anticorpos Antibacterianos/sangue , Líquido da Lavagem Broncoalveolar/microbiologia , Células Cultivadas , Contagem de Colônia Microbiana , Humanos , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções Oportunistas/complicações , Prevotella intermedia/crescimento & desenvolvimento , Prevotella intermedia/imunologia , Prevotella intermedia/isolamento & purificação , Pseudomonas aeruginosa/crescimento & desenvolvimento , Pseudomonas aeruginosa/isolamento & purificação , Infecções Respiratórias/complicações , Escarro/microbiologia , Virulência
18.
Int J Med Microbiol ; 300(8): 573-7, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20940108

RESUMO

In patients with cystic fibrosis (CF) prevention of lung infections with Pseudomonas aeruginosa is of major importance. Principles to achieve this goal include vaccination, immediate use of antibiotics in patients newly colonized with the pathogen, and hygienic measures. The purpose of this review is to discuss recent developments in this context.


Assuntos
Broncopneumonia/prevenção & controle , Fibrose Cística/complicações , Pneumonia Bacteriana/prevenção & controle , Infecções por Pseudomonas/prevenção & controle , Antibacterianos/uso terapêutico , Vacinas Bacterianas/administração & dosagem , Humanos , Controle de Infecções/métodos , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/imunologia
19.
Cell Microbiol ; 11(2): 208-16, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19068098

RESUMO

Innate immunity is essential for prevention of infection in vertebrates and plants and dysfunction of single components of innate immunity may provoke severe disease. Here we describe how mutations in the cystic fibrosis transmembrane conductance regulator gene dysregulate a variety of components of the innate immune system in individuals suffering from the hereditary disease cystic fibrosis. In the airways of these individuals, functions of the mucociliary clearance system, cationic antimicrobial (poly)peptides and neutrophils and macrophages are impaired and inflammatory signal transduction pathways exaggerated. Consequently, chronic airway colonization with opportunistic bacterial pathogens develops and leads to life-threatening lung disease.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/imunologia , Fibrose Cística/patologia , Imunidade Inata , Mutação , Animais , Humanos
20.
Am J Respir Crit Care Med ; 180(2): 138-45, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19423715

RESUMO

RATIONALE: During long-term lung infection in patients with cystic fibrosis (CF), Pseudomonas aeruginosa strains develop mutations leading to clonal expansion. This microevolution is believed to be correlated with a reduced virulence. OBJECTIVES: We tested this hypothesis in models of lung infection, using mice with different genetic backgrounds. METHODS: From infected airways of six patients with CF, 25 P. aeruginosa clones were isolated during a period of up to 16.3 years and genotypically and phenotypically characterized. Virulence of the 8 early, 6 intermediate, and 11 late CF isolates and 5 environmental strains was assessed by monitoring acute mortality versus survival and P. aeruginosa chronic persistence versus lung clearance in mice of different genetic backgrounds, including CF mice. MEASUREMENTS AND MAIN RESULTS: Different patients harbored clonally unrelated strains, but early, intermediate, and late P. aeruginosa isolates from single patients were clonally related, allowing comparative in vivo analysis. Although late isolates were attenuated in causing acute mortality in the mouse models, compared with early and intermediate clonal isolates and environmental strains, they did not differ from early and intermediate clonal isolates in their capacity to establish chronic infection and cause extensive inflammation in the murine respiratory tract. CONCLUSIONS: Our findings indicate that clonal expansion of P. aeruginosa strains during microevolution within CF lungs leads to populations with altered but not reduced virulence. These P. aeruginosa clones with adapted virulence play a critical role in the pathogenesis of chronic infections and may serve to define virulence determinants as targets for novel therapies.


Assuntos
Fibrose Cística/microbiologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/patogenicidade , Infecções Respiratórias/microbiologia , Infecções Respiratórias/patologia , Animais , Doença Crônica , Células Clonais/fisiologia , Fibrose Cística/patologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CFTR , Pseudomonas aeruginosa/isolamento & purificação , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA