Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 434
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34203338

RESUMO

Diffuse alveolar hemorrhage (DAH) in systemic lupus erythematosus (SLE) is associated with significant mortality, requiring a thorough understanding of its complex mechanisms to develop novel therapeutics for disease control. Activated p53-dependent apoptosis with dysregulated long non-coding RNA (lncRNA) expression is involved in the SLE pathogenesis and correlated with clinical activity. We examined the expression of apoptosis-related p53-dependent lncRNA, including H19, HOTAIR and lincRNA-p21 in SLE-associated DAH patients. Increased lincRNA-p21 levels were detected in circulating mononuclear cells, mainly in CD4+ and CD14+ cells. Higher expression of p53, lincRNA-p21 and cell apoptosis was identified in lung tissues. Lentivirus-based short hairpin RNA (shRNA)-transduced stable transfectants were created for examining the targeting efficacy in lncRNA. Under pristane stimulation, alveolar epithelial cells had increased p53, lincRNA-p21 and downstream Bax levels with elevated apoptotic ratios. After pristane injection, C57/BL6 mice developed DAH with increased pulmonary expression of p53, lincRNA-p21 and cell apoptosis. Intra-pulmonary delivery of shRNA targeting lincRNA-p21 reduced hemorrhage frequencies and improved anemia status through decreasing Bax expression and cell apoptosis. Our findings demonstrate increased p53-dependent lncRNA expression with accelerated cell apoptosis in the lungs of SLE-associated DAH patients, and show the therapeutic potential of targeting intra-pulmonary lncRNA expression in a pristane-induced model of DAH.


Assuntos
RNA Longo não Codificante/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/fisiologia , Modelos Animais de Doenças , Feminino , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pneumopatias/genética , Pneumopatias/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Masculino , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Supressora de Tumor p53/genética
2.
Am J Physiol Lung Cell Mol Physiol ; 319(2): L218-L227, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32519893

RESUMO

Few patients with bacteremia from a nonpulmonary source develop acute respiratory distress syndrome (ARDS). However, the mechanisms that protect the lung from injury in bacteremia have not been identified. We simulated bacteremia by adding Streptococcus pneumoniae to the perfusate of the ex vivo perfused human lung model. In contrast to a pneumonia model in which bacteria were instilled into the distal air spaces of one lobe, injection of high doses of S. pneumoniae into the perfusate was not associated with alveolar epithelial injury as demonstrated by low protein permeability of the alveolar epithelium, intact alveolar fluid clearance, and the absence of alveolar edema. Unexpectedly, the ex vivo human lung rapidly cleared large quantities of S. pneumoniae even though the perfusate had very few intravascular phagocytes and lacked immunoglobulins or complement. The bacteria were cleared in part by the small number of neutrophils in the perfusate, alveolar macrophages in the airspaces, and probably by interstitial pathways. Together, these findings identify one mechanism by which the lung and the alveolar epithelium are protected from injury in bacteremia.


Assuntos
Lesão Pulmonar Aguda/microbiologia , Lesão Pulmonar Aguda/patologia , Bacteriemia/patologia , Pulmão/patologia , Streptococcus pneumoniae/patogenicidade , Adulto , Bacteriemia/microbiologia , Epitélio/microbiologia , Epitélio/patologia , Feminino , Humanos , Pulmão/microbiologia , Macrófagos/microbiologia , Macrófagos/patologia , Masculino , Pessoa de Meia-Idade , Neutrófilos/microbiologia , Neutrófilos/patologia , Permeabilidade , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/patologia , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Síndrome do Desconforto Respiratório/microbiologia , Síndrome do Desconforto Respiratório/patologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/parasitologia
3.
EMBO Rep ; 19(1): 29-42, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29141986

RESUMO

The interaction of Mycobacterium tuberculosis (Mtb) with pulmonary epithelial cells is critical for early stages of bacillus colonization and during the progression of tuberculosis. Entry of Mtb into epithelial cells has been shown to depend on F-actin polymerization, though the molecular mechanisms are still unclear. Here, we demonstrate that mycobacterial uptake into epithelial cells requires rearrangements of the actin cytoskeleton, which are regulated by ADP-ribosylation factor 1 (Arf1) and phospholipase D1 (PLD1), and is dependent on the M3 muscarinic receptor (M3R). We show that this pathway is controlled by Arf GTPase-activating protein 1 (ArfGAP1), as its silencing has an impact on actin cytoskeleton reorganization leading to uncontrolled uptake and replication of Mtb. Furthermore, we provide evidence that this pathway is critical for mycobacterial entry, while the cellular infection with other pathogens, such as Shigella flexneri and Yersinia pseudotuberculosis, is not affected. Altogether, these results reveal how cortical actin plays the role of a barrier to prevent mycobacterial entry into epithelial cells and indicate a novel role for ArfGAP1 as a restriction factor of host-pathogen interactions.


Assuntos
Citoesqueleto de Actina/metabolismo , Actinas/genética , Proteínas Ativadoras de GTPase/genética , Interações Hospedeiro-Patógeno , Mycobacterium tuberculosis/patogenicidade , Alvéolos Pulmonares/metabolismo , Células A549 , Fator 1 de Ribosilação do ADP/genética , Fator 1 de Ribosilação do ADP/metabolismo , Citoesqueleto de Actina/microbiologia , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Mycobacterium tuberculosis/fisiologia , Fosfolipase D/genética , Fosfolipase D/metabolismo , Polimerização , Alvéolos Pulmonares/microbiologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor Muscarínico M3/genética , Receptor Muscarínico M3/metabolismo , Shigella flexneri/fisiologia , Transdução de Sinais , Especificidade da Espécie , Yersinia pseudotuberculosis/fisiologia
4.
Med Mycol ; 58(3): 333-340, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-31309220

RESUMO

Cladosporium is one of the most abundant spore. Fungi of this genus can cause respiratory allergy and intrabronchial lesion. We studied the differential expression of host genes after the interaction of Cladosporium sphaerospermum conidia with Human Bronchial Epithelial Cells (BEAS-2B) and Human Pulmonary Alveolar Epithelial Cells (HPAEpiC). C. sphaerospermum conidia were harvested and co-cultured with BEAS-2B cells or HPAEpiC cells for 48 hours respectively. This culture duration was chosen as it was associated with high germination rate. RNA was extracted from two biological replicates per treatment. RNA of BEAS-2B cells was used to assess changes in gene expression using AffymetrixGeneChip® Human Transcriptome Array 2.0. After co-culture with Cladosporium spores, 68 individual genes were found differentially expressed (P ≤ 0.05) and up-regulated ≥ 1.5 folds while 75 genes were found differentially expressed at ≤ -1.5 folds compared with controls. Reverse transcription and qPCR were performed on the RNA collected from both BEAS-2B cells and HPAEpiC cells to validate the microarray results with 7 genes. Based on the findings, infected pulmonary epithelial cells exhibited an increase in cell death-related genes and genes associated with innate immunity.


Assuntos
Células Epiteliais Alveolares/microbiologia , Cladosporium/patogenicidade , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , Alvéolos Pulmonares/microbiologia , Brônquios/citologia , Brônquios/microbiologia , Linhagem Celular , Humanos , Análise em Microsséries , Alvéolos Pulmonares/citologia , Regulação para Cima
5.
Am J Respir Crit Care Med ; 199(9): 1127-1138, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30789747

RESUMO

Rationale: Idiopathic pulmonary fibrosis (IPF) causes considerable global morbidity and mortality, and its mechanisms of disease progression are poorly understood. Recent observational studies have reported associations between lung dysbiosis, mortality, and altered host defense gene expression, supporting a role for lung microbiota in IPF. However, the causal significance of altered lung microbiota in disease progression is undetermined. Objectives: To examine the effect of microbiota on local alveolar inflammation and disease progression using both animal models and human subjects with IPF. Methods: For human studies, we characterized lung microbiota in BAL fluid from 68 patients with IPF. For animal modeling, we used a murine model of pulmonary fibrosis in conventional and germ-free mice. Lung bacteria were characterized using 16S rRNA gene sequencing with novel techniques optimized for low-biomass sample load. Microbiota were correlated with alveolar inflammation, measures of pulmonary fibrosis, and disease progression. Measurements and Main Results: Disruption of the lung microbiome predicts disease progression, correlates with local host inflammation, and participates in disease progression. In patients with IPF, lung bacterial burden predicts fibrosis progression, and microbiota diversity and composition correlate with increased alveolar profibrotic cytokines. In murine models of fibrosis, lung dysbiosis precedes peak lung injury and is persistent. In germ-free animals, the absence of a microbiome protects against mortality. Conclusions: Our results demonstrate that lung microbiota contribute to the progression of IPF. We provide biological plausibility for the hypothesis that lung dysbiosis promotes alveolar inflammation and aberrant repair. Manipulation of lung microbiota may represent a novel target for the treatment of IPF.


Assuntos
Fibrose Pulmonar Idiopática/microbiologia , Inflamação/microbiologia , Pulmão/microbiologia , Microbiota/fisiologia , Idoso , Animais , Líquido da Lavagem Broncoalveolar/microbiologia , Modelos Animais de Doenças , Progressão da Doença , Feminino , Citometria de Fluxo , Vida Livre de Germes , Humanos , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/genética , Pessoa de Meia-Idade , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , RNA Ribossômico 16S/genética
6.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-33182614

RESUMO

Bacterial pneumonia is one of the most prevalent infectious diseases and has high mortality in sensitive patients (children, elderly and immunocompromised). Although an infection, the disease alters the alveolar epithelium homeostasis and hinders normal breathing, often with fatal consequences. A special case is hospitalized aged patients, which present a high risk of infection and death because of the community acquired version of the Streptococcus pneumoniae pneumonia. There is evidence that early antibiotics treatment decreases the inflammatory response during pneumonia. Here, we investigate mechanistically this strategy using a multi-level mathematical model, which describes the 24 first hours after infection of a single alveolus from the key signaling networks behind activation of the epithelium to the dynamics of the local immune response. With the model, we simulated pneumonia in aged and young patients subjected to different antibiotics timing. The results show that providing antibiotics to elderly patients 8 h in advance compared to young patients restores in aged individuals the effective response seen in young ones. This result suggests the use of early, probably prophylactic, antibiotics treatment in aged hospitalized people with high risk of pneumonia.


Assuntos
Antibacterianos/administração & dosagem , Modelos Imunológicos , Infiltração de Neutrófilos , Pneumonia Pneumocócica/tratamento farmacológico , Pneumonia Pneumocócica/imunologia , Idoso , Envelhecimento/imunologia , Animais , Proteínas de Bactérias/biossíntese , Simulação por Computador , Citocinas/imunologia , Esquema de Medicação , Humanos , Conceitos Matemáticos , Camundongos , Pneumonia Pneumocócica/microbiologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Índice de Gravidade de Doença , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/biossíntese , Análise de Sistemas
7.
Thorax ; 74(1): 43-50, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30076187

RESUMO

BACKGROUND: We previously reported that microvesicles (MVs) released by human mesenchymal stem cells (MSC) were as effective as the cells themselves in both Escherichia coli lipopolysaccharide and live bacteria-induced acute lung injury (ALI) mice models. However, it remained unclear whether the biological effect of MSC MV can be applied to human ALI. METHODS: In the current study, we tested the therapeutic effects of MSC MVs in a well-established ex vivo perfused human model of bacterial pneumonia. Using human donor lungs not used for transplantation, we instilled E. coli bacteria intrabronchially and, 1 hour later, administered MSC MVs into the perfusate as therapy. RESULTS: After 6 hours, instillation of E. coli bacteria caused influx of inflammatory cells, which resulted in significant inflammation, lung protein permeability and pulmonary oedema formation. Administration of MSC MV significantly increased alveolar fluid clearance and reduced protein permeability and numerically lowered the bacterial load in the injured alveolus. The beneficial effect on bacterial killing was more pronounced with pretreatment of MSCs with a Toll-like receptor 3 agonist, polyinosinic:polycytidylic acid (Poly (I:C)), prior to the isolation of MVs. Isolated human alveolar macrophages had increased antimicrobial activity with MSC MV treatment in vitro as well. Although oxygenation and lung compliance levels were similar between injury and treatment groups, administration of MSC MVs numerically decreased median pulmonary artery pressure at 6 hours. CONCLUSIONS: In summary, MSC MVs increased alveolar fluid clearance and reduced lung protein permeability, and pretreatment with Poly (I:C) enhanced the antimicrobial activity of MVs in an ex vivo perfused human lung with severe bacteria pneumonia.


Assuntos
Lesão Pulmonar Aguda/fisiopatologia , Lesão Pulmonar Aguda/terapia , Terapia Baseada em Transplante de Células e Tecidos , Micropartículas Derivadas de Células , Infecções por Escherichia coli/complicações , Células-Tronco Mesenquimais , Pneumonia Bacteriana/complicações , Proteínas/metabolismo , Alvéolos Pulmonares/metabolismo , Lesão Pulmonar Aguda/microbiologia , Lesão Pulmonar Aguda/patologia , Adulto , Idoso , Pressão Arterial , Carga Bacteriana , Micropartículas Derivadas de Células/efeitos dos fármacos , Feminino , Humanos , Indutores de Interferon/farmacologia , Contagem de Leucócitos , Complacência Pulmonar , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Neutrófilos , Técnicas de Cultura de Órgãos , Oxigênio/metabolismo , Permeabilidade , Poli I-C/farmacologia , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Artéria Pulmonar , Edema Pulmonar/microbiologia , Edema Pulmonar/terapia , Receptor 3 Toll-Like/agonistas , Fator de Necrose Tumoral alfa/metabolismo
8.
Mol Cell Proteomics ; 16(12): 2184-2198, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28951444

RESUMO

The ubiquitous mold Aspergillus fumigatus threatens immunosuppressed patients as inducer of lethal invasive aspergillosis. A. fumigatus conidia are airborne and reach the alveoli, where they encounter alveolar epithelial cells (AEC). Previous studies reported the importance of the surfactant-producing AEC II during A. fumigatus infection via in vitro experiments using cell lines. We established a negative isolation protocol yielding untouched primary murine AEC II with a purity >90%, allowing ex vivo analyses of the cells, which encountered the mold in vivo By label-free proteome analysis of AEC II isolated from mice 24h after A. fumigatus or mock infection we quantified 2256 proteins and found 154 proteins to be significantly differentially abundant between both groups (ANOVA p value ≤ 0.01, ratio of means ≥1.5 or ≤0.67, quantified with ≥2 peptides). Most of these proteins were higher abundant in the infected condition and reflected a comprehensive activation of AEC II on interaction with A. fumigatus This was especially represented by proteins related to oxidative phosphorylation, hence energy production. However, the most strongly induced protein was the l-amino acid oxidase (LAAO) Interleukin 4 induced 1 (IL4I1) with a 42.9 fold higher abundance (ANOVA p value 2.91-10). IL4I1 has previously been found in B cells, macrophages, dendritic cells and rare neurons. Increased IL4I1 abundance in AEC II was confirmed by qPCR, Western blot and immunohistology. Furthermore, A. fumigatus infected lungs showed high levels of IL4I1 metabolic products. Importantly, higher IL4I1 abundance was also confirmed in lung tissue from human aspergilloma. Because LAAO are key enzymes for bactericidal product generation, AEC II might actively participate in pathogen defense. We provide insights into proteome changes of primary AEC II thereby opening new avenues to analyze the molecular changes of this central lung cell on infectious threats. Data are available via ProteomeXchange with identifier PXD005834.


Assuntos
Aspergillus fumigatus/patogenicidade , Flavoproteínas/metabolismo , L-Aminoácido Oxidase/metabolismo , Proteômica/métodos , Alvéolos Pulmonares/citologia , Aspergilose Pulmonar/metabolismo , Adulto , Idoso , Animais , Linhagem Celular , Metabolismo Energético , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Flavoproteínas/genética , Regulação da Expressão Gênica , Humanos , L-Aminoácido Oxidase/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Fosforilação Oxidativa , Mapas de Interação de Proteínas , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Aspergilose Pulmonar/genética
9.
Int J Mol Sci ; 20(23)2019 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-31766758

RESUMO

Nontuberculous mycobacteria (NTM) have recently emerged as important pathogens among cystic fibrosis (CF) patients worldwide. Mycobacterium abscessus is becoming the most worrisome NTM in this cohort of patients and recent findings clarified why this pathogen is so prone to this disease. M. abscessus drug therapy takes up to 2 years and its failure causes an accelerated lung function decline. The M. abscessus colonization of lung alveoli begins with smooth strains producing glycopeptidolipids and biofilm, whilst in the invasive infection, "rough" mutants are responsible for the production of trehalose dimycolate, and consequently, cording formation. Human-to-human M. abscessus transmission was demonstrated among geographically separated CF patients by whole-genome sequencing of clinical isolates worldwide. Using a M. abscessus infected CF zebrafish model, it was demonstrated that CFTR (cystic fibrosis transmembrane conductance regulator) dysfunction seems to have a specific role in the immune control of M. abscessus infections only. This pathogen is also intrinsically resistant to many drugs, thanks to its physiology and to the acquisition of new mechanisms of drug resistance. Few new compounds or drug formulations active against M. abscessus are present in preclinical and clinical development, but recently alternative strategies have been investigated, such as phage therapy and the use of ß-lactamase inhibitors.


Assuntos
Doenças Transmissíveis Emergentes , Fibrose Cística , Farmacorresistência Bacteriana Múltipla/imunologia , Infecções por Mycobacterium não Tuberculosas , Mycobacterium abscessus , Alvéolos Pulmonares , Animais , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/imunologia , Doenças Transmissíveis Emergentes/patologia , Fibrose Cística/epidemiologia , Fibrose Cística/imunologia , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/imunologia , Modelos Animais de Doenças , Humanos , Infecções por Mycobacterium não Tuberculosas/epidemiologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/patologia , Mycobacterium abscessus/imunologia , Mycobacterium abscessus/patogenicidade , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Peixe-Zebra
10.
Am J Respir Cell Mol Biol ; 59(5): 572-579, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29944387

RESUMO

Smoking is a major risk factor driving the tuberculosis epidemic, and smokers' alveolar macrophages (AM) demonstrate significant immune defects after infection. Recently, macrophage glycolytic reprogramming has emerged as crucial in the early host immune response to Mycobacterium tuberculosis (Mtb) infection. In the present study, we sought to compare baseline metabolic characteristics and the glycolytic response to infection of human AM from smokers and nonsmokers. AM were obtained at bronchoscopy, and extracellular flux analyses were performed to determine baseline metabolic characteristics compared with human monocyte-derived macrophages (MDM). Metabolic characterization of AM from smokers and nonsmokers was performed similarly. After infection with Mtb, differences in glycolytic response were measured by extracellular flux analyses and gene expression analyses and correlated with production of glycolysis-driven IL-1ß and prostaglandin E2. Similar experiments were performed in cigarette smoke extract-treated MDM as an alternative model. At baseline, human AM from nonsmokers have a significantly lower extracellular acidification rate/oxygen consumption rate ratio than MDM (P < 0.05), but they retain substantial glycolytic reserve. Compared with nonsmokers' AM, smokers' AM demonstrate reduced metabolic activity, reduced glycolytic reserve (P = 0.051), and reduced spare respiratory capacity (P < 0.01). After infection with Mtb, smokers' AM have significantly reduced glycolytic response, as measured by extracellular flux analyses (P < 0.05) and glycolytic gene expression analyses. Cigarette smoke extract-treated MDM similarly demonstrate reduced metabolic activity and reserves, as well as impaired glycolytic response to infection. Human AM demonstrate metabolic plasticity that allows glycolytic reprogramming to occur after Mtb infection. In smokers, this metabolic reserve is significantly attenuated, with consequent impairment of the glycolytic response to infection.


Assuntos
Fumar Cigarros/efeitos adversos , Metabolismo Energético/imunologia , Macrófagos Alveolares/imunologia , Metaboloma , Mycobacterium tuberculosis/imunologia , Alvéolos Pulmonares/imunologia , Tuberculose/imunologia , Células Cultivadas , Metabolismo Energético/efeitos dos fármacos , Glicólise , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Mycobacterium tuberculosis/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Testes de Função Respiratória , Tuberculose/microbiologia , Tuberculose/patologia
11.
Artigo em Inglês | MEDLINE | ID: mdl-30126953

RESUMO

ETX2514 is a novel ß-lactamase inhibitor that broadly inhibits Ambler class A, C, and D ß-lactamases. ETX2514 combined with sulbactam (SUL) in vitro restores sulbactam activity against Acinetobacter baumannii ETX2514-sulbactam (ETX2514SUL) is under development for the treatment of A. baumannii infections. The objective of this study was to determine and compare plasma, epithelial lining fluid (ELF), and alveolar macrophage (AM) concentrations following intravenous (i.v.) ETX2514 and sulbactam. Plasma, ELF, and AM concentrations of ETX2514 and sulbactam were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in 30 healthy adult subjects following repeated dosing (ETX2514 [1 g] and sulbactam [1 g] every 6 h [q6h], as a 3-h i.v. infusion, for a total of 3 doses). A bronchoalveolar lavage (BAL) was performed once in each subject at either 1, 2.5, 3.25, 4, or 6 h after the start of the last infusion. Penetration ratios were calculated from area under the concentration-time curve from 0 to 6 h (AUC0-6) values for total plasma and ELF using mean and median concentrations at the BAL fluid sampling times. Respective ELF AUC0-6 values, based on mean and median concentrations, were 40.1 and 39.4 mg · h/liter for ETX2514 and 34.7 and 34.5 mg · h/liter for sulbactam. Respective penetration ratios of ELF to total/unbound plasma concentrations, based on mean and median AUC0-6 values, of ETX2514 were 0.37/0.41 and 0.36/0.40, whereas these same ratio values were 0.50/0.81 and 0.50/0.80 for sulbactam. ETX2514 and sulbactam concentrations in AM were measurable and fairly constant throughout the dosing interval (median values of 1.31 and 1.01 mg/liter, respectively). These data support further study of ETX2514SUL for the treatment of pneumonia caused by multidrug-resistant A. baumannii (This study has been registered at ClinicalTrials.gov under identifier NCT03303924.).


Assuntos
Compostos Azabicíclicos/sangue , Compostos Azabicíclicos/metabolismo , Sulbactam/sangue , Sulbactam/metabolismo , Infecções por Acinetobacter/sangue , Infecções por Acinetobacter/tratamento farmacológico , Infecções por Acinetobacter/metabolismo , Acinetobacter baumannii/efeitos dos fármacos , Administração Intravenosa , Adulto , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Antibacterianos/metabolismo , Compostos Azabicíclicos/administração & dosagem , Lavagem Broncoalveolar/métodos , Líquido da Lavagem Broncoalveolar/microbiologia , Feminino , Voluntários Saudáveis , Humanos , Macrófagos Alveolares/microbiologia , Masculino , Pessoa de Meia-Idade , Alvéolos Pulmonares/microbiologia , Sulbactam/administração & dosagem
12.
Histochem Cell Biol ; 147(6): 707-719, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28247028

RESUMO

Loss of alveolar barrier function with subsequent respiratory failure is a hallmark of severe pneumonia. Although junctions between endo- and epithelial cells regulate paracellular fluid flux, little is known about their composition and regulation in the human alveolar compartment. High autofluorescence of human lung tissue in particular complicates the determination of subcellular protein localization. By comparing conventional channel mode confocal imaging with spectral imaging and linear unmixing, we demonstrate that background fluorescent spectra and fluorophore signals could be rigorously separated resulting in complete recovery of the specific signal at a high signal-to-noise ratio. Using this technique and Western blotting, we show the expression patterns of tight junction proteins occludin, ZO-1 as well as claudin-3, -4, -5 and -18 and adherence junction protein VE-cadherin in naive or Streptococcus pneumoniae-infected human lung tissue. In uninfected tissues, occludin and ZO-1 formed band-like structures in alveolar epithelial cells type I (AEC I), alveolar epithelial cells type II (AEC II) and lung capillaries, whereas claudin-3, -4 and -18 were visualised in AEC II. Claudin-5 was detected in the endothelium only. Claudin-3, -5, -18 displayed continuous band-like structures, while claudin-4 showed a dot-like expression. Pneumococcal infection reduced alveolar occludin, ZO-1, claudin-5 and VE-cadherin but did not change the presence of claudin-3, -4 and -18. Spectral confocal microscopy allows for the subcellular structural analysis of proteins in highly autofluorescent human lung tissue. The thereby observed deterioration of lung alveolar junctional organisation gives a structural explanation for alveolar barrier disruption in severe pneumococcal pneumonia.


Assuntos
Caderinas/metabolismo , Síndrome da Persistência do Padrão de Circulação Fetal/metabolismo , Infecções Pneumocócicas/metabolismo , Alvéolos Pulmonares/anormalidades , Humanos , Síndrome da Persistência do Padrão de Circulação Fetal/microbiologia , Infecções Pneumocócicas/microbiologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Streptococcus pneumoniae
13.
J Immunol ; 194(12): 6024-34, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25948816

RESUMO

Neutrophils are key effector cells of the innate immune response to pathogenic bacteria, but excessive neutrophilic inflammation can be associated with bystander tissue damage. The mechanisms responsible for neutrophil recruitment to the lungs during bacterial pneumonia are poorly defined. In this study, we focus on the potential role of the major high-affinity thrombin receptor, proteinase-activated receptor 1 (PAR-1), during the development of pneumonia to the common lung pathogen Streptococcus pneumoniae. Our studies demonstrate that neutrophils were indispensable for controlling S. pneumoniae outgrowth but contributed to alveolar barrier disruption. We further report that intra-alveolar coagulation (bronchoalveolar lavage fluid thrombin-antithrombin complex levels) and PAR-1 immunostaining were increased in this model of bacterial lung infection. Functional studies using the most clinically advanced PAR-1 antagonist, SCH530348, revealed a key contribution for PAR-1 signaling in influencing neutrophil recruitment to lung airspaces in response to both an invasive and noninvasive strain of S. pneumoniae (D39 and EF3030) but that PAR-1 antagonism did not impair the ability of the host to control bacterial outgrowth. PAR-1 antagonist treatment significantly decreased pulmonary levels of IL-1ß, CXCL1, CCL2, and CCL7 and attenuated alveolar leak. Ab neutralization studies further demonstrated a nonredundant role for IL-1ß, CXCL1, and CCL7 in mediating neutrophil recruitment in response to S. pneumoniae infection. Taken together, these data demonstrate a key role for PAR-1 during S. pneumoniae lung infection that is mediated, at least in part, by influencing multiple downstream inflammatory mediators.


Assuntos
Neutrófilos/imunologia , Neutrófilos/metabolismo , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Receptor PAR-1/metabolismo , Animais , Coagulação Sanguínea , Líquido da Lavagem Broncoalveolar/imunologia , Quimiocinas/metabolismo , Quimiotaxia/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Interações Hospedeiro-Patógeno/imunologia , Mediadores da Inflamação/metabolismo , Camundongos , Permeabilidade , Pneumonia Bacteriana/sangue , Pneumonia Bacteriana/patologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/patologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Receptor PAR-1/antagonistas & inibidores , Streptococcus pneumoniae/imunologia
14.
Am J Respir Cell Mol Biol ; 55(6): 837-847, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27468130

RESUMO

New approaches for improving tuberculosis (TB) control using adjunct host-directed cellular and repurposed drug therapies are needed. Autophagy plays a crucial role in the response to TB, and a variety of autophagy-inducing drugs that are currently available for various medical conditions may serve as an adjunct treatment in pulmonary TB. Here, we evaluated the potential of loperamide, carbamazepine, valproic acid, verapamil, and rapamycin to enhance the antimicrobial immune response to Mycobacterium tuberculosis (Mtb). Human monocyte-derived macrophages (MDMs) and murine alveolar cells (MACs) were infected with Mtb and treated with loperamide, carbamazepine, valproic acid, verapamil, and rapamycin in vitro. Balb/c mice were intraperitoneally administered loperamide, valproic acid, and verapamil, and MACs were infected in vitro with Mtb. The induction of autophagy, the containment of Mtb within autophagosomes and the intracellular Mtb burden were determined. Autophagy was induced by all of the drugs in human and mouse macrophages, and loperamide significantly increased the colocalization of microtubule-associated protein 1 light chain 3 with Mtb in MDMs. Carbamazepine, loperamide, and valproic acid induced microtubule-associated protein 1 light chain 3 and autophagy related 16- like protein 1 gene expression in MDMs and in MACs. Loperamide also induced a reduction in TNF-α production. Loperamide and verapamil induced autophagy, which was associated with a significant reduction in the intracellular growth of Mtb in MACs and alveolar macrophages. The intraperitoneal administration of loperamide and valproic acid induced autophagy in freshly isolated MACs. The antimycobacterial activity in MACs was higher after loperamide treatment and was associated with the degradation of p62. In conclusion, loperamide shows potential as an adjunctive therapy for the treatment of TB.


Assuntos
Espaço Intracelular/microbiologia , Loperamida/farmacologia , Pulmão/patologia , Macrófagos/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Carbamazepina/farmacologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Ácido Valproico/farmacologia , Virulência/efeitos dos fármacos
15.
Infect Immun ; 84(9): 2439-48, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27297388

RESUMO

Coxiella burnetii is a Gram-negative bacterium that causes Q fever in humans. Q fever is an atypical pneumonia transmitted through inhalation of contaminated aerosols. In mammalian lungs, C. burnetii infects and replicates in several cell types, including alveolar macrophages (AMs). The innate immunity and signaling pathways operating during infection are still poorly understood, in part because of the lack of relevant host cell models for infection in vitro In the study described here, we investigated and characterized the infection of primary murine AMs by C. burnetii phase II in vitro Our data reveal that AMs show a pronounced M2 polarization and are highly permissive to C. burnetii multiplication in vitro Murine AMs present an increased susceptibility to infection in comparison to primary bone marrow-derived macrophages. AMs support more than 2 logs of bacterial replication during 12 days of infection in culture, similar to highly susceptible host cells, such as Vero and THP-1 cells. As a proof of principle that AMs are useful for investigation of C. burnetii replication, we performed experiments with AMs from Nos2(-/-) or Ifng(-/-) mice. In the absence of gamma interferon and nitric oxide synthase 2 (NOS2), AMs were significantly more permissive than wild-type cells. In contrast, AMs from Il4(-/-) mice were more restrictive to C. burnetii replication, supporting the importance of M2 polarization for the permissiveness of AMs to C. burnetii replication. Collectively, our data account for understanding the high susceptibility of alveolar macrophages to bacterial replication and support the use of AMs as a relevant model of C. burnetii growth in primary macrophages.


Assuntos
Coxiella burnetii/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/microbiologia , Animais , Células Cultivadas , Imunidade Inata/imunologia , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/imunologia , Febre Q/imunologia , Febre Q/microbiologia , Transdução de Sinais/imunologia
16.
Am J Physiol Lung Cell Mol Physiol ; 310(9): L868-79, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26919897

RESUMO

Keratinocyte growth factor (KGF) is an epithelial mitogen that has been reported to protect the lungs from a variety of toxic and infectious insults. In prior studies we found that recombinant human KGF accelerates clearance of bacteria from the murine lung by augmenting the function of alveolar macrophages (AM). In this study we tested the hypothesis that endogenous KGF plays a role in the maintenance of innate pulmonary defense against gram-negative bacterial infections. KGF-deficient mice exhibited delayed clearance of Escherichia coli from the lungs, attenuated phagocytosis by AM, and decreased antimicrobial activity in bronchoalveolar lavage (BAL) fluid, due in part to reductions in levels of surfactant protein A, surfactant protein D, and lysozyme. These immune deficits were accompanied by lower alveolar type II epithelial cell counts and reduced alveolar type II epithelial cell expression of collectin and lysozyme genes on a per cell basis. No significant between-group differences were detected in selected inflammatory cytokines or BAL inflammatory cell populations at baseline or after bacterial challenge in the wild-type and KGF-deficient mice. A single intranasal dose of recombinant human KGF reversed defects in bacterial clearance, AM function, and BAL fluid antimicrobial activity. We conclude that KGF supports alveolar innate immune defense through maintenance of alveolar antimicrobial protein levels and functions of AM. Together these data demonstrate a role for endogenous KGF in maintenance of normal pulmonary innate immune function.


Assuntos
Infecções por Escherichia coli/imunologia , Fator 7 de Crescimento de Fibroblastos/fisiologia , Imunidade Inata , Macrófagos Alveolares/imunologia , Pneumonia Bacteriana/imunologia , Animais , Células Cultivadas , Colectinas/genética , Colectinas/metabolismo , Infecções por Escherichia coli/metabolismo , Feminino , Expressão Gênica , Humanos , Macrófagos Alveolares/microbiologia , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muramidase/genética , Muramidase/metabolismo , Pneumonia Bacteriana/metabolismo , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia
17.
Antimicrob Agents Chemother ; 60(1): 278-87, 2016 01.
Artigo em Inglês | MEDLINE | ID: mdl-26503648

RESUMO

Isavuconazonium sulfate is a novel triazole prodrug that has been recently approved for the treatment of invasive aspergillosis by the FDA. The active moiety (isavuconazole) has a broad spectrum of activity against many pathogenic fungi. This study utilized a dynamic in vitro model of the human alveolus to describe the pharmacodynamics of isavuconazole against two wild-type and two previously defined azole-resistant isolates of Aspergillus fumigatus. A human-like concentration-time profile for isavuconazole was generated. MICs were determined using CLSI and EUCAST methodologies. Galactomannan was used as a measure of fungal burden. Target values for the area under the concentration-time curve (AUC)/MIC were calculated using a population pharmacokinetics-pharmacodynamics (PK-PD) mathematical model. Isolates with higher MICs required higher AUCs in order to achieve maximal suppression of galactomannan. The AUC/MIC targets necessary to achieve 90% probability of galactomannan suppression of <1 were 11.40 and 11.20 for EUCAST and CLSI, respectively.


Assuntos
Antifúngicos/farmacocinética , Aspergillus fumigatus/efeitos dos fármacos , Modelos Estatísticos , Nitrilas/farmacocinética , Piridinas/farmacocinética , Triazóis/farmacocinética , Antifúngicos/farmacologia , Área Sob a Curva , Aspergillus fumigatus/crescimento & desenvolvimento , Reatores Biológicos , Cultura em Câmaras de Difusão , Farmacorresistência Fúngica , Galactose/análogos & derivados , Humanos , Mananas/metabolismo , Mananas/farmacologia , Testes de Sensibilidade Microbiana , Modelos Biológicos , Nitrilas/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/microbiologia , Piridinas/farmacologia , Triazóis/farmacologia
18.
Zhonghua Nei Ke Za Zhi ; 55(1): 40-4, 2016 Jan.
Artigo em Zh | MEDLINE | ID: mdl-26796652

RESUMO

OBJECTIVE: To investigate the function of triggering receptor expresses on myeloid cells receptor-1 (TREM-1) in lymphocyte differentiation and regulation of Aspergillus infected immunosuppressed rats. METHODS: Cyclophosphamide (CTX) was intraperitoneally injected and Fumigatus spore suspension was inhaled by percutaneous tracheostomy to establish the immunosuppressive invasive pulmonary aspergillosis (IPA) rat model. After 24 h, 48 h, 72 h and 96 h inoculation, rats were sacrificed. Lung tissue specimens, bronchoalveolar lavage fluid (BALF), and plasma samples were collected. Plasma and BALF sTREM-1, plasma T cell-specific transcription factor (T-box expressed in T cells, T-bet) and eomesodermin(Eomes) were detected by ELISA. Biopsy specimens of lung tissue were used for periodic acid-schiff (PAS) staining and culture. RESULTS: The mortality rate of immunosuppressed rats after Aspergillus inhalation for 96 h was as high as 54.4%. Biopsy of lung tissue suggested acute inflammatory cell infiltration, interstitial lung congestion, alveolar structural damage, and visible Aspergillus hyphae in alveoli. Compared with normal control group[(110.50 ± 7.70) ng/L], plasma sTREM-1 in study groups were significantly increased[IPA: (146.77 ± 10.41) ng/L; CXT+ IPA at 24 h: (226.00 ± 11.88) ng/L; CTX+ IPA at 48 h: (200.77 ± 10.63) ng/L; P<0.05], so were T-bet levels[IPA: (561.17 ± 7.23) µg/L; CXT+ IPA at 24 h: (647.00 ± 33.03) µg/L; CTX+ IPA at 48 h: (619.23 ± 87.44) µg/L; control group: (340.03 ± 26.32) µg/L; respectively, P < 0.05]. However, plasma Eomes levels in IPA group, CTX+ IPA at 24 h and 48 h were significantly lower compared with that in normal controls[IPA: (7.96 ± 0.65) ng/L; CXT+ IPA at 24 h: (3.97 ± 0.35) ng/L; CTX+ IPA at 48 h: (4.00 ± 0.74) ng/L; control group: (8.38 ± 0.51) ng/L; respectively, P < 0.001]. Compared with those in CTX+ IPA vaccination after 24 h and 48 h, plasma sTREM-1 [(106.67 ± 7.64) ng/L; (133.27 ± 32.79) ng/L] and T-bet [(299.64 ± 63.07) µg/L; (398.02 ± 109.22) µg/L] in CTX+ IPA at 72 h and 96 h inoculation were significantly lower (P < 0.001). While Eomes [(8.38 ± 0.54) ng/L; (8.40 ± 0.70) ng/L]raised significantly higher (P < 0.001). Compared with the control group, sTREM-1 levels in BALF of IPA+ CTX 24 h, 48 h, 72 h, and 96 h groups were consistently high (P < 0.05). Pearson correlation analysis showed that sTREM-1 and T-bet had a significant positive correlation (r = 0.91, P < 0.001), yet Eomes was negatively correlated with them (r = -0.788, P < 0.001). CONCLUSIONS: sTREM-1 in rat plasma and BALF appears highly expressed in immune compromised Aspergillus infected rat model. Plasma sTREM-1 is closely correlated with T-bet and Eomes levels, which suggests that TREM-1 may be involved in lymphocytic regulation and differentiation during fungal infection.


Assuntos
Terapia de Imunossupressão , Aspergilose Pulmonar Invasiva/metabolismo , Receptores Imunológicos/metabolismo , Proteínas com Domínio T/sangue , Administração por Inalação , Animais , Aspergillus , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Inflamação , Aspergilose Pulmonar Invasiva/patologia , Pulmão/microbiologia , Pulmão/patologia , Células Mieloides , Alvéolos Pulmonares/microbiologia , Alvéolos Pulmonares/patologia , Ratos , Receptores Imunológicos/sangue , Receptor Gatilho 1 Expresso em Células Mieloides
19.
Eur J Immunol ; 44(2): 440-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24214530

RESUMO

Mucosal boosting of BCG-immunised individuals with a subunit tuberculosis (TB) vaccine would be highly desirable, considering that the lungs are the principal port of entry for Mycobacterium tuberculosis (MTB) and the site of the primary infection and reactivation. However, the main roadblock for subunit TB vaccine development is the lack of suitable adjuvants that could induce robust local and systemic immune responses. Here, we describe a novel vaccine delivery system that was designed to mimic, in part, the MTB pathogen itself. The surface of yellow carnauba wax nanoparticles was coated with the highly immunogenic Ag85B Ag of MTB and they were directed to the alveolar epithelial surfaces by the incorporation of the heparin-binding hemagglutinin adhesion (HBHA) protein. Our results showed that the i.n. immunisation of BCG-primed BALB/c mice with nanoparticles adsorbed with Ag85B-HBHA (Nano-AH vaccine) induced robust humoral and cellular immune responses and IFN-γ production, and multifunctional CD4⁺ T cells expressing IFN-γ, IL-2 and TNF-α. Mice challenged with H37Rv MTB had a significantly reduced bacterial load in their lungs when compared with controls immunised with BCG alone. We therefore conclude that this immunisation approach is an effective means of boosting the BCG-induced anti-TB immunity.


Assuntos
Antígenos de Bactérias/imunologia , Mycobacterium tuberculosis/imunologia , Nanopartículas/administração & dosagem , Alvéolos Pulmonares/imunologia , Mucosa Respiratória/imunologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/imunologia , Aciltransferases/genética , Aciltransferases/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Animais , Antígenos de Bactérias/genética , Vacina BCG/imunologia , Carga Bacteriana/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Feminino , Interferon gama/imunologia , Interleucina-2/imunologia , Lectinas/genética , Lectinas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Alvéolos Pulmonares/microbiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Mucosa Respiratória/microbiologia , Tuberculose/microbiologia , Vacinas contra a Tuberculose/genética , Fator de Necrose Tumoral alfa/imunologia , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia
20.
Can J Microbiol ; 61(12): 938-47, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26492080

RESUMO

Despite the interactions known to occur between various lower respiratory tract pathogens and alveolar epithelial cells (AECs), few reports examine factors influencing the interplay between Mycobacterium tuberculosis bacilli and AECs during infection. Importantly, in vitro studies have demonstrated that the M. tuberculosis hbha and esxA gene products HBHA and ESAT6 directly or indirectly influence AEC survival. In this report, we identify Rv3351c as another M. tuberculosis gene that impacts the fate of both the pathogen and AEC host. Intracellular replication of an Rv3351c mutant in the human AEC type II pneumocyte cell line A549 was markedly reduced relative to the complemented mutant and parent strain. Deletion of Rv3351c diminished the release of lactate dehydrogenase and decreased uptake of trypan blue vital stain by host cells infected with M. tuberculosis bacilli, suggesting attenuated cytotoxic effects. Interestingly, an isogenic hbha mutant displayed reductions in AEC killing similar to those observed for the Rv3351c mutant. This opens the possibility that multiple M. tuberculosis gene products interact with AECs. We also observed that Rv3351c aids intracellular replication and survival of M. tuberculosis in macrophages. This places Rv3351c in the same standing as HBHA and ESAT6, which are important factors in AECs and macrophages. Defining the mechanism(s) by which Rv3351c functions to aid pathogen survival within the host may lead to new drug or vaccine targets.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/microbiologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/metabolismo , Alvéolos Pulmonares/microbiologia , Tuberculose/microbiologia , Proteínas de Bactérias/genética , Linhagem Celular , Sobrevivência Celular , Células Epiteliais/citologia , Humanos , Macrófagos/microbiologia , Mycobacterium tuberculosis/genética , Alvéolos Pulmonares/citologia , Tuberculose/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA