Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Respir Crit Care Med ; 206(4): 427-439, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35536732

RESUMO

Rationale: Chronic obstructive pulmonary disease (COPD) is variable in its development. Lung microbiota and metabolites collectively may impact COPD pathophysiology, but relationships to clinical outcomes in milder disease are unclear. Objectives: Identify components of the lung microbiome and metabolome collectively associated with clinical markers in milder stage COPD. Methods: We analyzed paired microbiome and metabolomic data previously characterized from bronchoalveolar lavage fluid in 137 participants in the SPIROMICS (Subpopulations and Intermediate Outcome Measures in COPD Study), or (GOLD [Global Initiative for Chronic Obstructive Lung Disease Stage 0-2). Datasets used included 1) bacterial 16S rRNA gene sequencing; 2) untargeted metabolomics of the hydrophobic fraction, largely comprising lipids; and 3) targeted metabolomics for a panel of hydrophilic compounds previously implicated in mucoinflammation. We applied an integrative approach to select features and model 14 individual clinical variables representative of known associations with COPD trajectory (lung function, symptoms, and exacerbations). Measurements and Main Results: The majority of clinical measures associated with the lung microbiome and metabolome collectively in overall models (classification accuracies, >50%, P < 0.05 vs. chance). Lower lung function, COPD diagnosis, and greater symptoms associated positively with Streptococcus, Neisseria, and Veillonella, together with compounds from several classes (glycosphingolipids, glycerophospholipids, polyamines and xanthine, an adenosine metabolite). In contrast, several Prevotella members, together with adenosine, 5'-methylthioadenosine, sialic acid, tyrosine, and glutathione, associated with better lung function, absence of COPD, or less symptoms. Significant correlations were observed between specific metabolites and bacteria (Padj < 0.05). Conclusions: Components of the lung microbiome and metabolome in combination relate to outcome measures in milder COPD, highlighting their potential collaborative roles in disease pathogenesis.


Assuntos
Microbiota , Doença Pulmonar Obstrutiva Crônica , Adenosina , Humanos , Pulmão/patologia , Doença Pulmonar Obstrutiva Crônica/diagnóstico , RNA Ribossômico 16S/genética
3.
Lancet Respir Med ; 9(6): 601-612, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33460570

RESUMO

BACKGROUND: Alterations in the respiratory microbiome are common in chronic lung diseases, correlate with decreased lung function, and have been associated with disease progression. The clinical significance of changes in the respiratory microbiome after lung transplant, specifically those related to development of chronic lung allograft dysfunction (CLAD), are unknown. The aim of this study was to evaluate the effect of lung microbiome characteristics in healthy lung transplant recipients on subsequent CLAD-free survival. METHODS: We prospectively studied a cohort of lung transplant recipients at the University of Michigan (Ann Arbor, MI, USA). We analysed characteristics of the respiratory microbiome in acellular bronchoalveolar lavage fluid (BALF) collected from asymptomatic patients during per-protocol surveillance bronchoscopy 1 year after lung transplantation. For our primary endpoint, we evaluated a composite of development of CLAD or death at 500 days after the 1-year surveillance bronchoscopy. Our primary microbiome predictor variables were bacterial DNA burden (total 16S rRNA gene copies per mL of BALF, quantified via droplet digital PCR) and bacterial community composition (determined by bacterial 16S rRNA gene sequencing). Patients' lung function was followed serially at least every 3 months by spirometry, and CLAD was diagnosed according to International Society of Heart and Lung Transplant 2019 guidelines. FINDINGS: We analysed BALF from 134 patients, collected during 1-year post-transplant surveillance bronchoscopy between Oct 21, 2005, and Aug 25, 2017. Within 500 days of follow-up from the time of BALF sampling, 24 (18%) patients developed CLAD, five (4%) died before confirmed development of CLAD, and 105 (78%) patients remained CLAD-free with complete follow-up. Lung bacterial burden was predictive of CLAD development or death within 500 days of the surveillance bronchoscopy, after controlling for demographic and clinical factors, including immunosuppression and bacterial culture results, in a multivariable survival model. This relationship was evident when burden was analysed as a continuous variable (per log10 increase in burden, HR 2·49 [95% CI 1·38-4·48], p=0·0024) or by tertiles (middle vs lowest bacterial burden tertile, HR 4·94 [1·25-19·42], p=0·022; and highest vs lowest, HR 10·56 [2·53-44·08], p=0·0012). In patients who developed CLAD or died, composition of the lung bacterial community significantly differed to that in patients who survived and remained CLAD-free (on permutational multivariate analysis of variance, p=0·047 at the taxonomic level of family), although differences in community composition were associated with bacterial burden. No individual bacterial taxa were definitively associated with CLAD development or death. INTERPRETATION: Among asymptomatic lung transplant recipients at 1-year post-transplant, increased lung bacterial burden is predictive of chronic rejection and death. The lung microbiome represents an understudied and potentially modifiable risk factor for lung allograft dysfunction. FUNDING: US National Institutes of Health, Cystic Fibrosis Foundation, Brian and Mary Campbell and Elizabeth Campbell Carr research gift fund.


Assuntos
Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/microbiologia , Transplante de Pulmão , Pulmão/microbiologia , Microbiota , Transplantados/estatística & dados numéricos , Doença Crônica , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos
4.
Sci Transl Med ; 12(556)2020 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-32801143

RESUMO

Inhaled oxygen, although commonly administered to patients with respiratory disease, causes severe lung injury in animals and is associated with poor clinical outcomes in humans. The relationship between hyperoxia, lung and gut microbiota, and lung injury is unknown. Here, we show that hyperoxia conferred a selective relative growth advantage on oxygen-tolerant respiratory microbial species (e.g., Staphylococcus aureus) as demonstrated by an observational study of critically ill patients receiving mechanical ventilation and experiments using neonatal and adult mouse models. During exposure of mice to hyperoxia, both lung and gut bacterial communities were altered, and these communities contributed to oxygen-induced lung injury. Disruption of lung and gut microbiota preceded lung injury, and variation in microbial communities correlated with variation in lung inflammation. Germ-free mice were protected from oxygen-induced lung injury, and systemic antibiotic treatment selectively modulated the severity of oxygen-induced lung injury in conventionally housed animals. These results suggest that inhaled oxygen may alter lung and gut microbial communities and that these communities could contribute to lung injury.


Assuntos
Microbioma Gastrointestinal , Hiperóxia , Lesão Pulmonar , Animais , Humanos , Pulmão , Lesão Pulmonar/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio
6.
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
7.
Int J Chron Obstruct Pulmon Dis ; 13: 2289-2299, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30104869

RESUMO

Background: The role of airway microbiota in COPD is highly debated. Symptomology assessment is vital for the management of clinically stable COPD patients; however, the link between symp toms and the airway microbiome is currently unknown. Purpose: The present study aimed to evaluate the relationship among stable COPD patients. Patients and methods: We conducted pyrosequencing of bacterial 16S rRNA using induced sputum samples in a Han Chinese cohort that included 40 clinically stable COPD patients and 19 healthy controls. Results: Alterations in community composition and core bacte rial taxa (Neisseria subflava, etc.) were observed in patients with severe symptoms compared with controls. The co-occurrence network indicated that the key microbiota enriched in COPD patients showed higher expression in patients with severe symptoms. The association pattern of symptoms with the sputum microbiome was obviously different from that of lung function in COPD patients. Conclusion: These findings broaden our insights into the relationship between the sputum microbiota and the symptom severity in COPD patients, emphasizing the role of symptoms in the airway microbiome, independent of lung function.


Assuntos
Microbiota , Doença Pulmonar Obstrutiva Crônica/microbiologia , Escarro/microbiologia , Avaliação de Sintomas , Corticosteroides , Adulto , Idoso , Estudos de Casos e Controles , Volume Expiratório Forçado , Humanos , Masculino , Microbiota/genética , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/diagnóstico , RNA Ribossômico 16S/genética , Fumar
8.
Am J Respir Crit Care Med ; 198(10): 1312-1321, 2018 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29878854

RESUMO

RATIONALE: Hematopoietic cell transplant (HCT) is a common treatment for hematological neoplasms and autoimmune disorders. Among HCT recipients, pulmonary complications are common, morbid, and/or lethal, and they have recently been associated with gut dysbiosis. The role of lung microbiota in post-HCT pulmonary complications is unknown. OBJECTIVES: To investigate the role of lung microbiota in post-HCT pulmonary complications using animal modeling and human BAL fluid. METHODS: For animal modeling, we used an established murine model of HCT with and without postengraftment herpes virus infection. For human studies, we characterized lung microbiota in BAL fluid from 43 HCT recipients. Lung bacteria were characterized using 16S ribosomal RNA gene sequencing and were compared with lung histology (murine) and with alveolar inflammation and pulmonary function testing (human). MEASUREMENTS AND MAIN RESULTS: Both HCT and viral infection independently altered the composition of murine lung microbiota, but they had no effect on lung microbial diversity. By contrast, combined HCT and viral infection profoundly altered lung microbiota, decreasing community diversity with an associated pneumonitis. Among human HCT recipients, increased relative abundance of the Proteobacteria phylum was associated with impaired pulmonary function, and lung microbiota were significantly associated with alveolar concentrations of inflammatory cytokines. CONCLUSIONS: In animal models and human subjects, lung dysbiosis is a prominent feature of HCT. Lung dysbiosis is correlated with histologic, immunologic, and physiologic features of post-HCT pulmonary complications. Our findings suggest the lung microbiome may be an unappreciated target for the prevention and treatment of post-HCT pulmonary complications.


Assuntos
Disbiose/epidemiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Inflamação/epidemiologia , Pneumopatias/epidemiologia , Complicações Pós-Operatórias/epidemiologia , Animais , Comorbidade , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Humanos , Inflamação/microbiologia , Pulmão/microbiologia , Pneumopatias/microbiologia , Masculino , Camundongos , Pessoa de Meia-Idade , Complicações Pós-Operatórias/microbiologia
9.
Genome Announc ; 6(7)2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29449399

RESUMO

We report here the first complete genome sequence of a human Pseudomonas lundensis isolate, strain AU1044, and the draft genomes of 11 other clinical P. lundensis strains, isolated from the lungs of cystic fibrosis patients. The genome of strain AU1044 is 4.81 Mb and encodes seven 16S rRNAs.

10.
mBio ; 8(1)2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28196961

RESUMO

Although culture-independent techniques have refuted lung sterility in health, controversy about contamination during bronchoscope passage through the upper respiratory tract (URT) has impeded research progress. We sought to establish whether bronchoscopic sampling accurately reflects the lung microbiome in health and to distinguish between two proposed routes of authentic microbial immigration, (i) dispersion along contiguous respiratory mucosa and (ii) subclinical microaspiration. During bronchoscopy of eight adult volunteers without lung disease, we performed seven protected specimen brushings (PSB) and bilateral bronchoalveolar lavages (BALs) per subject. We amplified, sequenced, and analyzed the bacterial 16S rRNA gene V4 regions by using the Illumina MiSeq platform. Rigorous attention was paid to eliminate potential sources of error or contamination, including a randomized processing order and the inclusion and analysis of exhaustive procedural and sequencing control specimens. Indices of mouth-lung immigration (mouth-lung community similarity, bacterial burden, and community richness) were all significantly greater in airway and alveolar specimens than in bronchoscope contamination control specimens, indicating minimal evidence of pharyngeal contamination. Ecological indices of mouth-lung immigration peaked at or near the carina, as predicted for a primary immigration route of microaspiration. Bacterial burden, diversity, and mouth-lung similarity were greater in BAL than PSB samples, reflecting differences in the sampled surface areas. (This study has been registered at ClinicalTrials.gov under registration no. NCT02392182.)IMPORTANCE This study defines the bacterial topography of the healthy human respiratory tract and provides ecological evidence that bacteria enter the lungs in health primarily by microaspiration, with potential contribution in some subjects by direct dispersal along contiguous mucosa. By demonstrating that contamination contributes negligibly to microbial communities in bronchoscopically acquired specimens, we validate the use of bronchoscopy to investigate the lung microbiome.


Assuntos
Bactérias/genética , Bactérias/isolamento & purificação , Líquido da Lavagem Broncoalveolar/microbiologia , Pulmão/microbiologia , Metagenoma , Microbiota , Adulto , Idoso , Bactérias/classificação , Lavagem Broncoalveolar/instrumentação , Lavagem Broncoalveolar/normas , Broncoscopia/efeitos adversos , Broncoscopia/normas , DNA Bacteriano/genética , Feminino , Voluntários Saudáveis , Humanos , Masculino , Microbiota/genética , Pessoa de Meia-Idade , Boca/microbiologia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
11.
Am J Respir Crit Care Med ; 196(2): 208-219, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28157391

RESUMO

RATIONALE: Differences in the lung microbial community influence idiopathic pulmonary fibrosis (IPF) progression. Whether the lung microbiome influences IPF host defense remains unknown. OBJECTIVES: To explore the host immune response and microbial interaction in IPF as they relate to progression-free survival (PFS), fibroblast function, and leukocyte phenotypes. METHODS: Paired microarray gene expression data derived from peripheral blood mononuclear cells as well as 16S ribosomal RNA sequencing data from bronchoalveolar lavage obtained as part of the COMET-IPF (Correlating Outcomes with Biochemical Markers to Estimate Time-Progression in Idiopathic Pulmonary Fibrosis) study were used to conduct association pathway analyses. The responsiveness of paired lung fibroblasts to Toll-like receptor 9 (TLR9) stimulation by CpG-oligodeoxynucleotide (CpG-ODN) was integrated into microbiome-gene expression association analyses for a subset of individuals. The relationship between associated pathways and circulating leukocyte phenotypes was explored by flow cytometry. MEASUREMENTS AND MAIN RESULTS: Down-regulation of immune response pathways, including nucleotide-binding oligomerization domain (NOD)-, Toll-, and RIG1-like receptor pathways, was associated with worse PFS. Ten of the 11 PFS-associated pathways correlated with microbial diversity and individual genus, with species accumulation curve richness as a hub. Higher species accumulation curve richness was significantly associated with inhibition of NODs and TLRs, whereas increased abundance of Streptococcus correlated with increased NOD-like receptor signaling. In a network analysis, expression of up-regulated signaling pathways was strongly associated with decreased abundance of operational taxonomic unit 1341 (OTU1341; Prevotella) among individuals with fibroblasts responsive to CpG-ODN stimulation. The expression of TLR signaling pathways was also linked to CpG-ODN responsive fibroblasts, OTU1341 (Prevotella), and Shannon index of microbial diversity in a network analysis. Lymphocytes expressing C-X-C chemokine receptor 3 CD8 significantly correlated with OTU1348 (Staphylococcus). CONCLUSIONS: These findings suggest that host-microbiome interactions influence PFS and fibroblast responsiveness.


Assuntos
Fibrose Pulmonar Idiopática/imunologia , Fibrose Pulmonar Idiopática/microbiologia , Imunidade Inata/imunologia , Microbiota/imunologia , Lavagem Broncoalveolar , Intervalo Livre de Doença , Regulação para Baixo/imunologia , Feminino , Citometria de Fluxo , Expressão Gênica/imunologia , Humanos , Masculino , Análise em Microsséries , Pessoa de Meia-Idade
12.
Nat Microbiol ; 1(10): 16113, 2016 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-27670109

RESUMO

Sepsis and the acute respiratory distress syndrome (ARDS) are major causes of mortality without targeted therapies. Although many experimental and clinical observations have implicated gut microbiota in the pathogenesis of these diseases, culture-based studies have failed to demonstrate translocation of bacteria to the lungs in critically ill patients. Here, we report culture-independent evidence that the lung microbiome is enriched with gut bacteria both in a murine model of sepsis and in humans with established ARDS. Following experimental sepsis, lung communities were dominated by viable gut-associated bacteria. Ecological analysis identified the lower gastrointestinal tract, rather than the upper respiratory tract, as the likely source community of post-sepsis lung bacteria. In bronchoalveolar lavage fluid from humans with ARDS, gut-specific bacteria (Bacteroides spp.) were common and abundant, undetected by culture and correlated with the intensity of systemic inflammation. Alveolar TNF-α, a key mediator of alveolar inflammation in ARDS, was significantly correlated with altered lung microbiota. Our results demonstrate that the lung microbiome is enriched with gut-associated bacteria in sepsis and ARDS, potentially representing a shared mechanism of pathogenesis in these common and lethal diseases.


Assuntos
Trato Gastrointestinal/microbiologia , Pulmão/microbiologia , Microbiota , Síndrome do Desconforto Respiratório/microbiologia , Sepse/microbiologia , Adulto , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bacteroides/crescimento & desenvolvimento , Bacteroides/isolamento & purificação , Líquido da Lavagem Broncoalveolar/microbiologia , Modelos Animais de Doenças , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Inflamação/microbiologia , Pulmão/imunologia , Pulmão/fisiopatologia , Masculino , Camundongos , Pessoa de Meia-Idade , Fator de Necrose Tumoral alfa/imunologia
13.
BMC Genomics ; 16: 1032, 2015 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-26644001

RESUMO

BACKGROUND: While the taxonomy and genomics of environmental strains from the P. fluorescens species-complex has been reported, little is known about P. fluorescens strains from clinical samples. In this report, we provide the first genomic analysis of P. fluorescens strains in which human vs. environmental isolates are compared. RESULTS: Seven P. fluorescens strains were isolated from respiratory samples from cystic fibrosis (CF) patients. The clinical strains could grow at a higher temperature (>34 °C) than has been reported for environmental strains. Draft genomes were generated for all of the clinical strains, and multi-locus sequence analysis placed them within subclade III of the P. fluorescens species-complex. All strains encoded type- II, -III, -IV, and -VI secretion systems, as well as the widespread colonization island (WCI). This is the first description of a WCI in P. fluorescens strains. All strains also encoded a complete I2/PfiT locus and showed evidence of horizontal gene transfer. The clinical strains were found to differ from the environmental strains in the number of genes involved in metal resistance, which may be a possible adaptation to chronic antibiotic exposure in the CF lung. CONCLUSIONS: This is the largest comparative genomics analysis of P. fluorescens subclade III strains to date and includes the first clinical isolates. At a global level, the clinical P. fluorescens subclade III strains were largely indistinguishable from environmental P. fluorescens subclade III strains, supporting the idea that identifying strains as 'environmental' vs 'clinical' is not a phenotypic trait. Rather, strains within P. fluorescens subclade III will colonize and persist in any niche that provides the requirements necessary for growth.


Assuntos
Genoma Bacteriano , Genômica , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas fluorescens/classificação , Pseudomonas fluorescens/genética , Sistemas de Secreção Bacterianos/genética , Composição de Bases , Fibrose Cística/complicações , Loci Gênicos , Genômica/métodos , Genótipo , Humanos , Metais/metabolismo , Família Multigênica , Fenótipo , Filogenia , Pseudomonas fluorescens/isolamento & purificação , Pseudomonas fluorescens/metabolismo , Metabolismo Secundário/genética , Análise de Sequência de DNA
14.
Ann Am Thorac Soc ; 12(6): 821-30, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25803243

RESUMO

RATIONALE: The lung microbiome is spatially heterogeneous in advanced airway diseases, but whether it varies spatially in health is unknown. We postulated that the primary determinant of lung microbiome constitution in health is the balance of immigration and elimination of communities from the upper respiratory tract (URT; "adapted island model of lung biogeography"), rather than differences in regional bacterial growth conditions. OBJECTIVES: To determine if the lung microbiome is spatially varied in healthy adults. METHODS: Bronchoscopy was performed on 15 healthy subjects. Specimens were sequentially collected in the lingula and right middle lobe (by bronchoalveolar lavage [BAL]), then in the right upper lobe, left upper lobe, and supraglottic space (by protected-specimen brush). Bacterial 16S ribosmal RNA-encoding genes were sequenced using MiSeq (Illumina, San Diego, CA). MEASUREMENTS AND MAIN RESULTS: There were no significant differences between specimens collected by BAL and protected-specimen brush. Spatially separated intrapulmonary sites, when compared with each other, did not contain consistently distinct microbiota. On average, intrasubject variation was significantly less than intersubject variation (P = 0.00003). By multiple ecologic parameters (community richness, community composition, intersubject variability, and similarity to source community), right upper lobe microbiota more closely resembled those of the URT than did microbiota from more distal sites. As predicted by the adapted island model, community richness decreased with increasing distance from the source community of the URT (P < 0.05). CONCLUSIONS: In healthy lungs, spatial variation in microbiota within an individual is significantly less than variation across individuals. The lung microbiome in health is more influenced by microbial immigration and elimination (the adapted island model) than by the effects of local growth conditions on bacterial reproduction rates, which are more determinant in advanced lung diseases. BAL of a single lung segment is an acceptable method of sampling the healthy lung microbiome. Clinical trial registered with www.clinicaltrials.gov (NCT02392182).


Assuntos
Líquido da Lavagem Broncoalveolar/microbiologia , Pulmão , Metagenoma/fisiologia , Microbiota/fisiologia , Adulto , Lavagem Broncoalveolar/métodos , Broncoscopia/métodos , DNA Bacteriano/análise , Feminino , Voluntários Saudáveis , Humanos , Pulmão/microbiologia , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , RNA Ribossômico 16S/análise , Análise de Sequência de DNA , Análise Espacial
15.
Genome Announc ; 3(1)2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25635025

RESUMO

We report here the first draft genome sequences of Pseudomonas fluorescens strains that have been isolated from humans. The seven assembled draft genomes contained an average of 60.1% G+C content, were an average genomic size of 6.3 Mbp, and mapped by multilocus sequence analysis to subclade III.

16.
Immunology ; 144(4): 704-16, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25399934

RESUMO

The host response to Clostridium difficile infection in antibiotic-treated mice is characterized by robust recruitment of Gr-1(+) cells, increased expression of inflammatory cytokines including tumour necrosis factor-α (TNF-α), and the development of severe epithelial damage. To investigate the role of Gr-1(+) cells and TNF-α during C. difficile colitis, we treated infected mice with monoclonal antibodies against Gr-1 or TNF-α. Mice were challenged with vegetative cells of C. difficile strain VPI 10463 following treatment with the third-generation cephalosporin ceftriaxone. Ceftriaxone treatment alone was associated with significant changes in cytokine expression within the colonic mucosa but not overt inflammatory histopathological changes. In comparison, C. difficile infection following ceftriaxone treatment was associated with increased expression of inflammatory cytokines and chemokines including Cxcl1, Cxcl2, Il1b, Il17f and Tnfa, as well as robust recruitment of Ly6C(Mid)  Gr-1(High) neutrophils and Ly6C(High) Gr-1(Mid) monocytes and the development of severe colonic histopathology. Anti-Gr-1 antibody treatment resulted in effective depletion of both Ly6C(Mid) Gr-1(High) neutrophils and Ly6C(High) Gr-1(Mid) monocytes: however, we observed no protection from the development of severe pathology or reduction in expression of the pro-inflammatory cytokines Il1b, Il6, Il33 and Tnfa following anti-Gr-1 treatment. By contrast, anti-TNF-α treatment did not affect Gr-1(+) cell recruitment, but was associated with increased expression of Il6 and Il1b. Additionally, Ffar2, Ffar3, Tslp, Tff and Ang4 expression was significantly reduced in anti-TNF-α-treated animals, in association with marked intestinal histopathology. These studies raise the possibility that TNF-α may play a role in restraining inflammation and protecting the epithelium during C. difficile infection.


Assuntos
Clostridioides difficile/patogenicidade , Colo/metabolismo , Enterocolite Pseudomembranosa/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Anticorpos Monoclonais/farmacologia , Ceftriaxona , Clostridioides difficile/imunologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Modelos Animais de Doenças , Enterocolite Pseudomembranosa/genética , Enterocolite Pseudomembranosa/imunologia , Enterocolite Pseudomembranosa/microbiologia , Enterocolite Pseudomembranosa/patologia , Enterocolite Pseudomembranosa/prevenção & controle , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Masculino , Camundongos Endogâmicos C57BL , Microbiota , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/imunologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/imunologia
17.
Ann Am Thorac Soc ; 10 Suppl: S170-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24313769

RESUMO

Chronic obstructive pulmonary disease (COPD) is one of few chronic disorders with rising mortality and morbidity. It is a highly prevalent disorder, characterized by highly heterogeneous clinical symptoms, health status, and disease progression. COPD is also characterized by an inflammatory/immune response that persists despite smoking cessation and varies by the patient population, method of assessment, and timing of measurement. Bacterial colonization or infection is ubiquitous in patients with COPD and, until recently, has been predominantly assessed using culture-based methodologies. This colonization has been believed to be biologically relevant. It has been estimated that more than 70% of the bacterial species on body surfaces cannot be cultured by standard techniques. As such, advanced culture-independent techniques have been developed that target bacterial genes, such as the 16S ribosomal RNA gene, that function as molecular chronometers. Application of these techniques in patients with COPD has suggested microbial diversity that varies by age, disease severity, and medication use. All of these data provide unique and rapidly evolving insight into the potential role of the respiratory microbiome in disease genesis and expression.


Assuntos
Infecções Bacterianas/microbiologia , DNA Bacteriano/análise , Microbiota/genética , Doença Pulmonar Obstrutiva Crônica/microbiologia , RNA Ribossômico 16S/genética , Infecções Respiratórias/microbiologia , Infecções Bacterianas/diagnóstico , Infecções Bacterianas/imunologia , Técnicas de Cultura , Progressão da Doença , Humanos , Doença Pulmonar Obstrutiva Crônica/imunologia , Infecções Respiratórias/diagnóstico , Infecções Respiratórias/imunologia , Fumar/imunologia
18.
Expert Rev Respir Med ; 7(3): 245-57, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23734647

RESUMO

Novel culture-independent techniques have recently demonstrated that the lower respiratory tract, historically considered sterile in health, contains diverse communities of microbes: the lung microbiome. Increasing evidence supports the concept that a distinct microbiota of the lower respiratory tract is present both in health and in various respiratory diseases, although the biological and clinical significance of these findings remains undetermined. In this article, the authors review and synthesize published reports of the lung microbiota of healthy and diseased subjects, discuss trends of microbial diversity and constitution across disease states, and look to the extrapulmonary microbiome for hypotheses and future directions for study.


Assuntos
Bactérias , Pulmão/microbiologia , Microbiota , Infecções Respiratórias/microbiologia , Animais , Asma/microbiologia , Bactérias/genética , Bactérias/crescimento & desenvolvimento , Bactérias/isolamento & purificação , Técnicas Bacteriológicas , Fibrose Cística/microbiologia , Interações Hospedeiro-Patógeno , Humanos , Pneumonias Intersticiais Idiopáticas/microbiologia , Doença Pulmonar Obstrutiva Crônica/microbiologia , Infecções Respiratórias/terapia , Fatores de Risco
19.
J Immunol ; 190(7): 3447-57, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23467934

RESUMO

The alveolar epithelium is characteristically abnormal in fibrotic lung disease, and we recently established a direct link between injury to the type II alveolar epithelial cell (AEC) and the accumulation of interstitial collagen. The mechanisms by which damage to the epithelium induces lung scarring remain poorly understood. It is particularly controversial whether an insult to the type II AEC initiates an inflammatory response that is required for the development of fibrosis. To explore whether local inflammation occurs following a targeted epithelial insult and contributes to lung fibrosis, we administered diphtheria toxin to transgenic mice with type II AEC-restricted expression of the diphtheria toxin receptor. We used immunophenotyping techniques and diphtheria toxin receptor-expressing, chemokine receptor-2-deficient (CCR2(-/-)) mice to determine the participation of lung leukocyte subsets in pulmonary fibrogenesis. Our results demonstrate that targeted type II AEC injury induces an inflammatory response that is enriched for CD11b(+) nonresident exudate macrophages (ExM) and their precursors, Ly-6C(high) monocytes. CCR2 deficiency abrogates the accumulation of both cell populations and protects mice from fibrosis, weight loss, and death. Further analyses revealed that the ExM are alternatively activated and that ExM and Ly-6C(high) monocytes express mRNA for IL-13, TGF-ß, and the collagen genes, COL1A1 and COLIIIA1. Furthermore, the accumulated ExM and Ly-6C(high) monocytes contain intracellular collagen, as detected by immunostaining. Together, these results implicate CCR2 and the accumulation of ExM and Ly-6C(high) monocytes as critical determinants of pulmonary fibrosis induced by selective type II AEC injury.


Assuntos
Exsudatos e Transudatos/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Fibrose Pulmonar/genética , Fibrose Pulmonar/imunologia , Receptores CCR2/genética , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/metabolismo , Animais , Antígenos Ly/imunologia , Colágeno/biossíntese , Citocinas/genética , Citocinas/imunologia , Exsudatos e Transudatos/citologia , Expressão Gênica , Marcação de Genes , Imunofenotipagem , Ativação de Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Fenótipo , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/mortalidade , Receptores CCR2/imunologia , Redução de Peso/genética , Redução de Peso/imunologia
20.
PLoS One ; 6(2): e16384, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21364979

RESUMO

Although culture-independent techniques have shown that the lungs are not sterile, little is known about the lung microbiome in chronic obstructive pulmonary disease (COPD). We used pyrosequencing of 16S amplicons to analyze the lung microbiome in two ways: first, using bronchoalveolar lavage (BAL) to sample the distal bronchi and air-spaces; and second, by examining multiple discrete tissue sites in the lungs of six subjects removed at the time of transplantation. We performed BAL on three never-smokers (NS) with normal spirometry, seven smokers with normal spirometry ("healthy smokers", HS), and four subjects with COPD (CS). Bacterial 16 s sequences were found in all subjects, without significant quantitative differences between groups. Both taxonomy-based and taxonomy-independent approaches disclosed heterogeneity in the bacterial communities between HS subjects that was similar to that seen in healthy NS and two mild COPD patients. The moderate and severe COPD patients had very limited community diversity, which was also noted in 28% of the healthy subjects. Both approaches revealed extensive membership overlap between the bacterial communities of the three study groups. No genera were common within a group but unique across groups. Our data suggests the existence of a core pulmonary bacterial microbiome that includes Pseudomonas, Streptococcus, Prevotella, Fusobacterium, Haemophilus, Veillonella, and Porphyromonas. Most strikingly, there were significant micro-anatomic differences in bacterial communities within the same lung of subjects with advanced COPD. These studies are further demonstration of the pulmonary microbiome and highlight global and micro-anatomic changes in these bacterial communities in severe COPD patients.


Assuntos
Saúde , Pulmão/microbiologia , Metagenoma , Doença Pulmonar Obstrutiva Crônica/microbiologia , Fumar/patologia , Adulto , Idoso , Líquido da Lavagem Broncoalveolar/microbiologia , Broncoscopia , DNA Bacteriano/análise , Feminino , Humanos , Pulmão/patologia , Masculino , Metagenoma/genética , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/patologia , RNA Ribossômico 16S/análise , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA