Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 859
Filter
1.
Front Cell Infect Microbiol ; 14: 1397940, 2024.
Article in English | MEDLINE | ID: mdl-38751999

ABSTRACT

Non-typeable Haemophilus influenzae (NTHi) and Moraxella catarrhalis (Mcat) are two common respiratory tract pathogens often associated with acute exacerbations in Chronic Obstructive Pulmonary Disease (COPD) as well as with otitis media (OM) in children. Although there is evidence that these pathogens can adopt persistence mechanisms such as biofilm formation, the precise means through which they contribute to disease severity and chronicity remains incompletely understood, posing challenges for their effective eradication. The identification of potential vaccine candidates frequently entails the characterization of the host-pathogen interplay in vitro even though this approach is limited by the fact that conventional models do not permit long term bacterial infections. In the present work, by using air-liquid-interface (ALI) human airway in vitro models, we aimed to recreate COPD-related persistent bacterial infections. In particular, we explored an alternative use of the ALI system consisting in the assembly of an inverted epithelium grown on the basal part of a transwell membrane with the aim to enable the functionality of natural defense mechanisms such as mucociliary clearance and cellular extrusion that are usually hampered during conventional ALI infection experiments. The inversion of the epithelium did not affect tissue differentiation and considerably delayed NTHi or Mcat infection progression, allowing one to monitor host-pathogen interactions for up to three weeks. Notably, the use of these models, coupled with confocal and transmission electron microscopy, revealed unique features associated with NTHi and Mcat infection, highlighting persistence strategies including the formation of intracellular bacterial communities (IBCs) and surface-associated biofilm-like structures. Overall, this study demonstrates the possibility to perform long term host-pathogen investigations in vitro with the aim to define persistence mechanisms adopted by respiratory pathogens and individuate potential new vaccine targets.


Subject(s)
Biofilms , Haemophilus influenzae , Moraxella catarrhalis , Moraxellaceae Infections , Moraxella catarrhalis/physiology , Humans , Haemophilus influenzae/physiology , Haemophilus influenzae/pathogenicity , Biofilms/growth & development , Moraxellaceae Infections/microbiology , Persistent Infection/microbiology , Host-Pathogen Interactions , Haemophilus Infections/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Models, Biological , Respiratory Tract Infections/microbiology , Epithelial Cells/microbiology
2.
Front Immunol ; 15: 1407439, 2024.
Article in English | MEDLINE | ID: mdl-38779669

ABSTRACT

Background: Increasing evidence indicates the microbial ecology of chronic obstructive pulmonary disease (COPD) is intricately associated with the disease's status and severity, and distinct microbial ecological variations exist between COPD and healthy control (HC). This systematic review and meta-analysis aimed to summarize microbial diversity indices and taxa relative abundance of oral, airway, and intestine microbiota of different stages of COPD and HC to make comparisons. Methods: A comprehensive systematic literature search was conducted in PubMed, Embase, the Web of Science, and the Cochrane Library databases to identify relevant English articles on the oral, airway, and intestine microbiota in COPD published between 2003 and 8 May 2023. Information on microbial diversity indices and taxa relative abundance of oral, airway, and intestine microbiota was collected for comparison between different stages of COPD and HC. Results: A total of 20 studies were included in this review, involving a total of 337 HC participants, 511 COPD patients, and 154 AECOPD patients. We observed that no significant differences in alpha diversity between the participant groups, but beta diversity was significantly different in half of the included studies. Compared to HC, Prevotella, Streptococcus, Actinomyces, and Veillonella of oral microbiota in SCOPD were reduced at the genus level. Most studies supported that Haemophilus, Lactobacillus, and Pseudomonas were increased, but Veillonella, Prevotella, Actinomyces, Porphyromonas, and Atopobium were decreased at the genus level in the airway microbiota of SCOPD. However, the abundance of Haemophilus, Lactobacillus and Pseudomonas genera exhibited an increase, whereas Actinomyces and Porphyromonas showed a decrease in the airway microbiota of AECOPD compared to HC. And Lachnospira of intestine microbiota in SCOPD was reduced at the genus level. Conclusion: The majority of published research findings supported that COPD exhibited decreased alpha diversity compared to HC. However, our meta-analysis does not confirm it. In order to further investigate the characteristics and mechanisms of microbiome in the oral-airway- intestine axis of COPD patients, larger-scale and more rigorous studies are needed. Systematic review registration: PROSPERO (https://www.crd.york.ac.uk/prospero/), identifier CRD42023418726.


Subject(s)
Gastrointestinal Microbiome , Pulmonary Disease, Chronic Obstructive , Pulmonary Disease, Chronic Obstructive/microbiology , Humans , Mouth/microbiology , Microbiota , Bacteria/classification , Bacteria/genetics
3.
Front Cell Infect Microbiol ; 14: 1379831, 2024.
Article in English | MEDLINE | ID: mdl-38746785

ABSTRACT

Objective: Exploring the effect of SJQJD on the pulmonary microbiota of chronic obstructive pulmonary disease (COPD) rats through 16S ribosomal RNA (rRNA) sequencing. Methods: A COPD rat model was constructed through smoking and lipopolysaccharide (LPS) stimulation, and the efficacy of SJQJD was evaluated by hematoxylin and eosin (H&E) staining and Enzyme-Linked Immunosorbnent Assay (ELISA). The alveolar lavage fluid of rats was subjected to 16S rRNA sequencing. The diversity of lung microbiota composition and community structure was analyzed and differential microbiota were screened. Additionally, machine learning algorithms were used for screening biomarkers of each group of the microbiota. Results: SJQJD could improve lung structure and inflammatory response in COPD rats. 16s rRNA sequencing analysis showed that SJQJD could significantly improve the abundance and diversity of bacterial communities in COPD rats. Through differential analysis and machine learning methods, potential microbial biomarkers were identified as Mycoplasmataceae, Bacillaceae, and Lachnospiraceae. Conclusion: SJQJD could improve tissue morphology and local inflammatory response in COPD rats, and its effect may be related to improve pulmonary microbiota.


Subject(s)
Disease Models, Animal , Drugs, Chinese Herbal , Lung , Microbiota , Pulmonary Disease, Chronic Obstructive , RNA, Ribosomal, 16S , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Microbiota/drug effects , Lung/microbiology , Lung/pathology , Rats , RNA, Ribosomal, 16S/genetics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Male , Bacteria/classification , Bacteria/genetics , Bacteria/drug effects , Bacteria/isolation & purification , Bronchoalveolar Lavage Fluid/microbiology , Rats, Sprague-Dawley
4.
Respir Res ; 25(1): 204, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730440

ABSTRACT

BACKGROUND: The impact of cigarette smoke (CS) on lung diseases and the role of microbiome dysbiosis in chronic obstructive pulmonary disease (COPD) have been previously reported; however, the relationships remain unclear. METHODS: Our research examined the effects of 20-week cigarette smoke (CS) exposure on the lung and intestinal microbiomes in C57BL/6JNarl mice, alongside a comparison with COPD patients' intestinal microbiome data from a public dataset. RESULTS: The study found that CS exposure significantly decreased forced vital capacity (FVC), thickened airway walls, and induced emphysema. Increased lung damage was observed along with higher lung keratinocyte chemoattractant (KC) levels by CS exposure. Lung microbiome analysis revealed a rise in Actinobacteriota, while intestinal microbiome showed significant diversity changes, indicating dysbiosis. Principal coordinate analysis highlighted distinct intestinal microbiome compositions between control and CS-exposed groups. In the intestinal microbiome, notable decreases in Patescibacteria, Campilobacterota, Defferibacterota, Actinobacteriota, and Desulfobacterota were observed. We also identified correlations between lung function and dysbiosis in both lung and intestinal microbiomes. Lung interleukins, interferon-É£, KC, and 8-isoprostane levels were linked to lung microbiome dysbiosis. Notably, dysbiosis patterns in CS-exposed mice were similar to those in COPD patients, particularly of Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage 4 patients. This suggests a systemic impact of CS exposure. CONCLUSION: In summary, CS exposure induces significant dysbiosis in lung and intestinal microbiomes, correlating with lung function decline and injury. These results align with changes in COPD patients, underscoring the important role of microbiome in smoke-related lung diseases.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Lung , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive , Animals , Pulmonary Disease, Chronic Obstructive/microbiology , Gastrointestinal Microbiome/physiology , Mice , Humans , Male , Lung/microbiology , Female , Middle Aged , Aged , Smoke/adverse effects
5.
Article in English | MEDLINE | ID: mdl-38596203

ABSTRACT

Introduction: Chronic obstructive pulmonary disease (COPD), an incurable chronic respiratory disease, has become a major public health problem. The relationship between the composition of intestinal microbiota and the important clinical factors affecting COPD remains unclear. This study aimed to identify specific intestinal microbiota with high clinical diagnostic value for COPD. Methods: The fecal microbiota of patients with COPD and healthy individuals were analyzed by 16S rDNA sequencing. Random forest classification was performed to analyze the different intestinal microbiota. Spearman correlation was conducted to analyze the correlation between different intestinal microbiota and clinical characteristics. A microbiota-disease network diagram was constructed using the gut MDisorder database to identify the possible pathogenesis of intestinal microorganisms affecting COPD, screen for potential treatment, and guide future research. Results: No significant difference in biodiversity was shown between the two groups but significant differences in microbial community structure. Fifteen genera of bacteria with large abundance differences were identified, including Bacteroides, Prevotella, Lachnospira, and Parabacteroides. Among them, the relative abundance of Lachnospira and Coprococcus was negatively related to the smoking index and positively related to lung function results. By contrast, the relative abundance of Parabacteroides was positively correlated with the smoking index and negatively correlated with lung function findings. Random forest classification showed that Lachnospira was the genus most capable of distinguishing between patients with COPD and healthy individuals suggesting it may be a potential biomarker of COPD. A Lachnospira disease network diagram suggested that Lachnospira decreased in some diseases, such as asthma, diabetes mellitus, and coronavirus disease 2019 (COVID-19), and increased in other diseases, such as irritable bowel syndrome, hypertension, and bovine lichen. Conclusion: The dominant intestinal microbiota with significant differences is related to the clinical characteristics of COPD, and the Lachnospira has the potential value to identify COPD.


Subject(s)
Asthma , Gastrointestinal Microbiome , Microbiota , Pulmonary Disease, Chronic Obstructive , Humans , Animals , Cattle , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/microbiology , Feces/microbiology
6.
Article in English | MEDLINE | ID: mdl-38650680

ABSTRACT

Introduction: The Lifei Decoction (LD) is a commonly utilized Chinese medicine for the treatment of sepsis and bronchial inflammation. However, its therapeutic potential in chronic obstructive pulmonary disease (COPD) remains unknown. Therefore, the objective of this study was to investigate the therapeutic efficacy and underlying mechanism of LD in a mouse model of COPD induced by cigarette smoke (CS) combined with lipopolysaccharide (LPS). Methods: Hematoxylin-eosin (H&E) staining was employed to observe the pathological alterations in lung tissue, while ELISA was utilized for the detection of levels of inflammatory factors in both lung tissue and bronchoalveolar lavage fluid (BALF). Additionally, Western blot analysis was conducted to assess the expression of p-NF-κB, GDF11, ZO-1, and Occludin-1 proteins. The changes in intestinal flora were evaluated using the viable bacteria count method. Results: The administration of LD demonstrates significant efficacy in mitigating pulmonary tissue damage in a murine model, while concurrently inhibiting the activation of the inflammatory pathway NF-κB to attenuate the levels of pro-inflammatory factors. Moreover, LD exhibits the capacity to enhance the expression of intestinal functional proteins ZO-1 and Occludin-1, thereby rectifying dysbiosis within the gut microbiota. Conclusion: The LD shows great promise as a potential treatment for COPD.


Subject(s)
Anti-Inflammatory Agents , Disease Models, Animal , Drugs, Chinese Herbal , Inflammation Mediators , Lipopolysaccharides , Lung , NF-kappa B , Occludin , Pulmonary Disease, Chronic Obstructive , Signal Transduction , Zonula Occludens-1 Protein , Animals , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/microbiology , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/microbiology , Drugs, Chinese Herbal/pharmacology , Zonula Occludens-1 Protein/metabolism , NF-kappa B/metabolism , Occludin/metabolism , Inflammation Mediators/metabolism , Anti-Inflammatory Agents/pharmacology , Male , Gastrointestinal Microbiome/drug effects , Mice, Inbred C57BL , Smoke/adverse effects , Bronchoalveolar Lavage Fluid , Cigarette Smoking/adverse effects , Mice
7.
Sci China Life Sci ; 67(5): 854-864, 2024 May.
Article in English | MEDLINE | ID: mdl-38265598

ABSTRACT

Tobacco smoking is a prevalent and detrimental habit practiced worldwide, increasing the risk of various diseases, including chronic obstructive pulmonary disease (COPD), cardiovascular disease, liver disease, and cancer. Although previous research has explored the detrimental health effects of tobacco smoking, recent studies suggest that gut microbiota dysbiosis may play a critical role in these outcomes. Numerous tobacco smoke components, such as nicotine, are found in the gastrointestinal tract and interact with gut microbiota, leading to lasting impacts on host health and diseases. This review delves into the ways tobacco smoking and its various constituents influence gut microbiota composition and functionality. We also summarize recent advancements in understanding how tobacco smoking-induced gut microbiota dysbiosis affects host health. Furthermore, this review introduces a novel perspective on how changes in gut microbiota following smoking cessation may contribute to withdrawal syndrome and the degree of health improvements in smokers.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Tobacco Smoking , Humans , Tobacco Smoking/adverse effects , Dysbiosis/microbiology , Nicotine/adverse effects , Nicotine/metabolism , Animals , Gastrointestinal Tract/microbiology , Smoking Cessation , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/etiology
8.
Int J Chron Obstruct Pulmon Dis ; 18: 2531-2542, 2023.
Article in English | MEDLINE | ID: mdl-38022823

ABSTRACT

Purpose: The aim of the study was to use RNA sequencing (RNA-seq) data of lung from chronic obstructive pulmonary disease (COPD) patients to identify the bacteria that are most commonly detected. Additionally, the study sought to investigate the differences in these infections between normal lung tissues and those affected by COPD. Patients and Methods: We re-analyzed RNA-seq data of lung from 99 COPD patients and 93 non-COPD smokers to determine the extent to which the metagenomes differed between the two groups and to assess the reliability of the metagenomes. We used unmapped reads in the RNA-seq data that were not aligned to the human reference genome to identify more common infections in COPD patients. Results: We identified 18 bacteria that exhibited significant differences between the COPD and non-COPD smoker groups. Among these, Yersinia enterocolitica was found to be more than 30% more abundant in COPD. Additionally, we observed difference in detection rate based on smoking history. To ensure the accuracy of our findings and distinguish them from false positives, we double-check the metagenomic profile using Basic Local Alignment Search Tool (BLAST). We were able to identify and remove specific species that might have been misclassified as other species in Kraken2 but were actually Staphylococcus aureus, as identified by BLAST analysis. Conclusion: This study highlighted the method of using unmapped reads, which were not typically used in sequencing data, to identify microorganisms present in patients with lung diseases such as COPD. This method expanded our understanding of the microbial landscape in COPD and provided insights into the potential role of microorganisms in disease development and progression.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/microbiology , Reproducibility of Results , Lung/microbiology , Bacteria/genetics , RNA , Sequence Analysis, RNA
10.
Int J Chron Obstruct Pulmon Dis ; 18: 1267-1276, 2023.
Article in English | MEDLINE | ID: mdl-37362620

ABSTRACT

Introduction: Increasing evidence suggests that seasonal changes can trigger the alternation of airway microbiome. However, the dynamics of the upper airway bacterial ecology of chronic obstructive pulmonary disease (COPD) patients across different seasons remains unclear. Methods: In this study, we present a 16S ribosomal RNA survey of the airway microbiome on 72 swab samples collected in different months (March, May, July, September, and November) in 2019 from 18 COPD patients and from six resampled patients in November in 2020. Results: Our study uncovered a dynamic airway microbiota where changes appeared to be associated with seasonal alternation in COPD patients. Twelve clusters of temporal patterns were displayed by differential and clustering analysis along the time course, systematically revealing distinct microbial taxa that prefer to grow in cool and warm seasons, respectively. Moreover, the upper airway microbiome composition was relatively stable in the same season in different years. Discussion: Given the tight association between airway microbiome and COPD disease progression, this study can provide useful information for clinically understanding the seasonal trend of disease phenotypes in COPD patients.


Subject(s)
Microbiota , Pulmonary Disease, Chronic Obstructive , Humans , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/microbiology , Seasons , Sputum/microbiology , Microbiota/genetics , Trachea
11.
Nat Med ; 29(7): 1750-1759, 2023 07.
Article in English | MEDLINE | ID: mdl-37349537

ABSTRACT

Exposure to environmental pollution influences respiratory health. The role of the airway microbial ecosystem underlying the interaction of exposure and respiratory health remains unclear. Here, through a province-wide chronic obstructive pulmonary disease surveillance program, we conducted a population-based survey of bacterial (n = 1,651) and fungal (n = 719) taxa and metagenomes (n = 1,128) from induced sputum of 1,651 household members in Guangdong, China. We found that cigarette smoking and higher PM2.5 concentration were associated with lung function impairment through the mediation of bacterial and fungal communities, respectively, and that exposure was associated with an enhanced inter-kingdom microbial interaction resembling the pattern seen in chronic obstructive pulmonary disease. Enrichment of Neisseria was associated with a 2.25-fold increased risk of high respiratory symptom burden, coupled with an elevation in Aspergillus, in association with occupational pollution. We developed an individualized microbiome-based health index, which covaried with exposure, respiratory symptoms and diseases, with potential generalizability to global datasets. Our results may inform environmental risk prevention and guide interventions that harness airway microbiome.


Subject(s)
Microbiota , Pulmonary Disease, Chronic Obstructive , Humans , Respiratory System , Pulmonary Disease, Chronic Obstructive/etiology , Pulmonary Disease, Chronic Obstructive/microbiology , Environmental Exposure/adverse effects , Sputum/microbiology
12.
Microbiol Spectr ; 11(1): e0386022, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36475849

ABSTRACT

Patients with chronic obstructive pulmonary disease (COPD) benefit from the immunomodulatory effect of azithromycin, but long-term administration may alter colonizing bacteria. Our goal was to identify changes in Haemophilus influenzae and Haemophilus parainfluenzae during azithromycin treatment. Fifteen patients were followed while receiving prolonged azithromycin treatment (Hospital Universitari de Bellvitge, Spain). Four patients (P02, P08, P11, and P13) were persistently colonized by H. influenzae for at least 3 months and two (P04 and P11) by H. parainfluenzae. Isolates from these patients (53 H. influenzae and 18 H. parainfluenzae) were included to identify, by whole-genome sequencing, antimicrobial resistance changes and genetic variation accumulated during persistent colonization. All persistent lineages isolated before treatment were azithromycin-susceptible but developed resistance within the first months, apart from those belonging to P02, who discontinued the treatment. H. influenzae isolates from P08-ST107 acquired mutations in 23S rRNA, and those from P11-ST2480 and P13-ST165 had changes in L4 and L22. In H. parainfluenzae, P04 persistent isolates acquired changes in rlmC, and P11 carried genes encoding MefE/MsrD efflux pumps in an integrative conjugative element, which was also identified in H. influenzae P11-ST147. Other genetic variation occurred in genes associated with cell wall and inorganic ion metabolism. Persistent H. influenzae strains all showed changes in licA and hgpB genes. Other genes (lex1, lic3A, hgpC, and fadL) had variation in multiple lineages. Furthermore, persistent strains showed loss, acquisition, or genetic changes in prophage-associated regions. Long-term azithromycin therapy results in macrolide resistance, as well as genetic changes that likely favor bacterial adaptation during persistent respiratory colonization. IMPORTANCE The immunomodulatory properties of azithromycin reduce the frequency of exacerbations and improve the quality of life of COPD patients. However, long-term administration may alter the respiratory microbiota, such as Haemophilus influenzae, an opportunistic respiratory colonizing bacteria that play an important role in exacerbations. This study contributes to a better understanding of COPD progression by characterizing the clinical evolution of H. influenzae in a cohort of patients with prolonged azithromycin treatment. The emergence of macrolide resistance during the first months, combined with the role of Haemophilus parainfluenzae as a reservoir and source of resistance dissemination, is a cause for concern that may lead to therapeutic failure. Furthermore, genetic variations in cell wall and inorganic ion metabolism coding genes likely favor bacterial adaptation to host selective pressures. Therefore, the bacterial pathoadaptive evolution in these severe COPD patients raise our awareness of the possible spread of macrolide resistance and selection of host-adapted clones.


Subject(s)
Haemophilus Infections , Pulmonary Disease, Chronic Obstructive , Humans , Azithromycin/therapeutic use , Azithromycin/pharmacology , Haemophilus/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Quality of Life , Haemophilus Infections/drug therapy , Haemophilus Infections/microbiology , Macrolides/pharmacology , Macrolides/therapeutic use , Drug Resistance, Bacterial/genetics , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/microbiology , Respiratory System , Haemophilus influenzae
13.
Am J Respir Cell Mol Biol ; 68(3): 245-255, 2023 03.
Article in English | MEDLINE | ID: mdl-36476129

ABSTRACT

Microorganisms colonize the human body. The lungs and respiratory tract, previously believed to be sterile, harbor diverse microbial communities and the genomes of bacteria (bacteriome), viruses (virome), and fungi (mycobiome). Recent advances in amplicon and shotgun metagenomic sequencing technologies and data-analyzing methods have greatly aided the identification and characterization of microbial populations from airways. The respiratory microbiome has been shown to play roles in human health and disease and is an area of rapidly emerging interest in pulmonary medicine. In this review, we provide updated information in the field by focusing on four lung conditions, including asthma, chronic obstructive pulmonary disease, cystic fibrosis, and idiopathic pulmonary fibrosis. We evaluate gut, oral, and upper airway microbiomes and how they contribute to lower airway flora. The discussion is followed by a systematic review of the lower airway microbiome in health and disease. We conclude with promising research avenues and implications for evolving therapeutics.


Subject(s)
Asthma , Cystic Fibrosis , Microbiota , Pulmonary Disease, Chronic Obstructive , Humans , Lung/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Cystic Fibrosis/microbiology
14.
mSphere ; 7(6): e0037722, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36342141

ABSTRACT

Inhaled corticosteroids (ICS) are commonly prescribed first-line treatments for asthma and chronic obstructive pulmonary disease (COPD). Recent evidence has shown that ICS use is associated with changes in the airway microbiome, which may impact clinical outcomes such as potential increased risk for pneumonia in COPD. Although the immunomodulatory effects of corticosteroids are well appreciated, whether ICS could directly influence the behavior of respiratory tract bacteria has been unknown. In this pilot study we explored the effects of fluticasone proprionate, a commonly prescribed inhaled corticosteroid, on respiratory bacteria with an expanded focus on Klebsiella pneumoniae, a species previously implicated in fluticasone-associated pneumonia in COPD. We observed significant effects of fluticasone proprionate on growth responses of K. pneumoniae, as well as other bacterial species isolated from asthmatic patients. Fluticasone-exposed K. pneumoniae displayed altered expression of several bacterial genes and reduced the metabolic activity of bronchial epithelial cells and their expression of human ß-defensin 2. Targeted assays identified a fluticasone metabolite from fluticasone-exposed K. pneumoniae cells, suggesting this species may be capable of metabolizing fluticasone proprionate. Collectively, these observations support the hypothesis that specific members of the airway microbiota possess the functional repertoire to respond to or potentially utilize corticosteroids in their microenvironment. These findings lay a foundation for novel research directions into the potential direct effects of ICS, often prescribed long term to patients, on the broader airway microbial community and on the behavior of specific microbial species implicated in asthma and COPD outcomes. IMPORTANCE Inhaled corticosteroids are widely prescribed for many respiratory diseases, including asthma and COPD. While they benefit many patients, corticosteroids can also have negative effects. Some patients do not improve with treatment and even experience adverse side effects. Recent studies have shown that inhaled corticosteroids can change the make-up of bacteria in the human respiratory tract. However, whether these medications can directly impact the behavior of such bacteria has been unknown. Here, we explored the effects of fluticasone propionate, a commonly prescribed inhaled corticosteroid, on Klebsiella pneumoniae and other airway bacteria of interest, including primary species isolated from adult asthma patients. We provide evidence of growth responses to direct fluticasone exposure in culture and further examined fluticasone's effects on K. pneumoniae, including gene expression changes and effects of fluticasone-exposed bacteria on airway cells. These findings indicate that members of the human airway bacterial community possess the functional ability to respond to corticosteroids, which may have implications for the heterogeneity of treatment response observed clinically.


Subject(s)
Asthma , Pneumonia , Pulmonary Disease, Chronic Obstructive , Humans , Fluticasone/adverse effects , Klebsiella pneumoniae , Pilot Projects , Asthma/drug therapy , Asthma/chemically induced , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/microbiology , Adrenal Cortex Hormones/adverse effects
17.
Jpn J Infect Dis ; 75(6): 560-568, 2022 Nov 22.
Article in English | MEDLINE | ID: mdl-35908874

ABSTRACT

Dynamic changes in the microbiome during chronic obstructive pulmonary disease (COPD) exacerbations remain unclear. Using 16S ribosomal DNA and fungal internal transcribed spacer DNA sequencing, we described the composition and changes in the bacterial and fungal microbiota of bronchoalveolar lavage fluid samples from 15 COPD patients and seven non-COPD patients. In patients with COPD, the dominant bacterial phyla were Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria. The most abundant fungal phyla were the Ascomycota and Basidiomycota. In terms of the genera of bacteria and fungi, the numbers of Veillonella, Prevotella-7, Haemophilus, and Aspergillus were significantly higher in patients with COPD than in the non-COPD patients. In addition, after the progression of COPD, the relative abundances of the fungal genera Aspergillus, Mortierella, Grifola, Thermoascus, Russula, and Thermomyces and the bacterial genus Proteobacteria increased significantly. Existing analyses have demonstrated changes in the diversity of bacterial and fungal communities, which appear to be related to COPD. Our results demonstrate the potential utility of microbiota as a possible biomarker for disease progression and provide therapeutic targets for COPD.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Humans , Bronchoalveolar Lavage Fluid/microbiology , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/analysis , Pulmonary Disease, Chronic Obstructive/microbiology , Bacteria/genetics , Proteobacteria/genetics , Fungi/genetics
18.
Life Sci ; 305: 120794, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-35835251

ABSTRACT

AIMS: Colonisation with non-typeable Haemophilus influenzae (NTHi) is common in COPD. Iron is required by bacteria for nutrition. Gallium is imported into bacteria using iron import proteins. Gallium cannot fulfill key metabolic functions, causing bactericidal effects. We tested the efficacy of gallium compounds as antimicrobials against NTHi in hemin rich conditions, and their ability to reduce NTHi induced pro-inflammatory responses in macrophages. MAIN METHODS: NTHi was cultured with the free iron analogue gallium nitrate (GaN) and heme iron analogue gallium protoporphyrin (GaPP) (0.5-4 µM; 24 h). Growth of NTHi reference strain (NCTC 12699) and 6 clinical isolates from COPD patients (including antibiotic resistant isolates) was assessed by optical density, and viability by Miles Misra. Monocyte derived macrophages (MDMs) were treated with GaPP before/after NTHi exposure. Viable intracellular NTHi was assessed by gentamicin protection assay. GaN or GaPP was added to NTHi cultures prior to culture with MDMs. Cytokine gene expression (qPCR) and protein secretion (ELISA) were measured. KEY FINDINGS: NTHi growth and viability were reduced by GaPP but not GaN. GaPP inhibited growth of COPD isolates (4 µM: 87 % reduction). GaPP reduced intracellular viability of NTHi in macrophage infection models. MDM cytokine gene expression and protein secretion (TNF-α, IL-6 and CXCL8) in response to NTHi was reduced (82, 66 and 86 % for gene expression) when cultured with GaPP 4 µM. SIGNIFICANCE: GaPP is an effective antimicrobial for NTHi while GaN showed no effect on growth or viability. Culture of NTHi with GaPP also reduced the pro-inflammatory cytokine response in MDMs.


Subject(s)
Gallium , Pulmonary Disease, Chronic Obstructive , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cytokines/pharmacology , Gallium/pharmacology , Gallium/therapeutic use , Haemophilus influenzae/metabolism , Humans , Iron/metabolism , Protoporphyrins/pharmacology , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/microbiology
19.
Article in English | MEDLINE | ID: mdl-35480555

ABSTRACT

Purpose: Data regarding the relationship between microbiologic features and comorbidities in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD) are limited. The aim of this study was to correlate microbiologic findings with comorbidities in patients with moderate to severe AECOPD. Patients and Methods: This multicenter observational study included patients with AECOPD seen at 28 hospitals in South Korea between January 2015 and December 2018, and the data were retrospectively collected. Pathogens were examined in patients with either pulmonary or extrapulmonary comorbidities, and compared to those of patients without comorbidities. The relationship between pathogen type and the number of comorbidities was also evaluated. Results: Bacterial infections (178 [37.2%] vs 203 [28.7%], p = 0.002) and co-infections with bacteria and viruses (65 [13.6%] vs 57 [8.1%], p = 0.002) were more prevalent in patients with pulmonary comorbidities. Bacterial pathogens (280 [34.7%] vs 101 [26.7%], p=0.006) were detected at a higher rate in patients with extrapulmonary comorbidities. Previous pulmonary tuberculosis (PTB), bronchiectasis, and diabetes mellitus were risk factors for bacterial infection, and congestive heart failure was a risk factor for bacterial and viral co-infection. As the number of comorbidities increased, the risk of bacterial infection increased considerably. Pseudomonas aeruginosa was more frequently identified in patients with previous PTB (57 [15.3%] vs 59 [7.4%], p < 0.001) and bronchiectasis (33 [19.6%] vs 83 [8.3%], p < 0.001). Conclusion: AECOPD patients with comorbidities were more likely to experience infection-related exacerbations compared to those without comorbidities. As the overall number of comorbidities increased, the risk of bacterial infection increased significantly.


Subject(s)
Bacterial Infections , Bronchiectasis , Coinfection , Pulmonary Disease, Chronic Obstructive , Virus Diseases , Bacteria , Bacterial Infections/diagnosis , Bacterial Infections/epidemiology , Bacterial Infections/microbiology , Humans , Lung , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/microbiology , Retrospective Studies
20.
Indian J Med Microbiol ; 40(2): 223-227, 2022.
Article in English | MEDLINE | ID: mdl-35292163

ABSTRACT

PURPOSE: This study was planned to determine the trends and susceptibility pattern of invasive pulmonary aspergillosis (IPA) in severely ill chronic obstructive pulmonary disease (COPD) patients admitted in pulmonary ward and ICU of our tertiary care centre. METHODS: Fifty COPD patients suspected of IPA from pulmonary ward and ICU from April 2017 to September 2018 were investigated. Samples were processed by standard methods, culture positive isolates were confirmed by MALDI-TOF MS and antifungal susceptibility testing was performed by microbroth dilution method. RESULTS: Twenty-two critically ill COPD patients were microbiologically positive for IA infection, of which 13 were classified as putative invasive aspergillosis. The most common comorbid illness associated was diabetes. A. flavus and A. fumigatus were the commonest species isolated. The minimum inhibitory concentration of the antifungals was low. Morbidity due to IPA in COPD patients was very high. CONCLUSIONS: Prevalence of IPA in the pulmonary ward and ICU was found to be 9.6%. MALDI-TOF seems to be a promising tool for aiding rapid identification especially for slow growing and non-sporulating fungi. Heightened awareness and suspicion for pulmonary mould infections along with early diagnosis can substantially alter the patient prognosis.


Subject(s)
Aspergillosis , Invasive Pulmonary Aspergillosis , Pulmonary Disease, Chronic Obstructive , Critical Illness , Humans , Intensive Care Units , Invasive Pulmonary Aspergillosis/diagnosis , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...