Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.242
Filter
1.
Nat Commun ; 15(1): 4889, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849369

ABSTRACT

Polymicrobial infection of the airways is a hallmark of obstructive lung diseases such as cystic fibrosis (CF), non-CF bronchiectasis, and chronic obstructive pulmonary disease. Pulmonary exacerbations (PEx) in these conditions are associated with accelerated lung function decline and higher mortality rates. Understanding PEx ecology is challenged by high inter-patient variability in airway microbial community profiles. We analyze bacterial communities in 880 CF sputum samples collected during an observational prospective cohort study and develop microbiome descriptors to model community reorganization prior to and during 18 PEx. We identify two microbial dysbiosis regimes with opposing ecology and dynamics. Pathogen-governed PEx show hierarchical community reorganization and reduced diversity, whereas anaerobic bloom PEx display stochasticity and increased diversity. A simulation of antimicrobial treatment predicts better efficacy for hierarchically organized communities. This link between PEx, microbiome organization, and treatment success advances the development of personalized clinical management in CF and, potentially, other obstructive lung diseases.


Subject(s)
Cystic Fibrosis , Dysbiosis , Microbiota , Sputum , Cystic Fibrosis/microbiology , Humans , Male , Sputum/microbiology , Prospective Studies , Female , Treatment Outcome , Dysbiosis/microbiology , Adult , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Lung/microbiology , Disease Progression , Pulmonary Disease, Chronic Obstructive/microbiology , Young Adult , Adolescent , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
2.
Med Mycol J ; 65(2): 29-32, 2024.
Article in English | MEDLINE | ID: mdl-38825527

ABSTRACT

Mucormycosis is a fungal infectious disease caused by Rhizopus oryzae and other members of the order Mucorales, and it is known as one of the most lethal fungal infections. Early diagnosis of mucormycosis improves prognosis because of limited effective treatments and the rapid progression of the disease. On the other hand, the lack of characteristic clinical findings in mucormycosis and the challenge of early definitive diagnosis make early treatment difficult. Our goal was to establish a serodiagnostic method to detect Rhizopus specific antigen (RSA), and we have developed a diagnostic kit by Enzyme-linked immuno-sorbent assay (ELISA) using a monoclonal antibody against this antigen. RSA increased over time in the serum and alveolar lavage fluid of R. oryzae-infected mice. RSA was also detected in serum and alveolar fluid, even at an early stage (Day 1), when the tissue invasion of R. oryzae mycelium was not histopathologically detectable in the lungs of R. oryzae-infected mice. Further evaluation is needed to determine the feasibility of using this assay in clinical practice.


Subject(s)
Antigens, Fungal , Biomarkers , Enzyme-Linked Immunosorbent Assay , Mucormycosis , Rhizopus oryzae , Mucormycosis/diagnosis , Animals , Mice , Antigens, Fungal/immunology , Antigens, Fungal/blood , Biomarkers/blood , Bronchoalveolar Lavage Fluid/microbiology , Disease Models, Animal , Antibodies, Monoclonal , Rhizopus/isolation & purification , Lung/microbiology , Lung/pathology , Humans , Serologic Tests/methods
3.
Nat Commun ; 15(1): 4708, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38830853

ABSTRACT

Critical illness can significantly alter the composition and function of the human microbiome, but few studies have examined these changes over time. Here, we conduct a comprehensive analysis of the oral, lung, and gut microbiota in 479 mechanically ventilated patients (223 females, 256 males) with acute respiratory failure. We use advanced DNA sequencing technologies, including Illumina amplicon sequencing (utilizing 16S and ITS rRNA genes for bacteria and fungi, respectively, in all sample types) and Nanopore metagenomics for lung microbiota. Our results reveal a progressive dysbiosis in all three body compartments, characterized by a reduction in microbial diversity, a decrease in beneficial anaerobes, and an increase in pathogens. We find that clinical factors, such as chronic obstructive pulmonary disease, immunosuppression, and antibiotic exposure, are associated with specific patterns of dysbiosis. Interestingly, unsupervised clustering of lung microbiota diversity and composition by 16S independently predicted survival and performed better than traditional clinical and host-response predictors. These observations are validated in two separate cohorts of COVID-19 patients, highlighting the potential of lung microbiota as valuable prognostic biomarkers in critical care. Understanding these microbiome changes during critical illness points to new opportunities for microbiota-targeted precision medicine interventions.


Subject(s)
COVID-19 , Dysbiosis , Gastrointestinal Microbiome , Lung , Microbiota , Humans , Female , Male , Dysbiosis/microbiology , Middle Aged , Lung/microbiology , COVID-19/microbiology , COVID-19/virology , Aged , Microbiota/genetics , Gastrointestinal Microbiome/genetics , Host Microbial Interactions/genetics , Longitudinal Studies , RNA, Ribosomal, 16S/genetics , Respiratory Insufficiency/microbiology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Adult , Respiration, Artificial , Bacteria/genetics , Bacteria/classification , Bacteria/isolation & purification , Critical Illness , Metagenomics/methods
4.
Front Cell Infect Microbiol ; 14: 1348685, 2024.
Article in English | MEDLINE | ID: mdl-38841114

ABSTRACT

Background: The microbiota-gut-lung axis has elucidated a potential association between gut microbiota and idiopathic pulmonary fibrosis (IPF). However, there is a paucity of population-level studies with providing robust evidence for establishing causality. This two-sample Mendelian randomization (MR) analysis aimed to investigate the causal relationship between the gut microbiota and IPF as well as lung function. Materials and methods: Adhering to Mendel's principle of inheritance, this MR analysis utilized summary-level data from respective genome-wide association studies (GWAS) involving 211 gut microbial taxa, IPF, and lung function indicators such as FEV1, FVC, and FEV1/FVC. A bidirectional two-sample MR design was employed, utilizing multiple MR analysis methods, including inverse variance-weighted (IVW), weighted median, MR-Egger, and weighted mode. Multivariable MR (MVMR) was used to uncover mediating factors connecting the exposure and outcome. Additionally, comprehensive sensitivity analyses were conducted to ensure the robustness of the results. Results: The MR results confirmed four taxa were found causally associated with the risk of IPF. Order Bifidobacteriales (OR=0.773, 95% CI: 0.610-0.979, p=0.033), Family Bifidobacteriaceae (OR=0.773, 95% CI: 0.610-0.979, p=0.033), and Genus RuminococcaceaeUCG009 (OR=0.793, 95% CI: 0.652-0.965, p=0.020) exerted protective effects on IPF, while Genus Coprococcus2 (OR=1.349, 95% CI: 1.021-1.783, p=0.035) promote the development of IPF. Several taxa were causally associated with lung function, with those in Class Deltaproteobacteria, Order Desulfovibrionales, Family Desulfovibrionaceae, Class Verrucomicrobiae, Order Verrucomicrobiales and Family Verrucomicrobiaceae being the most prominent beneficial microbiota, while those in Family Lachnospiraceae, Genus Oscillospira, and Genus Parasutterella were associated with impaired lung function. As for the reverse analysis, MR results confirmed the effects of FEV1 and FVC on the increased abundance of six taxa (Phylum Actinobacteria, Class Actinobacteria, Order Bifidobacteriales, Family Bifidobacteriaceae, Genus Bifidobacterium, and Genus Ruminiclostridium9) with a boosted level of evidence. MVMR suggested monounsaturated fatty acids, total fatty acids, saturated fatty acids, and ratio of omega-6 fatty acids to total fatty acids as potential mediating factors in the genetic association between gut microbiota and IPF. Conclusion: The current study suggested the casual effects of the specific gut microbes on the risk of IPF and lung function. In turn, lung function also exerted a positive role in some gut microbes. A reasonable dietary intake of lipid substances has a certain protective effect against the occurrence and progression of IPF. This study provides novel insights into the potential role of gut microbiota in IPF and indicates a possible gut microbiota-mediated mechanism for the prevention of IPF.


Subject(s)
Gastrointestinal Microbiome , Genome-Wide Association Study , Idiopathic Pulmonary Fibrosis , Lung , Mendelian Randomization Analysis , Humans , Idiopathic Pulmonary Fibrosis/microbiology , Gastrointestinal Microbiome/genetics , Lung/microbiology , Respiratory Function Tests , Genetic Predisposition to Disease
5.
Front Cell Infect Microbiol ; 14: 1392015, 2024.
Article in English | MEDLINE | ID: mdl-38841113

ABSTRACT

Trehalose-6-phosphate synthase (TPS1) was identified as a virulence factor for Cryptococcus neoformans and a promising therapeutic target. This study reveals previously unknown roles of TPS1 in evasion of host defenses during pulmonary and disseminated phases of infection. In the pulmonary infection model, TPS1-deleted (tps1Δ) Cryptococci are rapidly cleared by mouse lungs whereas TPS1-sufficent WT (H99) and revertant (tps1Δ:TPS1) strains expand in the lungs and disseminate, causing 100% mortality. Rapid pulmonary clearance of tps1Δ mutant is T-cell independent and relies on its susceptibility to lung resident factors and innate immune factors, exemplified by tps1Δ but not H99 inhibition in a coculture with dispersed lung cells and its rapid clearance coinciding with innate leukocyte infiltration. In the disseminated model of infection, which bypasses initial lung-fungus interactions, tps1Δ strain remains highly attenuated. Specifically, tps1Δ mutant is unable to colonize the lungs from the bloodstream or expand in spleens but is capable of crossing into the brain, where it remains controlled even in the absence of T cells. In contrast, strains H99 and tps1Δ:TPS1 rapidly expand in all studied organs, leading to rapid death of the infected mice. Since the rapid pulmonary clearance of tps1Δ mutant resembles a response to acapsular strains, the effect of tps1 deletion on capsule formation in vitro and in vivo was examined. Tps1Δ cryptococci form capsules but with a substantially reduced size. In conclusion, TPS1 is an important virulence factor, allowing C. neoformans evasion of resident pulmonary and innate defense mechanisms, most likely via its role in cryptococcal capsule formation.


Subject(s)
Cryptococcosis , Cryptococcus neoformans , Disease Models, Animal , Glucosyltransferases , Lung , Virulence Factors , Animals , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/genetics , Cryptococcus neoformans/enzymology , Cryptococcus neoformans/immunology , Cryptococcosis/microbiology , Cryptococcosis/immunology , Mice , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Lung/microbiology , Lung/pathology , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism , Host-Pathogen Interactions , Brain/microbiology , Spleen/microbiology , Female , Mice, Inbred C57BL , Immunity, Innate , Immune Evasion , Gene Deletion
7.
Front Immunol ; 15: 1398369, 2024.
Article in English | MEDLINE | ID: mdl-38835759

ABSTRACT

Introduction: Although many studies have underscored the importance of T cells, phenotypically and functionally, fewer have studied the functions of myeloid cells in COVID disease. In particular, the potential role of myeloid cells such as monocytes and low-density neutrophils (LDNs) in innate responses and particular in the defense against secondary bacterial infections has been much less documented. Methods: Here, we compared, in a longitudinal study, healthy subjects, idiopathic fibrosis patients, COVID patients who were either hospitalized/moderate (M-) or admitted to ICU (COV-ICU) and patients in ICU hospitalized for other reasons (non-COV-ICU). Results: We show that COVID patients have an increased proportion of low-density neutrophils (LDNs), which produce high levels of proteases (particularly, NE, MMP-8 and MMP-9) (unlike non-COV-ICU patients), which are partly responsible for causing type II alveolar cell damage in co-culture experiments. In addition, we showed that M- and ICU-COVID monocytes had reduced responsiveness towards further live Pseudomonas aeruginosa (PAO1 strain) infection, an important pathogen colonizing COVID patients in ICU, as assessed by an impaired secretion of myeloid cytokines (IL-1, TNF, IL-8,…). By contrast, lymphoid cytokines (in particular type 2/type 3) levels remained high, both basally and post PAO1 infection, as reflected by the unimpaired capacity of T cells to proliferate, when stimulated with anti-CD3/CD28 beads. Discussion: Overall, our results demonstrate that COVID circulatory T cells have a biased type 2/3 phenotype, unconducive to proper anti-viral responses and that myeloid cells have a dual deleterious phenotype, through their LDN-mediated damaging effect on alveolar cells and their impaired responsiveness (monocyte-mediated) towards bacterial pathogens such as P. aeruginosa.


Subject(s)
COVID-19 , Monocytes , Neutrophils , Pseudomonas Infections , Pseudomonas aeruginosa , SARS-CoV-2 , Humans , COVID-19/immunology , Pseudomonas aeruginosa/immunology , Monocytes/immunology , Male , Female , Middle Aged , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Pseudomonas Infections/immunology , Neutrophils/immunology , Aged , Cytokines/metabolism , Cytokines/immunology , Adult , Longitudinal Studies , Leukocytes, Mononuclear/immunology , Lung/immunology , Lung/pathology , Lung/microbiology
8.
Front Cell Infect Microbiol ; 14: 1385562, 2024.
Article in English | MEDLINE | ID: mdl-38846353

ABSTRACT

Background: Lower respiratory tract infections represent prevalent ailments. Nonetheless, current comprehension of the microbial ecosystems within the lower respiratory tract remains incomplete and necessitates further comprehensive assessment. Leveraging the advancements in metagenomic next-generation sequencing (mNGS) technology alongside the emergence of machine learning, it is now viable to compare the attributes of lower respiratory tract microbial communities among patients across diverse age groups, diseases, and infection types. Method: We collected bronchoalveolar lavage fluid samples from 138 patients diagnosed with lower respiratory tract infections and conducted mNGS to characterize the lung microbiota. Employing various machine learning algorithms, we investigated the correlation of key bacteria in patients with concurrent bronchiectasis and developed a predictive model for hospitalization duration based on these identified key bacteria. Result: We observed variations in microbial communities across different age groups, diseases, and infection types. In the elderly group, Pseudomonas aeruginosa exhibited the highest relative abundance, followed by Corynebacterium striatum and Acinetobacter baumannii. Methylobacterium and Prevotella emerged as the dominant genera at the genus level in the younger group, while Mycobacterium tuberculosis and Haemophilus influenzae were prevalent species. Within the bronchiectasis group, dominant bacteria included Pseudomonas aeruginosa, Haemophilus influenzae, and Klebsiella pneumoniae. Significant differences in the presence of Pseudomonas phage JBD93 were noted between the bronchiectasis group and the control group. In the group with concomitant fungal infections, the most abundant genera were Acinetobacter and Pseudomonas, with Acinetobacter baumannii and Pseudomonas aeruginosa as the predominant species. Notable differences were observed in the presence of Human gammaherpesvirus 4, Human betaherpesvirus 5, Candida albicans, Aspergillus oryzae, and Aspergillus fumigatus between the group with concomitant fungal infections and the bacterial group. Machine learning algorithms were utilized to select bacteria and clinical indicators associated with hospitalization duration, confirming the excellent performance of bacteria in predicting hospitalization time. Conclusion: Our study provided a comprehensive description of the microbial characteristics among patients with lower respiratory tract infections, offering insights from various perspectives. Additionally, we investigated the advanced predictive capability of microbial community features in determining the hospitalization duration of these patients.


Subject(s)
Bacteria , Bronchoalveolar Lavage Fluid , High-Throughput Nucleotide Sequencing , Machine Learning , Metagenomics , Microbiota , Respiratory Tract Infections , Humans , Metagenomics/methods , Middle Aged , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Aged , Male , Female , Adult , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bronchoalveolar Lavage Fluid/microbiology , Microbiota/genetics , Young Adult , Bronchiectasis/microbiology , Aged, 80 and over , Metagenome , Adolescent , Lung/microbiology , Lung/virology , Hospitalization
9.
Front Immunol ; 15: 1360412, 2024.
Article in English | MEDLINE | ID: mdl-38745652

ABSTRACT

A robust immune response is required for resistance to pulmonary tuberculosis (TB), the primary disease caused by Mycobacterium tuberculosis (Mtb). However, pharmaceutical inhibition of T cell immune checkpoint molecules can result in the rapid development of active disease in latently infected individuals, indicating the importance of T cell immune regulation. In this study, we investigated the potential role of CD200R during Mtb infection, a key immune checkpoint for myeloid cells. Expression of CD200R was consistently downregulated on CD14+ monocytes in the blood of subjects with active TB compared to healthy controls, suggesting potential modulation of this important anti-inflammatory pathway. In homogenized TB-diseased lung tissue, CD200R expression was highly variable on monocytes and CD11b+HLA-DR+ macrophages but tended to be lowest in the most diseased lung tissue sections. This observation was confirmed by fluorescent microscopy, which showed the expression of CD200R on CD68+ macrophages surrounding TB lung granuloma and found expression levels tended to be lower in macrophages closest to the granuloma core and inversely correlated with lesion size. Antibody blockade of CD200R in a biomimetic 3D granuloma-like tissue culture system led to significantly increased Mtb growth. In addition, Mtb infection in this system reduced gene expression of CD200R. These findings indicate that regulation of myeloid cells via CD200R is likely to play an important part in the immune response to TB and may represent a potential target for novel therapeutic intervention.


Subject(s)
Mycobacterium tuberculosis , Myeloid Cells , Tuberculosis, Pulmonary , Humans , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Orexin Receptors/metabolism , Macrophages/immunology , Macrophages/metabolism , Adult , Female , Male , Antigens, CD/metabolism , Antigens, CD/genetics , Middle Aged , Lung/immunology , Lung/microbiology , Lung/pathology , Lung/metabolism , Biomimetics , Monocytes/immunology , Monocytes/metabolism
11.
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
12.
Int J Mycobacteriol ; 13(1): 40-46, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38771278

ABSTRACT

BACKGROUND: Tuberculosis (TB) is one of the leading infectious causes of mortality globally. The purpose of this research is to examine the clinical and radiological characteristics of patients with TB and diabetes. METHODS: The research comprised 276 TB patients, 52 of whom were diabetic and 224 of whom were not. During the evaluation of the patients' clinical histories, age, gender, diagnostic indicator, and whether or not they had undergone prior treatment were questioned, as were the requirement of inpatient treatment and the existence of drug resistance. Radiographically, they were questioned in terms of bilateral-unilateral extent, percentage of parenchymal involvement, cavitation, tree-in-bud appearance, the presence of ground glass, consolidation, miliary involvement, sequela fibrotic changes, parenchymal calcification, mediastinal lymphadenopathy, pleural effusion, and pleural calcification. In addition, segmenting was used to assess involvement in the affected lobes. RESULTS: When we look at the results of 276 patients, 182 males and 94 females, the mean age is 46.01 ± 17.83. Diabetes and TB coexistence are more prevalent in male individuals (P = 0.029). Smear positivity and the need for inpatient treatment were found to be higher in the clinical features of diabetic patients (P = 0.05 and P = 0.01, respectively). Radiologically, diabetes individuals are more likely to have larger mediastinal lymph nodes (P = 0.032). CONCLUSION: In the coexistence of both TB and diabetes, there are variations in radiological findings, complexity in treatment response, and patient management.


Subject(s)
Tomography, X-Ray Computed , Tuberculosis, Pulmonary , Humans , Male , Female , Middle Aged , Tuberculosis, Pulmonary/diagnostic imaging , Tuberculosis, Pulmonary/complications , Tuberculosis, Pulmonary/microbiology , Adult , Aged , Diabetes Complications/diagnostic imaging , Lung/diagnostic imaging , Lung/pathology , Lung/microbiology , Diabetes Mellitus , Young Adult
13.
Microb Pathog ; 191: 106678, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718954

ABSTRACT

A conditionally pathogenic bacterium called Bibersteinia trehalosi inhabits the upper respiratory tract of ruminants and is becoming a significant cause of pneumonia, especially in goats. In this study, we identified a gram-negative bacteria strain isolated from dead goat's lungs, which was named M01. By integrating the outcomes of its morphological and biochemical characterization with the investigation of the 16S rRNA gene sequence analysis, the isolate was identified as B. trehalosi. Based on antibiotic susceptibility tests, the isolate was shown to be resistant to ß-lactams, tetracyclines, and amphenicols. Its genome was discovered to comprise 2115 encoded genes and a circular chromosome measuring 2,345,568 bp using whole genome sequencing. Annotation of the VFBD database revealed that isolate M01 had four virulence genes encoding three virulence factors. The CARD database revealed that its genome has two antibiotic-resistance genes. Based on pathogenicity testing, isolate M01 was highly pathogenic to mice, primarily causing pneumonia, with an LD50 of 1.31 × 107 CFU/ml. Moreover, histopathology showed loss of alveolar structure and infiltration of lung inflammatory cells. Hence, the current study could provide sufficient information for prevention and control strategies for future epidemics of B. trehalosi in goat species.


Subject(s)
Anti-Bacterial Agents , Genome, Bacterial , Goats , Lung , Microbial Sensitivity Tests , RNA, Ribosomal, 16S , Virulence Factors , Animals , Goats/microbiology , RNA, Ribosomal, 16S/genetics , Mice , Anti-Bacterial Agents/pharmacology , Lung/microbiology , Lung/pathology , Virulence Factors/genetics , Goat Diseases/microbiology , Whole Genome Sequencing , Phylogeny , Virulence , Drug Resistance, Bacterial , DNA, Bacterial/genetics
14.
Sci Rep ; 14(1): 9998, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38693196

ABSTRACT

It is estimated that more than half of the world population has been infected with Helicobacter pylori. Most newly acquired H. pylori infections occur in children before 10 years of age. We hypothesized that early life H. pylori infection could influence the composition of the microbiome at mucosal sites distant to the stomach. To test this hypothesis, we utilized the infant rhesus macaque monkey as an animal model of natural H. pylori colonization to determine the impact of infection on the lung and oral microbiome during a window of postnatal development. From a cohort of 4-7 month-old monkeys, gastric biopsy cultures identified 44% of animals infected by H. pylori. 16S ribosomal RNA gene sequencing of lung washes and buccal swabs from animals showed distinct profiles for the lung and oral microbiome, independent of H. pylori infection. In order of relative abundance, the lung microbiome was dominated by the phyla Proteobacteria, Firmicutes, Bacteroidota, Fusobacteriota, Campilobacterota and Actinobacteriota while the oral microbiome was dominated by Proteobacteria, Firmicutes, Bacteroidota, and Fusobacteriota. In comparison to the oral cavity, the lung was composed of more genera and species that significantly differed by H. pylori status, with a total of 6 genera and species that were increased in H. pylori negative infant monkey lungs. Lung, but not plasma IL-8 concentration was also associated with gastric H. pylori load and lung microbial composition. We found the infant rhesus macaque monkey lung harbors a microbiome signature that is distinct from that of the oral cavity during postnatal development. Gastric H. pylori colonization and IL-8 protein were linked to the composition of microbial communities in the lung and oral cavity. Collectively, these findings provide insight into how H. pylori infection might contribute to the gut-lung axis during early childhood and modulate future respiratory health.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Lung , Macaca mulatta , Microbiota , Mouth , RNA, Ribosomal, 16S , Animals , Macaca mulatta/microbiology , Lung/microbiology , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Mouth/microbiology , RNA, Ribosomal, 16S/genetics , Male , Disease Models, Animal
15.
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
16.
Mycopathologia ; 189(3): 42, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709375

ABSTRACT

Pneumocystis pneumonia is a serious lung infection caused by an original ubiquitous fungus with opportunistic behavior, referred to as Pneumocystis jirovecii. P. jirovecii is the second most common fungal agent among invasive fungal infections after Candida spp. Unfortunately, there is still an inability to culture P. jirovecii in vitro, and so a great impairment to improve knowledge on the pathogenesis of Pneumocystis pneumonia. In this context, animal models have a high value to address complex interplay between Pneumocystis and the components of the host immune system. Here, we propose a protocol for a murine model of Pneumocystis pneumonia. Animals become susceptible to Pneumocystis by acquiring an immunocompromised status induced by iterative administration of steroids within drinking water. Thereafter, the experimental infection is completed by an intranasal challenge with homogenates of mouse lungs containing Pneumocystis murina. The onset of clinical signs occurs within 5 weeks following the infectious challenge and immunosuppression can then be withdrawn. At termination, lungs and bronchoalveolar lavage (BAL) fluids from infected mice are analyzed for fungal load (qPCR) and immune response (flow cytometry and biochemical assays). The model is a useful tool in studies focusing on immune responses initiated after the establishment of Pneumocystis pneumonia.


Subject(s)
Bronchoalveolar Lavage Fluid , Disease Models, Animal , Lung , Pneumonia, Pneumocystis , Animals , Pneumonia, Pneumocystis/microbiology , Pneumonia, Pneumocystis/pathology , Pneumonia, Pneumocystis/immunology , Bronchoalveolar Lavage Fluid/microbiology , Lung/microbiology , Lung/pathology , Mice , Pneumocystis , Colony Count, Microbial , Pneumocystis carinii , Immunocompromised Host
17.
Trials ; 25(1): 298, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698471

ABSTRACT

BACKGROUND: The use of antibiotics is a key driver of antimicrobial resistance and is considered a major threat to global health. In Denmark, approximately 75% of antibiotic prescriptions are issued in general practice, with acute lower respiratory tract infections (LRTIs) being one of the most common indications. Adults who present to general practice with symptoms of acute LRTI often suffer from self-limiting viral infections. However, some patients have bacterial community-acquired pneumonia (CAP), a potential life-threatening infection, that requires immediate antibiotic treatment. Importantly, no single symptom or specific point-of-care test can be used to discriminate the various diagnoses, and diagnostic uncertainty often leads to (over)use of antibiotics. At present, general practitioners (GPs) lack tools to better identify those patients who will benefit from antibiotic treatment. The primary aim of the PLUS-FLUS trial is to determine whether adults who present with symptoms of an acute LRTI in general practice and who have FLUS performed in addition to usual care are treated less frequently with antibiotics than those who only receive usual care. METHODS: Adults (≥ 18 years) presenting to general practice with acute cough (< 21 days) and at least one other symptom of acute LRTI, where the GP suspects a bacterial CAP, will be invited to participate in this pragmatic randomized controlled trial. All participants will receive usual care. Subsequently, participants will be randomized to either the control group (usual care) or to an additional focused lung ultrasonography performed by the GP (+ FLUS). The primary outcome is the proportion of participants with antibiotics prescribed at the index consultation (day 0). Secondary outcomes include comparisons of the clinical course for participants in groups. DISCUSSION: We will examine whether adults who present with symptoms of acute LRTI in general practice, who have FLUS performed in addition to usual care, have antibiotics prescribed less frequently than those given usual care alone. It is highly important that a possible reduction in antibiotic prescriptions does not compromise patients' recovery or clinical course, which we will assess closely. TRIAL REGISTRATION: ClinicalTrials.gov NCT06210282. Registered on January 17, 2024.


Subject(s)
Anti-Bacterial Agents , General Practice , Lung , Practice Patterns, Physicians' , Pragmatic Clinical Trials as Topic , Respiratory Tract Infections , Ultrasonography , Humans , Anti-Bacterial Agents/therapeutic use , Denmark , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/diagnostic imaging , Respiratory Tract Infections/microbiology , Lung/diagnostic imaging , Lung/microbiology , Acute Disease , Treatment Outcome , Drug Prescriptions , Point-of-Care Testing , Adult
18.
Front Cell Infect Microbiol ; 14: 1367885, 2024.
Article in English | MEDLINE | ID: mdl-38784566

ABSTRACT

Objective: This study aims to investigate the clinical application value of Metagenome Next-Generation Sequencing (mNGS) for pulmonary diffuse exudative lesions. Methods: From January 1, 2014, to November 31, 2021, 136 cases with chest radiologic presentations of pulmonary diffuse exudative lesions admitted to Fujian Provincial Hospital were included in the study; of those, 77 patients underwent mNGS pathogen detection. Based on the pathogen detection outcomes and clinical diagnoses, patients were categorized into an infection group (IG) and a non-infection group (NIG). A comparison was made between the diagnostic efficacy of the mNGS technique and traditional culture methods. Meanwhile, 59 patients clinically identified as having infectious pulmonary diffuse exudative lesions but who did not receive mNGS testing were designated as the non-NGS infection group (non-IG). A retrospective cohort study was conducted on patients in both the IG and non-IG, with a 30-day all-cause mortality endpoint used for follow-up. Outcomes: When compared to conventional culture methods, mNGS demonstrated an approximate 35% increase in sensitivity (80.0% vs 45.5%, P<0.001), without significant disparity in specificity (77.3% vs 95.5%, P=0.185). Under antibiotic exposure, the positivity rate detected by mNGS was notably higher than that by traditional culture methods, indicating that mNGS is less affected by exposure to antibiotics (P<0.05). Within 30 days, the all-cause mortality rate for patients in the IG versus the non-IG was 14.55% and 37.29%, respectively (P<0.05). Following a COX regression analysis to adjust for confounding factors, the analysis revealed that a CURB-65 score ≥3 points (HR=3.348, P=0.001) and existing cardiovascular disease (HR=2.473, P=0.026) were independent risk factors for these patients. Conversely, mNGS testing (HR=0.368, P=0.017) proved to be an independent protective factor. Conclusion: mNGS technology makes it easier to pinpoint the cause of pulmonary diffuse infectious exudative lesions without much interference from antibiotics, helping doctors spot and diagnose these issues early on, thereby playing a key role in helping them decide the best treatment approach for patients. Such conclusions may have a bias, as the performance of traditional methods might be underestimated due to the absence of complete results from other conventional diagnostic techniques like serological testing and PCR.


Subject(s)
High-Throughput Nucleotide Sequencing , Metagenome , Humans , Retrospective Studies , Male , Female , High-Throughput Nucleotide Sequencing/methods , Middle Aged , Aged , Sensitivity and Specificity , Adult , Lung Diseases/microbiology , Lung Diseases/diagnosis , Lung/microbiology , Lung/pathology , Aged, 80 and over , Metagenomics/methods
19.
Nat Commun ; 15(1): 4326, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38773113

ABSTRACT

Resolving inflammation is thought to return the affected tissue back to homoeostasis but recent evidence supports a non-linear model of resolution involving a phase of prolonged immune activity. Here we show that within days following resolution of Streptococcus pneumoniae-triggered lung inflammation, there is an influx of antigen specific lymphocytes with a memory and tissue-resident phenotype as well as macrophages bearing alveolar or interstitial phenotype. The transcriptome of these macrophages shows enrichment of genes associated with prostaglandin biosynthesis and genes that drive T cell chemotaxis and differentiation. Therapeutic depletion of post-resolution macrophages, inhibition of prostaglandin E2 (PGE2) synthesis or treatment with an EP4 antagonist, MF498, reduce numbers of lung CD4+/CD44+/CD62L+ and CD4+/CD44+/CD62L-/CD27+ T cells as well as their expression of the α-integrin, CD103. The T cells fail to reappear and reactivate upon secondary challenge for up to six weeks following primary infection. Concomitantly, EP4 antagonism through MF498 causes accumulation of lung macrophages and marked tissue fibrosis. Our study thus shows that PGE2 signalling, predominantly via EP4, plays an important role during the second wave of immune activity following resolution of inflammation. This secondary immune activation drives local tissue-resident T cell development while limiting tissue injury.


Subject(s)
Dinoprostone , Disease Models, Animal , Lung , Macrophages , Mice, Inbred C57BL , Pneumonia, Pneumococcal , Receptors, Prostaglandin E, EP4 Subtype , Streptococcus pneumoniae , Animals , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/microbiology , Pneumonia, Pneumococcal/metabolism , Mice , Dinoprostone/metabolism , Streptococcus pneumoniae/immunology , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Macrophages/immunology , Macrophages/metabolism , Lung/immunology , Lung/pathology , Lung/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Integrin alpha Chains/metabolism , Integrin alpha Chains/genetics , Female , Antigens, CD/metabolism , Antigens, CD/genetics , T-Lymphocytes/immunology
20.
Clin Transl Sci ; 17(6): e13850, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38807464

ABSTRACT

Cold-inducible RNA-binding protein (CIRP) is a damage-associated molecular pattern that plays a critical role in triggering inflammatory responses. It remains unknown whether CIRP is strongly associated with bacterial load, inflammatory response, and mortality in sepsis model. Pneumonia was induced in specific pathogen-free 8-9-week old male rats by injecting bacteria via puncture of the tracheal cartilage. The expressions of CIRP and proinflammatory cytokines [tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and IL-1ß] in lung tissues, alveolar macrophages (AMs), plasma, and bronchoalveolar lavage fluid (BALF) were determined by reverse transcription-polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay. The numbers of bacteria recovered from the lungs were correlated with the bacterial loads injected and mortality. The expressions of CIRP increased sharply as the bacterial loads increased in the lung tissues and AMs. The amounts of TNF-α, IL-6 and IL-1ß proteins synthesized were dependent on the bacterial load in the lung tissues. Releases of CIRP, TNF-α, IL-6, and IL-1ß increased with the bacterial load in the blood plasma. The proteins confirmed similar patterns in the BALF. CIRP was strongly associated with the releases of TNF-α, IL-6, and IL-1ß in the lung tissues, blood plasma, and BALF, and showed a close correlation with mortality. CIRP demonstrated a strong association with bacterial load, which is new evidence, and close correlations with proinflammatory cytokines and mortality of pneumonia in rats, suggesting that it might be an interesting pneumonic biomarker for monitoring host response and predicting mortality, and a promising target for immunotherapy.


Subject(s)
Bacterial Load , Cytokines , RNA-Binding Proteins , Animals , Male , RNA-Binding Proteins/metabolism , Cytokines/metabolism , Cytokines/blood , Rats , Lung/microbiology , Lung/immunology , Lung/pathology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Pneumonia/microbiology , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/mortality , Rats, Sprague-Dawley , Interleukin-1beta/metabolism , Interleukin-1beta/blood , Disease Models, Animal , Inflammation Mediators/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/blood , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality
SELECTION OF CITATIONS
SEARCH DETAIL
...