Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 694
Filter
1.
Methods Mol Biol ; 2813: 137-144, 2024.
Article in English | MEDLINE | ID: mdl-38888776

ABSTRACT

Air-liquid interface (ALI) airway culture models serve as a powerful tool to emulate the characteristic features of the respiratory tract in vitro. These models are particularly valuable for studying emerging respiratory viral and bacterial infections. Here, we describe an optimized protocol to obtain the ALI airway culture models using normal human bronchial epithelial cells (NHBECs). The protocol outlined below enables the generation of differentiated mucociliary airway epithelial cultures by day 28 following exposure to air.


Subject(s)
Cell Culture Techniques , Epithelial Cells , Humans , Cell Culture Techniques/methods , Epithelial Cells/microbiology , Epithelial Cells/virology , Epithelial Cells/cytology , Bronchi/cytology , Respiratory Mucosa/cytology , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Air , Cells, Cultured , Communicable Diseases/microbiology
2.
Front Cell Infect Microbiol ; 14: 1346087, 2024.
Article in English | MEDLINE | ID: mdl-38736751

ABSTRACT

Epithelial cells form a resilient barrier and orchestrate defensive and reparative mechanisms to maintain tissue stability. This review focuses on gut and airway epithelia, which are positioned where the body interfaces with the outside world. We review the many signaling pathways and mechanisms by which epithelial cells at the interface respond to invading pathogens to mount an innate immune response and initiate adaptive immunity and communicate with other cells, including resident microbiota, to heal damaged tissue and maintain homeostasis. We compare and contrast how airway and gut epithelial cells detect pathogens, release antimicrobial effectors, collaborate with macrophages, Tregs and epithelial stem cells to mount an immune response and orchestrate tissue repair. We also describe advanced research models for studying epithelial communication and behaviors during inflammation, tissue injury and disease.


Subject(s)
Homeostasis , Immunity, Innate , Intestinal Mucosa , Humans , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Respiratory Mucosa/microbiology , Respiratory Mucosa/immunology , Epithelial Cells/microbiology , Signal Transduction , Adaptive Immunity , Macrophages/immunology , Macrophages/microbiology , Host-Pathogen Interactions
3.
PLoS Pathog ; 20(5): e1011453, 2024 May.
Article in English | MEDLINE | ID: mdl-38820569

ABSTRACT

Mucosa-associated biofilms are associated with many human disease states, but the host mechanisms promoting biofilm remain unclear. In chronic respiratory diseases like cystic fibrosis (CF), Pseudomonas aeruginosa establishes chronic infection through biofilm formation. P. aeruginosa can be attracted to interspecies biofilms through potassium currents emanating from the biofilms. We hypothesized that P. aeruginosa could, similarly, sense and respond to the potassium efflux from human airway epithelial cells (AECs) to promote biofilm. Using respiratory epithelial co-culture biofilm imaging assays of P. aeruginosa grown in association with CF bronchial epithelial cells (CFBE41o-), we found that P. aeruginosa biofilm was increased by potassium efflux from AECs, as examined by potentiating large conductance potassium channel, BKCa (NS19504) potassium efflux. This phenotype is driven by increased bacterial attachment and increased coalescence of bacteria into aggregates. Conversely, biofilm formation was reduced when AECs were treated with a BKCa blocker (paxilline). Using an agar-based macroscopic chemotaxis assay, we determined that P. aeruginosa chemotaxes toward potassium and screened transposon mutants to discover that disruption of the high-sensitivity potassium transporter, KdpFABC, and the two-component potassium sensing system, KdpDE, reduces P. aeruginosa potassium chemotaxis. In respiratory epithelial co-culture biofilm imaging assays, a KdpFABCDE deficient P. aeruginosa strain demonstrated reduced biofilm growth in association with AECs while maintaining biofilm formation on abiotic surfaces. Furthermore, we determined that the Kdp operon is expressed in vivo in people with CF and the genes are conserved in CF isolates. Collectively, these data suggest that P. aeruginosa biofilm formation can be increased by attracting bacteria to the mucosal surface and enhancing coalescence into microcolonies through aberrant AEC potassium efflux sensed by the KdpFABCDE system. These findings suggest host electrochemical signaling can enhance biofilm, a novel host-pathogen interaction, and potassium flux could be a therapeutic target to prevent chronic infections in diseases with mucosa-associated biofilms, like CF.


Subject(s)
Biofilms , Cystic Fibrosis , Epithelial Cells , Operon , Potassium , Pseudomonas Infections , Pseudomonas aeruginosa , Biofilms/growth & development , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/physiology , Humans , Cystic Fibrosis/microbiology , Cystic Fibrosis/metabolism , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Potassium/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology
4.
PLoS Pathog ; 20(5): e1012111, 2024 May.
Article in English | MEDLINE | ID: mdl-38718049

ABSTRACT

Infants are highly susceptible to invasive respiratory and gastrointestinal infections. To elucidate the age-dependent mechanism(s) that drive bacterial spread from the mucosa, we developed an infant mouse model using the prevalent pediatric respiratory pathogen, Streptococcus pneumoniae (Spn). Despite similar upper respiratory tract (URT) colonization levels, the survival rate of Spn-infected infant mice was significantly decreased compared to adults and corresponded with Spn dissemination to the bloodstream. An increased rate of pneumococcal bacteremia in early life beyond the newborn period was attributed to increased bacterial translocation across the URT barrier. Bacterial dissemination in infant mice was independent of URT monocyte or neutrophil infiltration, phagocyte-derived ROS or RNS, inflammation mediated by toll-like receptor 2 or interleukin 1 receptor signaling, or the pore-forming toxin pneumolysin. Using molecular barcoding of Spn, we found that only a minority of bacterial clones in the nasopharynx disseminated to the blood in infant mice, indicating the absence of robust URT barrier breakdown. Rather, transcriptional profiling of the URT epithelium revealed a failure of infant mice to upregulate genes involved in the tight junction pathway. Expression of many such genes was also decreased in early life in humans. Infant mice also showed increased URT barrier permeability and delayed mucociliary clearance during the first two weeks of life, which corresponded with tighter attachment of bacteria to the respiratory epithelium. Together, these results demonstrate a window of vulnerability during postnatal development when altered mucosal barrier function facilitates bacterial dissemination.


Subject(s)
Pneumococcal Infections , Streptococcus pneumoniae , Animals , Pneumococcal Infections/microbiology , Pneumococcal Infections/immunology , Mice , Humans , Animals, Newborn , Disease Models, Animal , Mice, Inbred C57BL , Respiratory Mucosa/microbiology , Respiratory Mucosa/metabolism , Female , Nasopharynx/microbiology
5.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38693120

ABSTRACT

Respiratory viral infection increases host susceptibility to secondary bacterial infections, yet the precise dynamics within airway epithelia remain elusive. Here, we elucidate the pivotal role of CD47 in the airway epithelium during bacterial super-infection. We demonstrated that upon influenza virus infection, CD47 expression was upregulated and localized on the apical surface of ciliated cells within primary human nasal or bronchial epithelial cells. This induced CD47 exposure provided attachment sites for Staphylococcus aureus, thereby compromising the epithelial barrier integrity. Through bacterial adhesion assays and in vitro pull-down assays, we identified fibronectin-binding proteins (FnBP) of S. aureus as a key component that binds to CD47. Furthermore, we found that ciliated cell-specific CD47 deficiency or neutralizing antibody-mediated CD47 inactivation enhanced in vivo survival rates. These findings suggest that interfering with the interaction between airway epithelial CD47 and pathogenic bacterial FnBP holds promise for alleviating the adverse effects of super-infection.


Subject(s)
CD47 Antigen , Epithelial Cells , Staphylococcal Infections , Staphylococcus aureus , Superinfection , CD47 Antigen/metabolism , CD47 Antigen/genetics , Humans , Animals , Superinfection/microbiology , Mice , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Epithelial Cells/virology , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Bacterial Adhesion , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Mice, Inbred C57BL , Bronchi/metabolism , Bronchi/cytology , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/virology , Mice, Knockout , Influenza A Virus, H1N1 Subtype
6.
Vet Res Commun ; 48(3): 1481-1495, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38336962

ABSTRACT

This study proposes an ecological approach for preventing respiratory tract infections caused by Bordetella bronchiseptica in mammals using a mixture of carbohydrates. In an in vivo study, 51-day-old New Zealand rabbits were treated with a solution containing 1 × 107 CFUs of B. bronchiseptica and 250 µg of one of the following carbohydrates: N acetylglucosamine (GlcNAc), N acetylgalactosamine (GalNAc), alpha methyl mannose (AmeMan), alpha methyl glucose (AmeGlc) and sialic acid (Neu5AC). Positive (B. bronchiseptica) and negative (Physiological Saline Solution (PSS)) controls were included. Animals treated with GlcNAc or AmeGlc showed no clinical signs of infection and exhibited a significant reduction (p < 0.05) in the severity of microscopic lesions evaluated in the nasal cavity and lung compared with the positive controls. Additionally, the presence of bacteria was not detected through microbiological isolation or PCR in the lungs of animals treated with these sugars. Use of a mixture of GlcNAc and AmeGlc resulted in greater inhibition of microscopic lesions, with a significant reduction (p < 0.05) in the severity of these lesions compared to the results obtained using individual sugars. Furthermore, the bacterium was not detected through microbiological isolation, Polymerase Chain Reaction (PCR) or indirect immunoperoxidase (IIP) in this group.


Subject(s)
Bordetella Infections , Bordetella bronchiseptica , Respiratory Mucosa , Animals , Rabbits , Bordetella bronchiseptica/drug effects , Bordetella Infections/veterinary , Bordetella Infections/microbiology , Respiratory Mucosa/drug effects , Respiratory Mucosa/microbiology , Bacterial Adhesion/drug effects , Carbohydrates/pharmacology , Acetylglucosamine/pharmacology , Respiratory Tract Infections/veterinary , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/drug therapy , Lung/microbiology , Lung/drug effects , Lung/pathology
7.
Am J Respir Crit Care Med ; 209(12): 1453-1462, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38324627

ABSTRACT

Rationale: Pseudomonas aeruginosa is the major bacterial pathogen colonizing the airways of adult patients with cystic fibrosis (CF) and causes chronic infections that persist despite antibiotic therapy. Intracellular bacteria may represent an unrecognized reservoir of bacteria that evade the immune system and antibiotic therapy. Although the ability of P. aeruginosa to invade and survive within epithelial cells has been described in vitro in different epithelial cell models, evidence of this intracellular lifestyle in human lung tissues is currently lacking. Objectives: To detect and characterize intracellular P. aeruginosa in CF airway epithelium from human lung explant tissues. Methods: We sampled lung explant tissues from patients with CF undergoing lung transplantation and non-CF lung donor control tissue. We analyzed lung tissue sections for the presence of intracellular P. aeruginosa using quantitative culture and microscopy, in parallel to histopathology and airway morphometry. Measurements and Main Results: P. aeruginosa was isolated from the lungs of seven patients with CF undergoing lung transplantation. Microscopic assessment revealed the presence of intracellular P. aeruginosa within airway epithelial cells in three of the seven patients analyzed at a varying but low frequency. We observed those events occurring in lung regions with high bacterial burden. Conclusions: This is the first study describing the presence of intracellular P. aeruginosa in CF lung tissues. Although intracellular P. aeruginosa in airway epithelial cells is likely relatively rare, our findings highlight the plausible occurrence of this intracellular bacterial reservoir in chronic CF infections.


Subject(s)
Cystic Fibrosis , Lung Transplantation , Lung , Pseudomonas Infections , Pseudomonas aeruginosa , Respiratory Mucosa , Humans , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Female , Male , Adult , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Pseudomonas Infections/microbiology , Lung/microbiology , Lung/pathology , Young Adult , Epithelial Cells/microbiology
8.
Indian J Tuberc ; 69 Suppl 2: S241-S245, 2022.
Article in English | MEDLINE | ID: mdl-36400517

ABSTRACT

The article reviews the immune changes in the elderly with particular reference to susceptibility of elderly to Tubercular infection whether new or LTBI and in the light of recent advances in the field of immune mechanisms of tubercular infection. An primary understanding of the host response to infections and M. tuberculosis (M.tb) infection in particular helps to better understand the various issues of immune response to tubercular infection in the elderly. Immune mechanisms of ageing in particular deal with the twin unique mechanisms and terms particular to aging- Immunosenescence and Inflammaging. In the elderly patient both the Innate and the Adaptive immune responses are affected at various levels. The M.tb bacteria encounters the innate immune system initially and thereafter the response is from the cells of the adaptive immune system. The M.tb bacillus which enters through the respiratory system to the bronchioles and alveolus encounters the immune system at three levels which are the Resident structural i.e. alveolar epithelium, Resident innate i.e. the alveolar and pulmonary macrophages and the Infiltrating innate i.e. the neutrophils and monocytes. Increased inflammatory changes present in the lung mucosa has been associated with changes in multiple innate molecular defence mechanisms that could influence the ability of M.tb to establish an infection, the various cellular mechanisms involved and the evasive strategies evolved by the M.tb to survive and disseminate are briefly described. The susceptibility of the elderly to develop and succumb to TB may be a direct impact of increased inflammation at every stage of infection. M.tb is a potent stimulator of multiple inflammatory responses and added to a basal inflammatory state with evasiveness of M.tb bacilli, enable it to overcome and disseminate, increasing the morbidity and mortality in the infected elderly. Hopefully a better understanding of the immune mechanisms involved will enable better preventive, diagnostic and treatment modalities.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Aged , Immunity, Innate , Tuberculosis/microbiology , Respiratory Mucosa/microbiology , Lung
9.
PLoS Pathog ; 18(1): e1010209, 2022 01.
Article in English | MEDLINE | ID: mdl-35085362

ABSTRACT

Haemophilus influenzae (Hi) infections are associated with recurring acute exacerbations of chronic respiratory diseases in children and adults including otitis media, pneumonia, chronic obstructive pulmonary disease and asthma. Here, we show that persistence and recurrence of Hi infections are closely linked to Hi metabolic properties, where preferred growth substrates are aligned to the metabolome of human airway epithelial surfaces and include lactate, pentoses, and nucleosides, but not glucose that is typically used for studies of Hi growth in vitro. Enzymatic and physiological investigations revealed that utilization of lactate, the preferred Hi carbon source, required the LldD L-lactate dehydrogenase (conservation: 98.8% of strains), but not the two redox-balancing D-lactate dehydrogenases Dld and LdhA. Utilization of preferred substrates was directly linked to Hi infection and persistence. When unable to utilize L-lactate or forced to rely on salvaged guanine, Hi showed reduced extra- and intra-cellular persistence in a murine model of lung infection and in primary normal human nasal epithelia, with up to 3000-fold attenuation observed in competitive infections. In contrast, D-lactate dehydrogenase mutants only showed a very slight reduction compared to the wild-type strain. Interestingly, acetate, the major Hi metabolic end-product, had anti-inflammatory effects on cultured human tissue cells in the presence of live but not heat-killed Hi, suggesting that metabolic endproducts also influence HI-host interactions. Our work provides significant new insights into the critical role of metabolism for Hi persistence in contact with host cells and reveals for the first time the immunomodulatory potential of Hi metabolites.


Subject(s)
Haemophilus Infections/metabolism , Haemophilus influenzae/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Animals , Host-Pathogen Interactions/physiology , Humans , Mice
10.
Infect Immun ; 90(1): e0042321, 2022 01 25.
Article in English | MEDLINE | ID: mdl-34662211

ABSTRACT

To understand protective immune responses against the onset of group A Streptococcus respiratory infection, we investigated whether MyD88 KO mice were susceptible to acute infection through transmission. After commingling with mice that had intranasal group A Streptococcus (GAS) inoculation, MyD88-/- recipient mice had increased GAS loads in the nasal cavity and throat that reached a mean throat colonization of 6.3 × 106 CFU/swab and mean GAS load of 5.2 × 108 CFU in the nasal cavity on day 7. Beyond day 7, MyD88-/- recipient mice became moribund, with mean 1.6 × 107 CFU/swab and 2.5 × 109 CFU GAS in the throat and nasal cavity, respectively. Systemic GAS infection occurred a couple of days after the upper respiratory infection. GAS infects the lip, the gingival sulcus of the incisor teeth, and the lamina propria of the turbinate but not the nasal cavity and nasopharyngeal tract epithelia, and C57BL/6J recipient mice had no or low levels of GAS in the nasal cavity and throat. Direct nasal GAS inoculation of MyD88-/- mice caused GAS infection, mainly in the lamina propria of the turbinate. In contrast, C57BL/6J mice with GAS inoculation had GAS bacteria in the nasal cavity but not in the lamina propria of the turbinates. Thus, MyD88-/- mice are highly susceptible to acute and lethal GAS infection through transmission, and MyD88 signaling is critical for protection of the respiratory tract lamina propria but not nasal and nasopharyngeal epithelia against GAS infection.


Subject(s)
Epithelium/microbiology , Host-Pathogen Interactions , Myeloid Differentiation Factor 88/deficiency , Respiratory Mucosa/microbiology , Respiratory Tract Infections/etiology , Streptococcal Infections/etiology , Streptococcal Infections/transmission , Streptococcus pyogenes/physiology , Animals , Biopsy , Disease Susceptibility , Epithelium/pathology , Genetic Predisposition to Disease , Immunohistochemistry , Mice , Mice, Knockout , Neutrophil Infiltration , Organ Specificity , Respiratory Mucosa/pathology , Respiratory Tract Infections/pathology , Streptococcal Infections/pathology
11.
Sci Rep ; 11(1): 19534, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34599245

ABSTRACT

Next-generation sequencing (NGS) technologies have been applied in bacterial flora analysis. However, there is no standardized protocol, and the optimal clustering threshold for estimating bacterial species in respiratory infection specimens is unknown. This study was conducted to investigate the optimal threshold for clustering 16S ribosomal RNA gene sequences into operational taxonomic units (OTUs) by comparing the results of NGS technology with those of the Sanger method, which has a higher accuracy of sequence per single read than NGS technology. This study included 45 patients with pneumonia with aspiration risks and 35 patients with lung abscess. Compared to Sanger method, the concordance rates of NGS technology (clustered at 100%, 99%, and 97% homology) with the predominant phylotype were 78.8%, 71.3%, and 65.0%, respectively. With respect to the specimens dominated by the Streptococcus mitis group, containing several important causative agents of pneumonia, Bray Curtis dissimilarity revealed that the OTUs obtained at 100% clustering threshold (versus those obtained at 99% and 97% thresholds; medians of 0.35, 0.69, and 0.71, respectively) were more similar to those obtained by the Sanger method, with statistical significance (p < 0.05). Clustering with 100% sequence identity is necessary when analyzing the microbiota of respiratory infections using NGS technology.


Subject(s)
Microbiota , Respiratory Mucosa/microbiology , Respiratory Tract Infections/etiology , Aged , Aged, 80 and over , Disease Susceptibility , Female , Gene Library , High-Throughput Nucleotide Sequencing , Humans , Male , Metagenome , Metagenomics/methods , Middle Aged , Respiratory Tract Infections/diagnosis , Retrospective Studies
12.
EBioMedicine ; 71: 103538, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34425308

ABSTRACT

BACKGROUND: Normal airway microbial communities play a central role in respiratory health but are poorly characterized. Cigarette smoking is the dominant global environmental influence on lung function, and asthma has become the most prevalent chronic respiratory disease worldwide. Both conditions have major microbial components that are incompletely defined. METHODS: We investigated airway bacterial communities in a general population sample of 529 Australian adults. Posterior oropharyngeal swabs were analyzed by sequencing of the 16S rRNA gene. The microbiota were characterized according to their prevalence, abundance and network memberships. FINDINGS: The microbiota were similar across the general population, and were strongly organized into co-abundance networks. Smoking was associated with diversity loss, negative effects on abundant taxa, profound alterations to network structure and expansion of Streptococcus spp. By contrast, the asthmatic microbiota were selectively affected by an increase in Neisseria spp. and by reduced numbers of low abundance but prevalent organisms. INTERPRETATION: Our study shows that the healthy airway microbiota in this population were contained within a highly structured ecosystem, suggesting balanced relationships between the microbiome and human host factors. The marked abnormalities in smokers may contribute to chronic obstructive pulmonary disease (COPD) and lung cancer. The narrow spectrum of abnormalities in asthmatics encourages investigation of damaging and protective effects of specific bacteria. FUNDING: The study was funded by the Asmarley Trust and a Wellcome Joint Senior Investigator Award to WOCC and MFM (WT096964MA and WT097117MA). The Busselton Healthy Ageing Study is supported by the Government of Western Australia (Office of Science, Department of Health) the City of Busselton, and private donations.


Subject(s)
Asthma/epidemiology , Microbiota , Respiratory Mucosa/microbiology , Smoking/epidemiology , Adult , Aged , Asthma/etiology , Australia/epidemiology , Computational Biology/methods , Disease Susceptibility , Female , Humans , Male , Metagenomics/methods , Middle Aged , Population Surveillance , RNA, Ribosomal, 16S , Smoking/adverse effects , Tobacco Smoking
13.
Sci Rep ; 11(1): 17148, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34433845

ABSTRACT

The low biomass of respiratory samples makes it difficult to accurately characterise the microbial community composition. PCR conditions and contaminating microbial DNA can alter the biological profile. The objective of this study was to benchmark the currently available laboratory protocols to accurately analyse the microbial community of low biomass samples. To study the effect of PCR conditions on the microbial community profile, we amplified the 16S rRNA gene of respiratory samples using various bacterial loads and different number of PCR cycles. Libraries were purified by gel electrophoresis or AMPure XP and sequenced by V2 or V3 MiSeq reagent kits by Illumina sequencing. The positive control was diluted in different solvents. PCR conditions had no significant influence on the microbial community profile of low biomass samples. Purification methods and MiSeq reagent kits provided nearly similar microbiota profiles (paired Bray-Curtis dissimilarity median: 0.03 and 0.05, respectively). While profiles of positive controls were significantly influenced by the type of dilution solvent, the theoretical profile of the Zymo mock was most accurately analysed when the Zymo mock was diluted in elution buffer (difference compared to the theoretical Zymo mock: 21.6% for elution buffer, 29.2% for Milli-Q, and 79.6% for DNA/RNA shield). Microbiota profiles of DNA blanks formed a distinct cluster compared to low biomass samples, demonstrating that low biomass samples can accurately be distinguished from DNA blanks. In summary, to accurately characterise the microbial community composition we recommend 1. amplification of the obtained microbial DNA with 30 PCR cycles, 2. purifying amplicon pools by two consecutive AMPure XP steps and 3. sequence the pooled amplicons by V3 MiSeq reagent kit. The benchmarked standardized laboratory workflow presented here ensures comparability of results within and between low biomass microbiome studies.


Subject(s)
Benchmarking/methods , Microbiota , Reagent Kits, Diagnostic/standards , Respiratory Mucosa/microbiology , Biomass , Humans , Metagenomics/methods , Metagenomics/standards , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , RNA, Ribosomal, 16S/genetics , Saliva/microbiology
14.
Cell Rep ; 36(3): 109392, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289364

ABSTRACT

Chitin, a major component of fungal cell walls, has been associated with allergic disorders such as asthma. However, it is unclear how mammals recognize chitin and the principal receptor(s) on epithelial cells that sense chitin remain to be determined. In this study, we show that LYSMD3 is expressed on the surface of human airway epithelial cells and demonstrate that LYSMD3 is able to bind chitin, as well as ß-glucan, on the cell walls of fungi. Knockdown or knockout of LYSMD3 also sharply blunts the production of inflammatory cytokines by epithelial cells in response to chitin and fungal spores. Competitive inhibition of the LYSMD3 ectodomain by soluble LYSMD3 protein, multiple ligands, or antibody against LYSMD3 also blocks chitin signaling. Our study reveals LYSMD3 as a mammalian pattern recognition receptor (PRR) for chitin and establishes its role in epithelial cell inflammatory responses to chitin and fungi.


Subject(s)
Chitin , Mammals , Membrane Proteins , Receptors, Pattern Recognition , Animals , Humans , Mice , beta-Glucans/metabolism , Candida albicans/physiology , Cell Membrane/metabolism , Chitin/metabolism , Epithelial Cells/metabolism , HeLa Cells , Immunity, Innate , Inflammation/pathology , Mammals/metabolism , Membrane Proteins/metabolism , RAW 264.7 Cells , Receptors, Pattern Recognition/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Signal Transduction
15.
Am J Respir Crit Care Med ; 204(6): 692-702, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34170795

ABSTRACT

Rationale: Although it is clear that cystic fibrosis (CF) airway disease begins at a very young age, the early and subsequent steps in disease pathogenesis and the relative contribution of infection, mucus, and inflammation are not well understood. Objectives: As one approach to assessing the early contribution of infection, we tested the hypothesis that early and continuous antibiotics would decrease the airway bacterial burden. We believed that, if they do, this might reveal aspects of the disease that are more or less sensitive to decreasing infection. Methods: Three groups of pigs were studied from birth until ∼3 weeks of age: 1) wild-type, 2) CF, and 3) CF pigs treated continuously with broad-spectrum antibiotics from birth until study completion. Disease was assessed with chest computed tomography, histopathology, microbiology, and BAL. Measurements and Main Results: Disease was present by 3 weeks of age in CF pigs. Continuous antibiotics from birth improved chest computed tomography imaging abnormalities and airway mucus accumulation but not airway inflammation in the CF pig model. However, reducing bacterial infection did not improve two disease features already present at birth in CF pigs: air trapping and submucosal gland duct plugging. In the CF sinuses, antibiotics did not prevent the development of infection or disease or the number of bacteria but did alter the bacterial species. Conclusions: These findings suggest that CF airway disease begins immediately after birth and that early and continuous antibiotics impact some, but not all, aspects of CF lung disease development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/microbiology , Lung/drug effects , Respiratory Mucosa/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , Bacteria/isolation & purification , Bronchoalveolar Lavage Fluid/microbiology , Cystic Fibrosis/diagnostic imaging , Cystic Fibrosis/pathology , Lung/diagnostic imaging , Lung/microbiology , Lung/pathology , Multidetector Computed Tomography , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , Swine
16.
J Cyst Fibros ; 20(4): 682-691, 2021 07.
Article in English | MEDLINE | ID: mdl-34112603

ABSTRACT

BACKGROUND: In Cystic Fibrosis (CF) airways, the dehydrated, thick mucus promotes the establishment of persistent polymicrobial infections and drives chronic airways inflammation. This also predisposes the airways to further infections, the vicious, self-perpetuating cycle causing lung damage and progressive lung function decline. The airways are a poly-microbial environment, containing both aerobic and anaerobic bacterial species. Pseudomonas aeruginosa (P. aeruginosa) infections contribute to the excessive inflammatory response in CF, but the role of anaerobic Prevotella spp., frequently found in CF airways, is not known. MATERIALS: We assessed innate immune signalling in CF airway epithelial cells in response to clinical strains of P. histicola, P. nigresens and P. aeruginosa. CFBE41o- cells were infected with P. aeruginosa (MOI 100, 2h) followed by infection with P. histicola or P. nigrescens (MOI 100, 2h). Cells were incubated under anaerobic conditions for the duration of the experiments. RESULTS: Our study shows that P. histicola and P. nigresens can reduce the growth of P. aeruginosa and dampen the inflammatory response in airway epithelial cells. We specifically illustrate that the presence of the investigated Prevotella spp. reduces Toll-like-receptor (TLR)-4, MAPK, NF-κB(p65) signalling and cytokine release (Interleukin (IL)-6, IL-8) in mixed infections. CONCLUSION: Our work, for the first time, strongly indicates a relationship between P. aeruginosa and anaerobic Prevotella spp.. The observed modified NF-κB and MAPK signalling indicates some mechanisms underlying this interaction that could offer a novel therapeutic approach to combat chronic P. aeruginosa infection in people with CF.


Subject(s)
Bronchi/cytology , Bronchi/microbiology , Cystic Fibrosis/complications , Cystic Fibrosis/microbiology , Epithelial Cells/immunology , Inflammation/etiology , Inflammation/microbiology , Prevotella/physiology , Pseudomonas Infections/etiology , Pseudomonas aeruginosa/physiology , Respiratory Mucosa/cytology , Respiratory Mucosa/microbiology , Cells, Cultured , Cystic Fibrosis/immunology , Humans
17.
Front Immunol ; 12: 679829, 2021.
Article in English | MEDLINE | ID: mdl-34108973

ABSTRACT

Commensal microbiota has emerged as an essential biomarker and regulator of both tumorigenesis and response to cancer therapy. However, our current knowledge about microbiota in cancer has been largely limited to intestinal microbiota. As a mucosal organ harboring one of the largest surface areas in the body, the lung is exposed to a variety of microbes through inhalation and micro-aspiration, and is colonized by a diverse bacterial community in both physiological and pathological conditions. Importantly, increasing evidence has linked the lung microbiome to cancer development. Studies in lung cancer patients and mouse models have revealed tumor-associated dysregulation of the local microbiome in the lung, which in turn impacts cancer progression by shaping the tumor microenvironment and modulating the activity of tumor-infiltrating immune cells. These findings not only provide novel mechanistic insight into the biology of lung cancer but also shed light on new therapeutic targets and strategies for lung cancer prevention and treatment. The goal of this review is to discuss the key findings, remaining questions, and future directions in this new and exciting field.


Subject(s)
Disease Susceptibility , Host Microbial Interactions , Lung Neoplasms/etiology , Microbiota , Cell Transformation, Neoplastic , Disease Management , Dysbiosis , Gastrointestinal Microbiome , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung Neoplasms/therapy , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/microbiology , Tumor Microenvironment
18.
Am J Respir Cell Mol Biol ; 65(6): 593-602, 2021 12.
Article in English | MEDLINE | ID: mdl-34192508

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is associated with colonization by bacterial pathogens and repeated airway infections, leading to exacerbations and impaired lung function. The highly glycosylated mucins in the mucus lining the airways are an important part of the host defense against pathogens. However, mucus accumulation can contribute to COPD pathology. Here, we examined whether inflammation is associated with glycosylation changes that affect interactions between airway mucins and pathogens. We isolated mucins from lower airway samples (n = 4-9) from long-term smokers with and without COPD and from never-smokers. The most abundant terminal glycan moiety was N-acetylneuraminic acid (Neu5Ac) among smokers with and without COPD and N-acetyl-hexoseamine among never-smokers. Moraxella catarrhalis bound to MUC5 mucins from smokers with and without COPD. M. catarrhalis binding correlated with inflammatory parameters and Neu5Ac content. M. catarrhalis binding was abolished by enzymatic removal of Neu5Ac. Furthermore, M. catarrhalis bound to α2,6 sialyl-lactose, suggesting that α2,6 sialic acid contributes to M. catarrhalis binding to mucins. Furthermore, we detected more M. catarrhalis binding to mucins from patients with pneumonia than to those from control subjects (n = 8-13), and this binding correlated with C-reactive protein and Neu5Ac levels. These results suggest a key role of inflammation-induced Neu5Ac in the adhesion of M. catarrhalis to airway mucins. The inflammation-induced ability of MUC5 mucins to bind M. catarrhalis is likely a host defense mechanism in the healthy lung, although it cannot be excluded that impaired mucociliary clearance limits the effectiveness of this defense in patients with COPD.


Subject(s)
Lung/metabolism , Moraxella catarrhalis/metabolism , Mucin-5B/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory Mucosa/metabolism , Humans , Inflammation , Lung/microbiology , Pulmonary Disease, Chronic Obstructive/microbiology , Respiratory Mucosa/microbiology , Sialic Acids/metabolism
19.
J Immunol Res ; 2021: 6660379, 2021.
Article in English | MEDLINE | ID: mdl-33855090

ABSTRACT

Tissue-resident memory T cells (TRM) are different from effector memory T cells (TEM) and central memory T cells (TCM) and contribute to the protective immunity against local challenges. Currently, we found that CD4+ and CD8+ TRM cells in the nasal mucosa, trachea, lungs, and lavage fluids were heterogeneous on the expression of CD69 and CD103 as well as the production of cytokines including IFN-γ, IL-2, and TNF-α. After intranasal vaccination of mice with BCG, respiratory tissues expressed higher levels of the chemokine CXCL16 and TRM cells expressed CXCR6 to CXCL16. In addition, antigen-specific CD4+ and CD8+ TRM cells expressed cytokines following the stimulation with BCG and persisted in the nasal mucosa, trachea, and lungs for more than a hundred days. At the same time, mice were infected intranasally with live BCG and the results showed that vaccinated mice cleared up live BCG faster than nonvaccinated mice in the respiratory system. Taken together, our data demonstrated that intranasal vaccination of mice with BCG could induce antigen-specific CD4+ and CD8+ TRM cells in the respiratory system and have the ability to provide protection against pulmonary reinfection.


Subject(s)
BCG Vaccine/administration & dosage , Reinfection/prevention & control , T-Lymphocyte Subsets/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Administration, Intranasal , Animals , BCG Vaccine/immunology , Disease Models, Animal , Female , Humans , Immunogenicity, Vaccine , Immunologic Memory , Mice , Mycobacterium bovis/immunology , Mycobacterium bovis/isolation & purification , Reinfection/immunology , Reinfection/microbiology , Reinfection/pathology , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology , Respiratory Mucosa/pathology , T-Lymphocyte Subsets/metabolism , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
20.
Vet Microbiol ; 257: 109067, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33862331

ABSTRACT

Respiratory diseases negatively impact the global goat industry, but are understudied. There is a shortage of established and biological relevant in vitro or ex vivo assays to study caprine respiratory infections. Here, we describe the establishment of an in vitro system based on well-differentiated caprine airway epithelial cell (AEC) cultures grown under air liquid interface conditions as an experimental platform to study caprine respiratory pathogens. The functional differentiation of the AEC cultures was monitored and confirmed by light and immunofluorescence microscopy, scanning electron microscopy and examination of histological sections. We validated the functionality of the platform by studying Influenza D Virus (IDV) infection and Mycoplasma mycoides subsp. capri (Mmc) colonization over 5 days, including monitoring of infectious agents by titration and qPCR as well as colour changing units, respectively. The inoculation of caprine AEC cultures with IDV showed that efficient viral replication takes place, and revealed that IDV has a marked cell tropism for ciliated cells. Furthermore, AEC cultures were successfully infected with Mmc using a multiplicity of infection of 0.1 and colonization was monitored over several days. Altogether, these results demonstrate that our newly-established caprine AEC cultures can be used to investigate host-pathogen interactions of caprine respiratory pathogens.


Subject(s)
Cell Culture Techniques/methods , Cell Culture Techniques/veterinary , Epithelial Cells/microbiology , Epithelial Cells/virology , Respiratory Mucosa/microbiology , Respiratory Mucosa/virology , Respiratory System/cytology , Animals , Bronchi/cytology , Cell Differentiation , Cells, Cultured , Goats , Host-Pathogen Interactions , Microscopy, Electron, Scanning , Mycoplasma/physiology , Thogotovirus/physiology , Viral Tropism , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...