Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.110
Filter
1.
Clin Immunol ; 263: 110221, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636891

ABSTRACT

Staphylococcus aureus mucosal biofilms are associated with recalcitrant chronic rhinosinusitis (CRS). However, S. aureus colonisation of sinus mucosa is frequent in the absence of mucosal inflammation. This questions the relevance of S. aureus biofilms in CRS etiopathogenesis. This study aimed to investigate whether strain-level variation in in vitro-grown S. aureus biofilm properties relates to CRS disease severity, in vitro toxicity, and immune B cell responses in sinonasal tissue from CRS patients and non-CRS controls. S. aureus clinical isolates, tissue samples, and matched clinical datasets were collected from CRS patients with nasal polyps (CRSwNP), CRS without nasal polyps (CRSsNP), and controls. B cell responses in tissue samples were characterised by FACS. S. aureus biofilms were established in vitro, followed by measuring their properties of metabolic activity, biomass, colony-forming units, and exoprotein production. S. aureus virulence was evaluated using whole-genome sequencing, mass spectrometry and application of S. aureus biofilm exoproteins to air-liquid interface cultures of primary human nasal epithelial cells (HNEC-ALI). In vitro S. aureus biofilm properties were correlated with increased CRS severity scores, infiltration of antibody-secreting cells and loss of regulatory B cells in tissue samples. Biofilm exoproteins from S. aureus with high biofilm metabolic activity had enriched virulence genes and proteins, and negatively affected the barrier function of HNEC-ALI cultures. These findings support the notion of strain-level variation in S. aureus biofilms to be critical in the pathophysiology of CRS.


Subject(s)
Biofilms , Rhinitis , Sinusitis , Staphylococcal Infections , Staphylococcus aureus , Humans , Sinusitis/immunology , Sinusitis/microbiology , Staphylococcus aureus/immunology , Rhinitis/immunology , Rhinitis/microbiology , Chronic Disease , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Male , Female , Middle Aged , Nasal Polyps/immunology , Nasal Polyps/microbiology , Adult , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , B-Lymphocytes/immunology , Severity of Illness Index , Aged , Rhinosinusitis
2.
Microbiol Spectr ; 12(6): e0351623, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38687064

ABSTRACT

Recent case reports and epidemiological data suggest that fungal infections represent an underappreciated complication among people with severe COVID-19. However, the frequency of fungal colonization in patients with COVID-19 and associations with specific immune responses in the airways remain incompletely defined. We previously generated a single-cell RNA-sequencing data set characterizing the upper respiratory microenvironment during COVID-19 and mapped the relationship between disease severity and the local behavior of nasal epithelial cells and infiltrating immune cells. Our previous study, in agreement with findings from related human cohorts, demonstrated that a profound deficiency in host immunity, particularly in type I and type III interferon signaling in the upper respiratory tract, is associated with rapid progression to severe disease and worse clinical outcomes. We have now performed further analysis of this cohort and identified a subset of participants with severe COVID-19 and concurrent detection of Candida species-derived transcripts within samples collected from the nasopharynx and trachea. Here, we present the clinical characteristics of these individuals. Using matched single-cell transcriptomic profiles of these individuals' respiratory mucosa, we identify epithelial immune signatures suggestive of IL17 stimulation and anti-fungal immunity. Further, we observe a significant expression of anti-fungal inflammatory cascades in the nasal and tracheal epithelium of all participants who went on to develop severe COVID-19, even among participants without detectable genetic material from fungal pathogens. Together, our data suggest that IL17 stimulation-in part driven by Candida colonization-and blunted interferon signaling represent a common feature of severe COVID-19 infection. IMPORTANCE: In this paper, we present an analysis suggesting that symptomatic and asymptomatic fungal coinfections can impact patient disease progression during COVID-19 hospitalization. By looking into the presence of other pathogens and their effect on the host immune response during COVID-19 hospitalizations, we aim to offer insight into an underestimated scenario, furthering our current knowledge of determinants of severity that could be considered for future diagnostic and intervention strategies.


Subject(s)
COVID-19 , Coinfection , Epithelial Cells , Interferon Type I , Interleukin-17 , SARS-CoV-2 , Humans , Interleukin-17/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , COVID-19/immunology , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Interferon Type I/metabolism , Interferon Type I/immunology , Male , SARS-CoV-2/immunology , Middle Aged , Female , Epithelial Cells/immunology , Epithelial Cells/microbiology , Adult , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Aged , Nasopharynx/microbiology , Candidiasis/immunology , Candidiasis/microbiology , Mycoses/immunology
3.
Int Arch Allergy Immunol ; 185(5): 466-479, 2024.
Article in English | MEDLINE | ID: mdl-38354721

ABSTRACT

INTRODUCTION: Chronic rhinosinusitis with nasal polyps (CRSwNP) is a common chronic inflammatory disease of the nose characterized by barrier disruption and environmental susceptibility, and the deletion of ZNF365 may be a factor inducing these manifestations. However, there is no study on the mechanism of action between CRSwNP and ZNF365. Therefore, this study focuses on the effect of the zinc finger protein ZNF365 on the proliferation of nasal mucosal epithelial cells and their defense against Staphylococcus aureus (S. aureus). METHODS: Immunohistochemistry and Western blot were applied to verify the changes of ZNF365 expression in nasal polyp tissues and control tissues, as well as in primary epithelial cells. ZNF365 was knocked down in human nasal mucosa epithelial cell line (HNEpc), and the proliferation, migration, and transdifferentiation of epithelium were observed by immunofluorescence, QPCR, CCK8, and cell scratch assay. The changes of mesenchymal markers and TLR4-MAPK-NF-κB pathway were also observed after the addition of S. aureus. RESULTS: ZNF365 expression was reduced in NP tissues and primary nasal mucosal epithelial cells compared to controls. Knockdown of ZNF365 in HNEpc resulted in decreased proliferation and migration ability of epithelial cells and abnormal epithelial differentiation (decreased expression of tight junction proteins). S. aureus stimulation further inhibited epithelial cell proliferation and migration, while elevated markers of epithelial-mesenchymal transition and inflammatory responses occurred. CONCLUSION: ZNF365 is instrumental in maintaining the proliferative capacity of nasal mucosal epithelial cells and defending against the invasion of S. aureus. The findings suggest that ZNF365 may participate in the development of CRSwNP.


Subject(s)
Cell Proliferation , Nasal Mucosa , Staphylococcus aureus , Humans , Cell Line , Cell Movement/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/immunology , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nasal Mucosa/metabolism , Nasal Polyps/immunology , Nasal Polyps/microbiology , Rhinitis/immunology , Rhinitis/microbiology , Signal Transduction , Sinusitis/immunology , Sinusitis/microbiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology
4.
Cell Commun Signal ; 21(1): 306, 2023 10 30.
Article in English | MEDLINE | ID: mdl-37904180

ABSTRACT

Chronic rhinosinusitis (CRS) is a pathological condition characterized by persistent inflammation in the upper respiratory tract and paranasal sinuses. The epithelium serves as the first line of defense against potential threats and protects the nasal mucosa. The fundamental mechanical barrier is formed by the cell-cell contact and mucociliary clearance (MCC) systems. The physical-mechanical barrier is comprised of many cellular structures, including adhesion junctions and tight junctions (TJs). To this end, different factors, such as the dysfunction of MCC, destruction of epithelial barriers, and tissue remodeling, are related to the onset and development of CRS. Recently published studies reported the critical role of different microorganisms, such as Staphylococcus aureus and Pseudomonas aeruginosa, in the induction of the mentioned factors. Bacteria could result in diminished ciliary stimulation capacity, and enhance the chance of CRS by reducing basal ciliary beat frequency. Additionally, bacterial exoproteins have been demonstrated to disrupt the epithelial barrier and induce downregulation of transmembrane proteins such as occludin, claudin, and tricellulin. Moreover, bacteria exert an influence on TJ proteins, leading to an increase in the permeability of polarized epithelial cells. Noteworthy, it is evident that the activation of TLR2 by staphylococcal enterotoxin can potentially undermine the structural integrity of TJs and the epithelial barrier through the induction of pro-inflammatory cytokines. The purpose of this article is an attempt to investigate the possible role of the most important microorganisms associated with CRS and their pathogenic mechanisms against mucosal surfaces and epithelial barriers in the paranasal sinuses. Video Abstract.


Subject(s)
Pseudomonas aeruginosa , Sinusitis , Humans , Staphylococcus aureus , Mucociliary Clearance , Sinusitis/microbiology , Sinusitis/pathology , Nasal Mucosa/metabolism , Nasal Mucosa/microbiology , Nasal Mucosa/pathology , Tight Junctions , Bacteria , Chronic Disease
5.
Expert Rev Clin Immunol ; 19(8): 911-920, 2023.
Article in English | MEDLINE | ID: mdl-37378564

ABSTRACT

INTRODUCTION: Chronic rhinosinusitis (CRS) is characterized by inflammation of the paranasal sinus mucosa persisting for more than 12 weeks. This condition is associated with reduced quality-of-life and causes a high direct and indirect economic burden. Several pathogenic factors have been attributed to CRS, including bacterial and fungal biofilms on the sinonasal mucosa. Biofilms are well-established contributors to recalcitrance to treatment in other chronic inflammatory mucosal conditions such as cystic fibrosis and otitis media. AREAS COVERED: This review will present an overview of the role of biofilms in CRS, including the evidence for biofilms being present on the sinonasal mucosa and their implications for disease severity. Furthermore, the interactions between biofilms and host-mediated immune factors are explored. EXPERT OPINION: The eradication of biofilms has been a focus of research shortly after their recognition as a cause of disease. The currently available methodologies for identifying biofilms on mucosal surfaces are not sufficiently well-developed to be used in a clinical setting. A more accurate, cheaper, faster approach for biofilm detection is necessary, and molecular techniques may provide the possibility for this.


Subject(s)
Rhinitis , Sinusitis , Humans , Nasal Mucosa/microbiology , Biofilms , Chronic Disease
6.
Toxins (Basel) ; 15(4)2023 03 24.
Article in English | MEDLINE | ID: mdl-37104175

ABSTRACT

Staphylococcus aureus is a Gram-positive bacterium, which can be found, as a commensal microorganism, on the skin surface or in the nasal mucosa of the human population. However, S. aureus may become pathogenic and cause severe infections, especially in hospitalized patients. As an opportunistic pathogen, in fact, S. aureus interferes with the host Ca2+ signaling, favoring the spread of the infection and tissue destruction. The identification of novel strategies to restore calcium homeostasis and prevent the associated clinical outcomes is an emerging challenge. Here, we investigate whether harzianic acid, a bioactive metabolite derived from fungi of the genus Trichoderma, could control S. aureus-induced Ca2+ movements. First, we show the capability of harzianic acid to complex calcium divalent cations, using mass spectrometric, potentiometric, spectrophotometric, and nuclear magnetic resonance techniques. Then, we demonstrate that harzianic acid significantly modulates Ca2+ increase in HaCaT (human keratinocytes) cells incubated with S. aureus. In conclusion, this study suggests harzianic acid as a promising therapeutical alternative against diseases associated with Ca2+ homeostasis alteration.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Staphylococcus aureus/metabolism , Calcium/metabolism , Keratinocytes , Nasal Mucosa/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
7.
Infect Control Hosp Epidemiol ; 44(3): 447-452, 2023 03.
Article in English | MEDLINE | ID: mdl-35450544

ABSTRACT

OBJECTIVE: To assess the incidence rate of S. aureus colonization at baseline along with the mupirocin susceptibility (or resistance) rate in patients in a neonatal intensive care unit (NICU) and a pediatric intensive care unit (PICU) in conjunction with the implementation of universal decolonization as the standard of care. DESIGN: Prospective cohort study. SETTING: Children's Hospital of Michigan (CHM) inpatient intensive care units (ICUs). PARTICIPANTS: Newly admitted pediatric patients to the CHM NICU or PICU aged between 1 day and ≤21 years. INTERVENTIONS: Baseline and follow-up S. aureus screening cultures were obtained before patients underwent universal decolonization with mupirocin 2% antibiotic ointment (intranasal and umbilical) and chlorhexidine baths as standard of care to reduce CLABSI rates. RESULTS: Baseline S. aureus colonization rates of new admissions to the CHM NICU and PICU were high at 32% and 29%, respectively. Baseline mupirocin susceptibility to any S. aureus growth was 98.4%. All baseline culture isolates whether positive for MRSA or MSSA, with one exception, had minimum inhibitory concentrations (MICs) of ≤0.19 µg/mL. All follow-up study cultures after universal decolonization at 7 days or beyond with any S. aureus growth had mupirocin MICs of ≤0.125 µg/mL. CONCLUSIONS: Baseline S. aureus colonization rates of new admissions to the CHM ICUs were high as was baseline mupirocin susceptibility. Follow-up cultures, albeit limited in number, did not detect increasing mupirocin MICs over 1 year, despite broad mupirocin exposure due to the implementation of universal decolonization.


Subject(s)
Drug Resistance, Bacterial , Intensive Care Units, Neonatal , Intensive Care Units, Pediatric , Mupirocin , Staphylococcus aureus , Staphylococcus aureus/drug effects , Mupirocin/pharmacology , Mupirocin/therapeutic use , Humans , Infant, Newborn , Child , Microbial Sensitivity Tests , Infant , Child, Preschool , Adolescent , Young Adult , Male , Female , Nasal Mucosa/drug effects , Nasal Mucosa/microbiology , Umbilical Cord/drug effects , Umbilical Cord/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Cohort Studies , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Bacterial/drug effects
8.
Mem Inst Oswaldo Cruz ; 117: e220058, 2022.
Article in English | MEDLINE | ID: mdl-36259791

ABSTRACT

BACKGROUND: Leprosy is curable by multidrug therapy (MDT) treatment regimen ranging from six to 12 months. The variable levels of tolerance and adherence among patients can, however, result in treatment failure and the emergence of drug-resistant strains. OBJECTIVES: Describe the impact of MDT over Mycobacterium leprae viability in patient's oral and nasal mucosa along treatment. METHODS: Mycobacterium leprae viability was monitored by quantitative polymerase chain reaction (qPCR) quantification of 16S rRNA in lateral and contralateral scrapings of oral and nasal mucosa of 10 multibacillary patients along the initial five months of treatment. FINDINGS: The results demonstrated high heterogenicity of M. leprae viability among patients and between nasal and oral samples. Of six patients who presented good adherence and tolerance to the treatment, only four displayed absence of M. leprae viability in both samples three months after the first MDT dose, while for the other two, the absence of M. leprae viability in the oral and nasal cavities was only detected five months after the first dose. MAIN CONCLUSIONS: We concluded that qPCR of 16S rRNA for the determination of M. leprae viability in nasal and oral scraping samples could represent an interesting approach to monitor treatment efficacy.


Subject(s)
Leprostatic Agents , Mycobacterium leprae , Humans , Mycobacterium leprae/genetics , RNA, Ribosomal, 16S/genetics , Leprostatic Agents/therapeutic use , Drug Therapy, Combination , Nasal Mucosa/microbiology , DNA, Bacterial/genetics
9.
NPJ Biofilms Microbiomes ; 8(1): 26, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35418111

ABSTRACT

Our recent study presented that human nasal commensal Staphylococcus epidermidis could potentiate antiviral immunity in the nasal mucosa through interferon-related innate responses. Here, we found that human nasal commensal S. epidermidis promoted protease-protease inhibitor balance in favor of the host and prevented influenza A virus (IAV) replication in the nasal mucosa and lungs. A relatively higher induction of Serpine1 exhibited in S. epidermidis-inoculated nasal epithelium and S. epidermidis-induced Serpine1 significantly decreased the expression of serine proteases. Furthermore, the transcription of urokinase plasminogen activator (uPA) and Serpine1 was biologically relevant in S. epidermidis-inoculated nasal epithelium, and the induction of uPA might be related to the sequential increase of Serpine1 in human nasal epithelium. Our findings reveal that human nasal commensal S. epidermidis manipulates the cellular environment lacking serine proteases in the nasal epithelium through Serpine1 induction and disturbs IAV spread to the lungs at the level of the nasal mucosa.


Subject(s)
Influenza A virus , Nasal Mucosa , Staphylococcus epidermidis , Virus Internalization , Humans , Influenza A virus/physiology , Interferons , Nasal Mucosa/microbiology , Nasal Mucosa/virology , Serine Proteases
10.
Front Cell Infect Microbiol ; 12: 786481, 2022.
Article in English | MEDLINE | ID: mdl-35252024

ABSTRACT

Age-related changes in nasal bacterial microbiota of patients with chronic rhinosinusitis (CRS) remains unclear. In this study, we aimed to identify distinct characteristics of nasal bacterial microbiota between aged and younger patients with CRS through 16S rDNA gene sequencing. Patients with CRS undergoing endoscopic sinus surgery were recruited and separated into aged (≥60 years, median age = 66 years, N = 17) and younger (<60 years, median age = 35.5 years, N = 14) patients. Diversity, bacterial composition and metabolic activities of nasal microbiota between aged and younger patients were compared. Results have shown that levels of OTUs (p = 0.0173) and microbiota diversity (all p < 0.05) decreased significantly in aged patients. The abundance of phylum Actinobacteria, and genus Corynebacterium were significantly higher in aged patients, while the abundance of phylum Bacteroidetes, Fusobacteria, and genus Fusobacterium, Peptoniphilus were significantly higher in younger patients. In addition, predicted functional profiles have revealed that 41 KEGG pathways involving in 12 metabolic pathways, 4 genetic information processing, 3 environmental information processing, 4 cellular processes, 8 organismal systems, 6 human diseases, and 4 unclassified pathways were identified. Among which, the vast majority of metabolic activities are involved in replication and repair, membrane transport, translation, and the metabolism of amino acid, carbohydrate, energy, cofactors and vitamins, and nucleotide. On the level of the thirdly bacterial metabolic pathways, purine metabolism, glycine, serine and threonine metabolism, valine, leucine and isoleucine biosynthesis, glycolysis/gluconeogenesis and phenylalanine, tyrosine and tryptophan biosynthesis are significantly up-regulated while carbon fixation pathways in prokaryotesand methane metabolism are significantly down-regulated in aged patients. Overall, our analysis revealed that age-related physiological and pathological changes on the nasal mucosal surface may alter the host immune response and be highly associated with the nasal bacterial microbiota of patients with CRS. However, future studies are needed to elucidate the causal relationship.


Subject(s)
Microbiota , Rhinitis , Sinusitis , Adult , Aged , Chronic Disease , Humans , Middle Aged , Nasal Mucosa/microbiology , RNA, Ribosomal, 16S/genetics , Sinusitis/microbiology
11.
PLoS Negl Trop Dis ; 16(2): e0010191, 2022 02.
Article in English | MEDLINE | ID: mdl-35192629

ABSTRACT

Leptospirosis is a fatal zoonosis caused by contact between skin or a mucosal surface and contaminated soil or water. Hamsters were infected by intraperitoneal injection fto establish experimental leptospirosis, which is not a natural route of infection. There are no reports of nasal mucosal infection in hamsters. In this study, infection of the nasal mucosa was performed to establish a model of natural infection. Both methods of infection can cause lethal models with similar symptoms in the later stages of infection, such as weight loss, blood concentration, increased neutrophils (GRAN), and decreased lymphocytes (LYM) in the blood, severe organ damage and liver function obstruction. The burden of Leptospira in the organs and blood was lower in the mucosal inoculation groups at 1 day after infection. However, mucosal infection induced a higher Leptospira burden in urine than intraperitoneal infection in the late stages of infection. After nasal mucosal infection, antibody levels were higher and lasted longer. These results indicated that the route of nasal mucosal infection is a good choice for studying leptospirosis in hamsters.


Subject(s)
Disease Models, Animal , Leptospira/physiology , Leptospirosis/microbiology , Nasal Mucosa/microbiology , Animals , Antibodies, Bacterial/blood , Cricetinae , Female , Humans , Leptospira/immunology , Leptospirosis/blood , Leptospirosis/mortality , Liver/microbiology
12.
Minerva Pediatr (Torino) ; 74(5): 586-592, 2022 10.
Article in English | MEDLINE | ID: mdl-32731730

ABSTRACT

Chronic rhinosinusitis (CRS) is defined as an inflammatory disorder of the paranasal sinuses and of the nasal mucosa that lasts 12 weeks or longer. In CRS microbes contribute to the disease pathogenesis. Clinical microbiology is focused on finding single pathogens that causes the disease and the main goal is the use of antibiotics to kill bacteria. Efforts to achieve a better understanding of CRS include the study of the sinus microbiome, and to evaluate the ability of probiotics to augment homeostasis and modulate the immune response of the host mucosa. This review provides an update on the role of the microbiome in CRS. The study was conducted using two databases: PubMed and Science Direct. We searched for articles in English that matched the review topic. We first used the abstracts of articles to assess whether they met the inclusion criteria. We also reviewed the references of the selected articles and read those with titles that might be of interest. Several studies have shown that endogenous microbiome dysbiosis can impact mucosa health and disease severity. Some bacterial species presenting protective or pathogenic effect. Antimicrobial agents can create a similar disruption and impact the nasal microbiome balance. On the other hand, probiotics offers a promising avenue for developing systemic and topical therapies geared towards strategic manipulation of the biological host load, thereby augmenting immune homeostasis. A better comprehension of sinus-nasal microbiome in healthy and in CRS patients and the link with different CRS phenotype can help in developing new prognostics, diagnostics, and therapeutics strategies. Going forward, the use of probiotics can restore the native sinus ecology with significant therapeutic and preventive implications.


Subject(s)
Microbiota , Paranasal Sinuses , Rhinitis , Sinusitis , Humans , Rhinitis/therapy , Rhinitis/microbiology , Sinusitis/therapy , Sinusitis/microbiology , Paranasal Sinuses/microbiology , Microbiota/genetics , Nasal Mucosa/microbiology , Bacteria/genetics , Chronic Disease
13.
Front Immunol ; 12: 732029, 2021.
Article in English | MEDLINE | ID: mdl-34804016

ABSTRACT

Transient receptor potential (TRP) channels, neuronal stimulations widely known to be associated with thermal responses, pain induction, and osmoregulation, have been shown in recent studies to have underlying mechanisms associated with inflammatory responses. The role of TRP channels on inflammatory milieu during bacterial infections has been widely demonstrated. It may vary among types of channels/pathogens, however, and it is not known how TRP channels function during pneumococcal infections. Streptococcus pneumoniae can cause severe infections such as pneumonia, bacteremia, and meningitis, with systemic inflammatory responses. This study examines the role of TRP channels (TRPV1 and TRPV4) for pneumococcal nasal colonization and subsequent development of invasive pneumococcal disease in a mouse model. Both TRPV1 and TRPV4 channels were shown to be related to regulation of the development of pneumococcal diseases. In particular, the influx of neutrophils (polymorphonuclear cells) in the nasal cavity and the bactericidal activity were significantly suppressed among TRPV4 knockout mice. This may lead to severe pneumococcal pneumonia, resulting in dissemination of the bacteria to various organs and causing high mortality during influenza virus coinfection. Regulating host immune responses by TRP channels could be a novel strategy against pathogenic microorganisms causing strong local/systemic inflammation.


Subject(s)
Nasal Mucosa/metabolism , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/pathogenicity , TRPV Cation Channels/metabolism , Animals , Coinfection , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions , Inflammation Mediators/metabolism , Influenza A Virus, H3N2 Subtype/pathogenicity , Mice, Inbred C57BL , Mice, Knockout , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nasal Mucosa/virology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/microbiology , Phagocytosis , Pneumococcal Infections/genetics , Pneumococcal Infections/immunology , Pneumococcal Infections/microbiology , Signal Transduction , Streptococcus pneumoniae/immunology , TRPV Cation Channels/genetics , Virulence
14.
Microbiol Spectr ; 9(2): e0036721, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34668729

ABSTRACT

The objective of this study was to examine the nasal microbiota in relation to otitis media (OM) status and nose health in Indigenous Australian children. Children 2 to 7 years of age were recruited from two northern Australian (Queensland) communities. Clinical histories were obtained through parent interviews and reviews of the medical records. Nasal cavity swab samples were obtained, and the children's ears, nose, and throat were examined. DNA was extracted and analyzed by 16S rRNA amplicon next-generation sequencing of the V3/V4 region, in combination with previously generated culture data. A total of 103 children were recruited (mean age, 4.7 years); 17 (16.8%) were healthy, i.e., normal examination results and no history of OM. The nasal microbiota differed significantly in relation to OM status and nose health. Children with historical OM had greater relative abundance of Moraxella, compared to healthy children, despite both having healthy ears at the time of swabbing. Children with healthy noses had greater relative abundance of Staphylococcus aureus, compared to those with rhinorrhea. Dolosigranulum was correlated with Corynebacterium in healthy children. Haemophilus and Streptococcus were correlated across phenotypes. Ornithobacterium was absent or was present with low relative abundance in healthy children and clustered around otopathogens. It correlated with Helcococcus and Dichelobacter. Dolosigranulum and Corynebacterium form a synergism that promotes upper respiratory tract (URT)/ear health in Indigenous Australian children. Ornithobacterium likely represents "Candidatus Ornithobacterium hominis" and in this population is correlated with a novel bacterium that appears to be related to poor URT/ear health. IMPORTANCE Recurring and chronic infections of the ear (OM) are disproportionately prevalent in disadvantaged communities across the globe and, in particular, within Indigenous communities. Despite numerous intervention strategies, OM persists as a major health issue and is the leading cause of preventable hearing loss. In disadvantaged communities, this hearing loss is associated with negative educational and social development outcomes, and consequently, poorer employment prospects and increased contact with the justice system in adulthood. Thus, a better understanding of the microbial ecology is needed in order to identify new targets to treat, as well as to prevent the infections. This study used a powerful combination of 16S rRNA gene sequencing and extended culturomics to show that Dolosigranulum pigrum, a bacterium previously identified as a candidate protective species, may require cocolonization with Corynebacterium pseudodiphtheriticum in order to prevent OM. Additionally, emerging and potentially novel pathogens and bacteria were identified.


Subject(s)
Bacteria/classification , Ear/microbiology , Microbiota/genetics , Nasal Cavity/microbiology , Native Hawaiian or Other Pacific Islander/statistics & numerical data , Otitis Media/epidemiology , Australia/epidemiology , Bacteria/genetics , Bacteria/isolation & purification , Child , Child, Preschool , Female , Health Status , Humans , Male , Microbiota/physiology , Nasal Mucosa/microbiology , Nasopharynx/microbiology , Otitis Media/microbiology , Persistent Infection/microbiology , RNA, Ribosomal, 16S/genetics , Respiratory System/microbiology
15.
EBioMedicine ; 72: 103579, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34571365

ABSTRACT

BACKGROUND: Persistent carriage of pneumococcal vaccine serotypes has occurred after introduction of PCV13 vaccination in Africa but the mechanisms are unclear. We tested the feasibility of using a human pneumococcal challenge model in Malawi to understand immune correlates of protection against carriage and to trial alternative vaccine candidates. We aimed to identify a dose of Streptococcus pneumoniae serotype 6B sufficient to establish nasopharyngeal carriage in 40% of those nasally inoculated and evaluate nasal mucosal immunity before and after experimental inoculation. METHODS: Healthy student volunteers were recruited and inoculated with saline, 20,000 CFU/naris or 80,000 CFU/naris of Streptococcus pneumoniae serotype 6B Post inoculation carriage was determined by nasal sampling for bacterial culture and lytA PCR. Immunological responses were measured in serum and nasal mucosal biopsies before and after bacterial inoculation. FINDINGS: Twenty-four subjects completed the feasibility protocol with minimal side effects. pneumococcal carriage was established in 0/6, 3/9 and 4/9 subjects in the saline, 20,000 CFU/naris and 80,000 CFU/naris groups, respectively. Incidental (natural) serotype carriage was common (7/24 participants carried non-6B strains, 29.2%. Experimentally induced type 6B pneumococcal carriage was associated with pro-inflammatory nasal mucosal responses prior to inoculation and altered mucosal recruitment of immune cells post bacterial challenge. There was no association with serum anti-capsular antibody. INTERPRETATION: The serotype 6B experimental human pneumococcal carriage model is feasible in Malawi and can now be used to determine the immunological correlates of protection against carriage and vaccine efficacy in this population. FUNDING: None.


Subject(s)
Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Streptococcus pneumoniae/immunology , Adult , Antibodies, Bacterial/immunology , Carrier State/immunology , Carrier State/microbiology , Feasibility Studies , Female , Humans , Malawi , Male , Nasal Mucosa/immunology , Nasal Mucosa/microbiology , Nasopharynx/immunology , Nasopharynx/microbiology , Pneumococcal Vaccines/immunology , Serogroup , Vaccination/methods , Vaccine Efficacy , Young Adult
16.
J Infect Dev Ctries ; 15(7): 1014-1020, 2021 07 31.
Article in English | MEDLINE | ID: mdl-34343127

ABSTRACT

INTRODUCTION: Aspergillus exhibits a wide variation of susceptibility against antifungals according to genetic and environmental factors. Identification to the species level is necessary for appropriate treatment. Our objective was to determine the Aspergillus species involved in invasive pulmonary aspergillosis (IPA) among ICU patients in Jakarta, Indonesia. METHODOLOGY: The incidence of IPA in ICU patients at six hospitals in Jakarta from October 2012 - January 2015 was investigated. It involved a collection of endotracheal aspirates (ETA), nasal swabs and environmental samples around the hospitals, phenotypic screening, molecular characterization, and antifungal susceptibility testing. RESULTS: Of the 405 patients investigated, 31 patients (7.7%) were diagnosed with putative IPA, from whom 45 Aspergillus isolates were collected. Aspergillus isolates were identified from pulmonary secretions in 24 patients, from nasal swabs in 7 patients and from both pulmonary secretions and nasal swabs in 7 patients. The phenotypic method showed 33 isolates of Aspergillus flavus (73.4%), nine Aspergillus fumigatus (20%), two Aspergillus niger (4.4%), and one Aspergillus nidulans (2.2%) isolate. Molecular identification showed 27 isolates of A. flavus (60.0%), eight isolates of A. fumigatus (17.8%), two isolates of A. niger (4.4%) and one isolate of A. nidulans (2.2%), while seven isolates (15.6%) were cryptic species or mixed isolates. CONCLUSIONS: Susceptibility testing showed all isolates were susceptible to amphotericin B, azoles and micafungin. Aspergillus flavus was the main causative organism in IPA cases in Jakarta, followed by A. fumigatus.


Subject(s)
Antifungal Agents/pharmacology , Aspergillus/classification , Aspergillus/drug effects , Intensive Care Units , Invasive Pulmonary Aspergillosis/microbiology , Antifungal Agents/therapeutic use , Aspergillus/genetics , Aspergillus/isolation & purification , Cohort Studies , Environmental Microbiology , Humans , Incidence , Indonesia/epidemiology , Invasive Pulmonary Aspergillosis/diagnosis , Invasive Pulmonary Aspergillosis/drug therapy , Invasive Pulmonary Aspergillosis/epidemiology , Microsatellite Repeats , Nasal Mucosa/microbiology , Phenotype , Prospective Studies , Trachea/microbiology
17.
mBio ; 12(4): e0159821, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34399617

ABSTRACT

The gut microbiota plays a critical role in the induction of adaptive immune responses to influenza virus infection. However, the role of nasal bacteria in the induction of the virus-specific adaptive immunity is less clear. Here, we found that disruption of nasal bacteria by intranasal application of antibiotics before influenza virus infection enhanced the virus-specific antibody response in a MyD88-dependent manner. Similarly, disruption of nasal bacteria by lysozyme enhanced antibody responses to intranasally administered influenza virus hemagglutinin (HA) vaccine in a MyD88-dependent manner, suggesting that intranasal application of antibiotics or lysozyme could release bacterial pathogen-associated molecular patterns (PAMPs) from disrupted nasal bacteria that act as mucosal adjuvants by activating the MyD88 signaling pathway. Since commensal bacteria in the nasal mucosal surface were significantly lower than those in the oral cavity, intranasal administration of HA vaccine alone was insufficient to induce the vaccine-specific antibody response. However, intranasal supplementation of cultured oral bacteria from a healthy human volunteer enhanced antibody responses to an intranasally administered HA vaccine. Finally, we demonstrated that oral bacteria combined with an intranasal vaccine protect from influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Our results reveal the role of nasal bacteria in the induction of the virus-specific adaptive immunity and provide clues for developing better intranasal vaccines. IMPORTANCE Intranasal vaccination induces the nasal IgA antibody which is protective against respiratory viruses, such as influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, understanding how mucosal immune responses are elicited following viral infection is important for developing better vaccines. Here, we focused on the role of nasal commensal bacteria in the induction of immune responses following influenza virus infection. To deplete nasal bacteria, we intranasally administered antibiotics to mice before influenza virus infection and found that antibiotic-induced disruption of nasal bacteria could release bacterial components which stimulate the virus-specific antibody responses. Since commensal bacteria in nasal mucosa were significantly lower than those in the oral cavity, intranasal administration of split virus vaccine alone was insufficient to induce the vaccine-specific antibody response. However, intranasal supplementation of cultured oral bacteria from a healthy human volunteer enhanced antibody responses to the intranasally administered vaccine. Therefore, both integrity and amounts of nasal bacteria may be critical for an effective intranasal vaccine.


Subject(s)
Bacteria/immunology , COVID-19 Vaccines/immunology , COVID-19/prevention & control , Influenza Vaccines/immunology , Nasal Mucosa/microbiology , Orthomyxoviridae Infections/prevention & control , Adaptive Immunity/immunology , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antibodies, Viral/immunology , Cell Line , Chlorocebus aethiops , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Mucosal/immunology , Influenza A Virus, H1N1 Subtype/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Myeloid Differentiation Factor 88/metabolism , Nasal Mucosa/immunology , Pathogen-Associated Molecular Pattern Molecules/immunology , SARS-CoV-2/immunology , Vaccination/methods , Vero Cells
18.
Mucosal Immunol ; 14(5): 1183-1202, 2021 09.
Article in English | MEDLINE | ID: mdl-33976385

ABSTRACT

Understanding the mechanism of protective immunity in the nasal mucosae is central to the design of more effective vaccines that prevent nasal infection and transmission of Bordetella pertussis. We found significant infiltration of IL-17-secreting CD4+ tissue-resident memory T (TRM) cells and Siglec-F+ neutrophils into the nasal tissue during primary infection with B. pertussis. Il17A-/- mice had significantly higher bacterial load in the nasal mucosae, associated with significantly reduced infiltration of Siglec-F+ neutrophils. Re-infected convalescent mice rapidly cleared B. pertussis from the nasal cavity and this was associated with local expansion of IL-17-producing CD4+ TRM cells. Depletion of CD4 T cells from the nasal tissue during primary infection or after re-challenge of convalescent mice significantly delayed clearance of bacteria from the nasal mucosae. Protection was lost in Il17A-/- mice and this was associated with significantly less infiltration of Siglec-F+ neutrophils and antimicrobial peptide (AMP) production. Finally, depletion of neutrophils reduced the clearance of B. pertussis following re-challenge of convalescent mice. Our findings demonstrate that IL-17 plays a critical role in natural and acquired immunity to B. pertussis in the nasal mucosae and this effect is mediated by mobilizing neutrophils, especially Siglec-F+ neutrophils, which have high neutrophil extracellular trap (NET) activity.


Subject(s)
Bordetella pertussis/immunology , Interleukin-17/genetics , Neutrophils/immunology , Neutrophils/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Whooping Cough/etiology , Whooping Cough/metabolism , Adaptive Immunity , Animals , Biomarkers , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunohistochemistry , Immunophenotyping , Interleukin-17/metabolism , Lymphocyte Depletion , Mice , Mice, Knockout , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Nasal Mucosa/microbiology , Neutrophil Activation/genetics , Neutrophil Activation/immunology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology
19.
Int J Mol Sci ; 22(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33947066

ABSTRACT

The pathogenesis of nasal inflammatory diseases is related to various factors such as anatomical structure, heredity, and environment. The nasal microbiota play a key role in coordinating immune system functions. Dysfunction of the microbiota has a significant impact on the occurrence and development of nasal inflammation. This review will introduce the positive and negative roles of microbiota involved in immunity surrounding nasal mucosal diseases such as chronic sinusitis and allergic rhinitis. In addition, we will also introduce recent developments in DNA sequencing, metabolomics, and proteomics combined with computation-based bioinformatics.


Subject(s)
Microbiota , Nasal Cavity/microbiology , Nasal Mucosa/microbiology , Rhinitis/microbiology , Sinusitis/microbiology , Adult , Antigens, Bacterial/immunology , Child , Chronic Disease , Dysbiosis/immunology , Dysbiosis/microbiology , Humans , Metabolomics/methods , Nasal Cavity/immunology , Nasal Mucosa/immunology , Proteomics/methods , Rhinitis/immunology , Rhinitis, Allergic/immunology , Rhinitis, Allergic/microbiology , Sequence Analysis, DNA/methods , Sinusitis/immunology
20.
Otolaryngol Head Neck Surg ; 165(6): 895-898, 2021 12.
Article in English | MEDLINE | ID: mdl-33781114

ABSTRACT

This proof-of-concept study demonstrates that sinonasal microbiome sequencing with suctioned nasal secretions in children is technically feasible. Eighteen children were enrolled: 11 with symptoms of rhinosinusitis and 7 controls. Samples were collected via a sterile flexible respiratory suction catheter into a suction trap. Samples were analyzed with 16S ribosomal RNA sequencing and subsequent phylogenic mapping. The log2-fold differential abundance in class demonstrated significantly higher quantities of bacteria from the classes Negativicutes, Bacilli, Mollicutes, and Alphaproteobacteria as compared with controls, with a false discovery rate-corrected P value <.01 for all 4. The experimental group showed a higher range of alpha diversity than the control group, although none of the measures of alpha diversity showed a statistically significant difference. Overall our study demonstrates that sinonasal microbiome sequencing with suctioned nasal secretions is a readily available and technically feasible alternative for study of the pediatric sinonasal microbiome.


Subject(s)
Bacteria/isolation & purification , Microbiota , Nasal Mucosa/microbiology , Rhinitis/microbiology , Sinusitis/microbiology , Case-Control Studies , Child , Child, Preschool , Chronic Disease , Female , Humans , Infant , Male , Pilot Projects , Proof of Concept Study
SELECTION OF CITATIONS
SEARCH DETAIL
...