Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 693
Filtrar
1.
Nat Commun ; 15(1): 5817, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38987270

RESUMO

Respiratory infections caused by the human fungal pathogen Aspergillus fumigatus are a major cause of mortality for immunocompromised patients. Exposure to these pathogens occurs through inhalation, although the role of the respiratory epithelium in disease pathogenesis has not been fully defined. Employing a primary human airway epithelial model, we demonstrate that fungal melanins potently block the post-translational secretion of the chemokines CXCL1 and CXCL8 independent of transcription or the requirement of melanin to be phagocytosed, leading to a significant reduction in neutrophil recruitment to the apical airway both in vitro and in vivo. Aspergillus-derived melanin, a major constituent of the fungal cell wall, dampened airway epithelial chemokine secretion in response to fungi, bacteria, and exogenous cytokines. Furthermore, melanin muted pathogen-mediated calcium fluxing and hindered actin filamentation. Taken together, our results reveal a critical role for melanin interaction with airway epithelium in shaping the host response to fungal and bacterial pathogens.


Assuntos
Aspergillus fumigatus , Cálcio , Quimiocina CXCL1 , Interleucina-8 , Melaninas , Melaninas/metabolismo , Humanos , Interleucina-8/metabolismo , Cálcio/metabolismo , Quimiocina CXCL1/metabolismo , Animais , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Camundongos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Quimiocinas/metabolismo , Camundongos Endogâmicos C57BL
2.
Nat Microbiol ; 9(7): 1725-1737, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38858595

RESUMO

Pseudomonas aeruginosa, a leading cause of severe hospital-acquired pneumonia, causes infections with up to 50% mortality rates in mechanically ventilated patients. Despite some knowledge of virulence factors involved, it remains unclear how P. aeruginosa disseminates on mucosal surfaces and invades the tissue barrier. Using infection of human respiratory epithelium organoids, here we observed that P. aeruginosa colonization of apical surfaces is promoted by cyclic di-GMP-dependent asymmetric division. Infection with mutant strains revealed that Type 6 Secretion System activities promote preferential invasion of goblet cells. Type 3 Secretion System activity by intracellular bacteria induced goblet cell death and expulsion, leading to epithelial rupture which increased bacterial translocation and dissemination to the basolateral epithelium. These findings show that under physiological conditions, P. aeruginosa uses coordinated activity of a specific combination of virulence factors and behaviours to invade goblet cells and breach the epithelial barrier from within, revealing mechanistic insight into lung infection dynamics.


Assuntos
Células Caliciformes , Infecções por Pseudomonas , Pseudomonas aeruginosa , Mucosa Respiratória , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Pseudomonas aeruginosa/fisiologia , Células Caliciformes/microbiologia , Células Caliciformes/metabolismo , Humanos , Mucosa Respiratória/microbiologia , Mucosa Respiratória/citologia , Infecções por Pseudomonas/microbiologia , Sistemas de Secreção Tipo VI/genética , Sistemas de Secreção Tipo VI/metabolismo , Fatores de Virulência/metabolismo , Fatores de Virulência/genética , Sistemas de Secreção Tipo III/metabolismo , Sistemas de Secreção Tipo III/genética , GMP Cíclico/análogos & derivados , GMP Cíclico/metabolismo , Organoides/microbiologia , Translocação Bacteriana
3.
Methods Mol Biol ; 2813: 137-144, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38888776

RESUMO

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.


Assuntos
Técnicas de Cultura de Células , Células Epiteliais , Humanos , Técnicas de Cultura de Células/métodos , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Células Epiteliais/citologia , Brônquios/citologia , Mucosa Respiratória/citologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Ar , Células Cultivadas , Doenças Transmissíveis/microbiologia
4.
mBio ; 15(7): e0113024, 2024 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-38934617

RESUMO

Type III interferon signaling contributes to the pathogenesis of the important human pathogen Staphylococcus aureus in the airway. Little is known of the cellular factors important in this response. Using Ifnl2-green fluorescent protein reporter mice combined with flow cytometry and cellular depletion strategies, we demonstrate that the alveolar macrophage is the primary producer of interferon lambda (IFN-λ) in response to S. aureus in the airway. Bone marrow chimeras showed reduced bacterial burden in IFN-λ receptor (IFNLR1)-deficient recipient mice, indicative that non-hematopoietic cells were important for pathogenesis, in addition to significant reductions in pulmonary inflammation. These observations were confirmed through the use of an airway epithelial-specific IFNLR knockout mouse. Our data suggest that upon entry to the airway, S. aureus activates alveolar macrophages to produce type III IFN that is subsequently sensed by the airway epithelium. Future steps will determine how signaling from the epithelium then exerts its influence on bacterial clearance. These results highlight the important, yet sometimes detrimental, role of type III IFN signaling during infection and the impact the airway epithelium plays during host-pathogen interactions.IMPORTANCEThe contribution of type III interferon signaling to the control of bacterial infections is largely unknown. We have previously demonstrated that it contributes to the pathogenesis of acute Staphylococcus aureus respiratory infection. In this report, we document the importance of two cell types that underpin this pathogenesis. We demonstrate that the alveolar macrophage is the cell that is responsible for the production of type III interferon and that this molecule is sensed by airway epithelial cells, which impacts both bacterial clearance and induction of inflammation. This work sheds light on the first two aspects of this important pathogenic cascade.


Assuntos
Interferons , Macrófagos Alveolares , Camundongos Knockout , Infecções Estafilocócicas , Staphylococcus aureus , Animais , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/genética , Camundongos , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/imunologia , Infecções Estafilocócicas/microbiologia , Interferons/metabolismo , Interferons/genética , Interferons/imunologia , Camundongos Endogâmicos C57BL , Interações Hospedeiro-Patógeno , Transdução de Sinais , Mucosa Respiratória/microbiologia , Interferon lambda , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Virulência
5.
PLoS Pathog ; 20(5): e1011453, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38820569

RESUMO

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.


Assuntos
Biofilmes , Fibrose Cística , Células Epiteliais , Óperon , Potássio , Infecções por Pseudomonas , Pseudomonas aeruginosa , Biofilmes/crescimento & desenvolvimento , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/fisiologia , Humanos , Fibrose Cística/microbiologia , Fibrose Cística/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/metabolismo , Potássio/metabolismo , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia
6.
Nat Commun ; 15(1): 3666, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38693120

RESUMO

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.


Assuntos
Antígeno CD47 , Células Epiteliais , Infecções Estafilocócicas , Staphylococcus aureus , Superinfecção , Antígeno CD47/metabolismo , Antígeno CD47/genética , Humanos , Animais , Superinfecção/microbiologia , Camundongos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Influenza Humana/metabolismo , Influenza Humana/imunologia , Influenza Humana/virologia , Aderência Bacteriana , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Camundongos Endogâmicos C57BL , Brônquios/metabolismo , Brônquios/citologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Camundongos Knockout , Vírus da Influenza A Subtipo H1N1
7.
PLoS Pathog ; 20(5): e1012111, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38718049

RESUMO

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.


Assuntos
Infecções Pneumocócicas , Streptococcus pneumoniae , Animais , Infecções Pneumocócicas/microbiologia , Infecções Pneumocócicas/imunologia , Camundongos , Humanos , Animais Recém-Nascidos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Mucosa Respiratória/microbiologia , Mucosa Respiratória/metabolismo , Feminino , Nasofaringe/microbiologia
8.
Front Cell Infect Microbiol ; 14: 1346087, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38736751

RESUMO

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.


Assuntos
Homeostase , Imunidade Inata , Mucosa Intestinal , Humanos , Mucosa Intestinal/microbiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Animais , Mucosa Respiratória/microbiologia , Mucosa Respiratória/imunologia , Células Epiteliais/microbiologia , Transdução de Sinais , Imunidade Adaptativa , Macrófagos/imunologia , Macrófagos/microbiologia , Interações Hospedeiro-Patógeno
9.
Vet Res Commun ; 48(3): 1481-1495, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38336962

RESUMO

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.


Assuntos
Infecções por Bordetella , Bordetella bronchiseptica , Mucosa Respiratória , Animais , Coelhos , Bordetella bronchiseptica/efeitos dos fármacos , Infecções por Bordetella/veterinária , Infecções por Bordetella/microbiologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/microbiologia , Aderência Bacteriana/efeitos dos fármacos , Carboidratos/farmacologia , Acetilglucosamina/farmacologia , Infecções Respiratórias/veterinária , Infecções Respiratórias/microbiologia , Infecções Respiratórias/tratamento farmacológico , Pulmão/microbiologia , Pulmão/efeitos dos fármacos , Pulmão/patologia
10.
Am J Respir Crit Care Med ; 209(12): 1453-1462, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38324627

RESUMO

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.


Assuntos
Fibrose Cística , Transplante de Pulmão , Pulmão , Infecções por Pseudomonas , Pseudomonas aeruginosa , Mucosa Respiratória , Humanos , Fibrose Cística/microbiologia , Fibrose Cística/complicações , Feminino , Masculino , Adulto , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Infecções por Pseudomonas/microbiologia , Pulmão/microbiologia , Pulmão/patologia , Adulto Jovem , Células Epiteliais/microbiologia
11.
Indian J Tuberc ; 69 Suppl 2: S241-S245, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36400517

RESUMO

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.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Idoso , Imunidade Inata , Tuberculose/microbiologia , Mucosa Respiratória/microbiologia , Pulmão
12.
PLoS Pathog ; 18(1): e1010209, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35085362

RESUMO

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.


Assuntos
Infecções por Haemophilus/metabolismo , Haemophilus influenzae/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Animais , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Camundongos
13.
Infect Immun ; 90(1): e0042321, 2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-34662211

RESUMO

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.


Assuntos
Epitélio/microbiologia , Interações Hospedeiro-Patógeno , Fator 88 de Diferenciação Mieloide/deficiência , Mucosa Respiratória/microbiologia , Infecções Respiratórias/etiologia , Infecções Estreptocócicas/etiologia , Infecções Estreptocócicas/transmissão , Streptococcus pyogenes/fisiologia , Animais , Biópsia , Suscetibilidade a Doenças , Epitélio/patologia , Predisposição Genética para Doença , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Infiltração de Neutrófilos , Especificidade de Órgãos , Mucosa Respiratória/patologia , Infecções Respiratórias/patologia , Infecções Estreptocócicas/patologia
14.
Sci Rep ; 11(1): 19534, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34599245

RESUMO

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.


Assuntos
Microbiota , Mucosa Respiratória/microbiologia , Infecções Respiratórias/etiologia , Idoso , Idoso de 80 Anos ou mais , Suscetibilidade a Doenças , Feminino , Biblioteca Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Metagenoma , Metagenômica/métodos , Pessoa de Meia-Idade , Infecções Respiratórias/diagnóstico , Estudos Retrospectivos
15.
EBioMedicine ; 71: 103538, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34425308

RESUMO

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.


Assuntos
Asma/epidemiologia , Microbiota , Mucosa Respiratória/microbiologia , Fumar/epidemiologia , Adulto , Idoso , Asma/etiologia , Austrália/epidemiologia , Biologia Computacional/métodos , Suscetibilidade a Doenças , Feminino , Humanos , Masculino , Metagenômica/métodos , Pessoa de Meia-Idade , Vigilância da População , RNA Ribossômico 16S , Fumar/efeitos adversos , Fumar Tabaco
16.
Sci Rep ; 11(1): 17148, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34433845

RESUMO

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.


Assuntos
Benchmarking/métodos , Microbiota , Kit de Reagentes para Diagnóstico/normas , Mucosa Respiratória/microbiologia , Biomassa , Humanos , Metagenômica/métodos , Metagenômica/normas , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/normas , RNA Ribossômico 16S/genética , Saliva/microbiologia
17.
Cell Rep ; 36(3): 109392, 2021 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-34289364

RESUMO

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.


Assuntos
Quitina , Mamíferos , Proteínas de Membrana , Receptores de Reconhecimento de Padrão , Animais , Humanos , Camundongos , beta-Glucanas/metabolismo , Candida albicans/fisiologia , Membrana Celular/metabolismo , Quitina/metabolismo , Células Epiteliais/metabolismo , Células HeLa , Imunidade Inata , Inflamação/patologia , Mamíferos/metabolismo , Proteínas de Membrana/metabolismo , Células RAW 264.7 , Receptores de Reconhecimento de Padrão/metabolismo , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Transdução de Sinais
18.
Am J Respir Crit Care Med ; 204(6): 692-702, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34170795

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Fibrose Cística/tratamento farmacológico , Fibrose Cística/microbiologia , Pulmão/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Animais , Antibacterianos/uso terapêutico , Bactérias/isolamento & purificação , Líquido da Lavagem Broncoalveolar/microbiologia , Fibrose Cística/diagnóstico por imagem , Fibrose Cística/patologia , Pulmão/diagnóstico por imagem , Pulmão/microbiologia , Pulmão/patologia , Tomografia Computadorizada Multidetectores , Mucosa Respiratória/microbiologia , Mucosa Respiratória/patologia , Suínos
19.
J Cyst Fibros ; 20(4): 682-691, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34112603

RESUMO

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.


Assuntos
Brônquios/citologia , Brônquios/microbiologia , Fibrose Cística/complicações , Fibrose Cística/microbiologia , Células Epiteliais/imunologia , Inflamação/etiologia , Inflamação/microbiologia , Prevotella/fisiologia , Infecções por Pseudomonas/etiologia , Pseudomonas aeruginosa/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/microbiologia , Células Cultivadas , Fibrose Cística/imunologia , Humanos
20.
Front Immunol ; 12: 679829, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34108973

RESUMO

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.


Assuntos
Suscetibilidade a Doenças , Interações entre Hospedeiro e Microrganismos , Neoplasias Pulmonares/etiologia , Microbiota , Transformação Celular Neoplásica , Gerenciamento Clínico , Disbiose , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Imunidade , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/terapia , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...