Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.352
Filtrar
1.
Int J Mol Sci ; 25(16)2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39201753

RESUMEN

An application of CO2/HCO3--free solution (Zero-CO2) did not increase intracellular pH (pHi) in ciliated human nasal epithelial cells (c-hNECs), leading to no increase in frequency (CBF) or amplitude (CBA) of the ciliary beating. This study demonstrated that the pHi of c-hNECs expressing carbonic anhydrase IV (CAIV) is high (7.64), while the pHi of ciliated human bronchial epithelial cells (c-hBECs) expressing no CAIV is low (7.10). An extremely high pHi of c-hNECs caused pHi, CBF and CBA to decrease upon Zero-CO2 application, while a low pHi of c-hBECs caused them to increase. An extremely high pHi was generated by a high rate of HCO3- influx via interactions between CAIV and Na+/HCO3- cotransport (NBC) in c-hNECs. An NBC inhibitor (S0859) decreased pHi, CBF and CBA and increased CBF and CBA in c-hNECs upon Zero-CO2 application. In conclusion, the interactions of CAIV and NBC maximize HCO3- influx to increase pHi in c-hNECs. This novel mechanism causes pHi to decrease, leading to no increase in CBF and CBA in c-hNECs upon Zero-CO2 application, and appears to play a crucial role in maintaining pHi, CBF and CBA in c-hNECs periodically exposed to air (0.04% CO2) with respiration.


Asunto(s)
Bicarbonatos , Dióxido de Carbono , Anhidrasa Carbónica IV , Cilios , Células Epiteliales , Mucosa Nasal , Humanos , Concentración de Iones de Hidrógeno , Dióxido de Carbono/metabolismo , Cilios/metabolismo , Bicarbonatos/metabolismo , Células Epiteliales/metabolismo , Mucosa Nasal/metabolismo , Mucosa Nasal/citología , Anhidrasa Carbónica IV/metabolismo , Anhidrasa Carbónica IV/genética , Células Cultivadas , Simportadores de Sodio-Bicarbonato/metabolismo , Simportadores de Sodio-Bicarbonato/genética
2.
Nature ; 632(8025): 637-646, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39085603

RESUMEN

Nasal vaccination elicits a humoral immune response that provides protection from airborne pathogens1, yet the origins and specific immune niches of antigen-specific IgA-secreting cells in the upper airways are unclear2. Here we define nasal glandular acinar structures and the turbinates as immunological niches that recruit IgA-secreting plasma cells from the nasal-associated lymphoid tissues (NALTs)3. Using intact organ imaging, we demonstrate that nasal vaccination induces B cell expansion in the subepithelial dome of the NALT, followed by invasion into commensal-bacteria-driven chronic germinal centres in a T cell-dependent manner. Initiation of the germinal centre response in the NALT requires pre-expansion of antigen-specific T cells, which interact with cognate B cells in interfollicular regions. NALT ablation and blockade of PSGL-1, which mediates interactions with endothelial cell selectins, demonstrated that NALT-derived IgA-expressing B cells home to the turbinate region through the circulation, where they are positioned primarily around glandular acinar structures. CCL28 expression was increased in the turbinates in response to vaccination and promoted homing of IgA+ B cells to this site. Thus, in response to nasal vaccination, the glandular acini and turbinates provide immunological niches that host NALT-derived IgA-secreting cells. These cellular events could be manipulated in vaccine design or in the treatment of upper airway allergic responses.


Asunto(s)
Inmunoglobulina A , Tejido Linfoide , Mucosa Nasal , Células Plasmáticas , Linfocitos T , Cornetes Nasales , Animales , Femenino , Masculino , Ratones , Bacterias/inmunología , Movimiento Celular , Quimiocinas CC/inmunología , Quimiocinas CC/metabolismo , Centro Germinal/inmunología , Centro Germinal/citología , Inmunoglobulina A/inmunología , Inmunoglobulina A/metabolismo , Tejido Linfoide/inmunología , Tejido Linfoide/citología , Ratones Endogámicos C57BL , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Linfocitos T/inmunología , Linfocitos T/citología , Linfocitos T/metabolismo , Cornetes Nasales/citología , Cornetes Nasales/inmunología , Vacunación , Administración Intranasal , Vacunas/inmunología , Simbiosis
3.
Biofabrication ; 16(4)2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39038483

RESUMEN

The function of a well-differentiated nasal epithelium is largely affected by airflow-induced wall shear stress, yet fewin vitromodels recapitulate this dynamic condition. Models which do expose cells to airflow exclusively initiate flow after the differentiation process has occurred.In vivo, basal cells are constantly replenishing the epithelium under airflow conditions, indicating that airflow may affect the development and function of the differentiated epithelium. To address this gap in the field, we developed a physiologically relevant microphysiological model of the human nasal epithelium and investigated the effects of exposing cells to airflow during epithelial maturation at the air-liquid interface. The nasal airway-on-chip platform was engineered to mimic bi-directional physiological airflow during normal breathing. Primary human nasal epithelial cells were seeded on chips and subjected to either: (1) no flow, (2) single flow (0.5 dyne cm-2flow on Day 21 of ALI only), or (3) pre-conditioning flow (0.05 dyne cm-2on Days 14-20 and 0.5 dyne cm-2flow on Day 21) treatments. Cells exposed to pre-conditioning showed decreased morphological changes and mucus secretions, as well as decreased inflammation, compared to unconditioned cells. Our results indicate that flow exposure only post-differentiation may impose acute stress on cells, while pre-conditioning may potentiate a properly functioning epitheliumin vitro.


Asunto(s)
Células Epiteliales , Dispositivos Laboratorio en un Chip , Mucosa Nasal , Humanos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Mucosa Nasal/citología , Mucosa Nasal/metabolismo , Diferenciación Celular , Células Cultivadas , Aire , Modelos Biológicos , Ingeniería de Tejidos
5.
Cell Immunol ; 401-402: 104840, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38880071

RESUMEN

Sepsis is characterized by an exacerbated inflammatory response, driven by the overproduction of cytokines, a phenomenon known as a cytokine storm. This condition is further compounded by the extensive infiltration of M1 macrophages and the pyroptosis of these cells, leading to immune paralysis. To counteract this, we sought to transition M1 macrophages into the M2 phenotype and safeguard them from pyroptosis. For this purpose, we employed ectodermal mesenchymal stem cells (EMSCs) sourced from the nasal mucosa to examine their impact on both macrophages and septic animal models. The co-culture protocol involving LPS-stimulated rat bone marrow macrophages and EMSCs was employed to examine the paracrine influence of EMSCs on macrophages. The intravenous administration of EMSCs was utilized to observe the enhancement in the survival rate of septic rat models and the protection of associated organs. The findings indicated that EMSCs facilitated M2 polarization of macrophages, which were stimulated by LPS, and significantly diminished levels of pro-inflammatory cytokines and NLRP3. Furthermore, EMSCs notably restored the mitochondrial membrane potential (MMP) of macrophages through paracrine action, eliminated excess reactive oxygen species (ROS), and inhibited macrophage pyroptosis. Additionally, the systemic integration of EMSCs substantially reduced injuries to multiple organs and preserved the fundamental functions of the heart, liver, and kidney in CLP rats, thereby extending their survival.


Asunto(s)
Macrófagos , Células Madre Mesenquimatosas , Mucosa Nasal , Piroptosis , Sepsis , Animales , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratas , Mucosa Nasal/inmunología , Mucosa Nasal/citología , Sepsis/inmunología , Masculino , Ratas Sprague-Dawley , Trasplante de Células Madre Mesenquimatosas/métodos , Lipopolisacáridos , Citocinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Modelos Animales de Enfermedad , Técnicas de Cocultivo , Potencial de la Membrana Mitocondrial , Células Cultivadas
6.
Sci Rep ; 14(1): 14558, 2024 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-38914666

RESUMEN

Plants offer a cost-effective and scalable pharmaceutical platform devoid of host-derived contamination risks. However, their medical application is complicated by the potential for acute allergic reactions to external proteins. Developing plant-based protein therapeutics for localized diseases with non-invasive treatment modalities may capitalize on the benefits of plant proteins while avoiding their inherent risks. Dupilumab, which is effective against a variety of allergic and autoimmune diseases but has systemic responses and injection-related side effects, may be more beneficial if delivered locally using a small biological form. In this study, we engineered a single-chain variable fragment (scFv) of dupilumab, termed Dup-scFv produced by Nicotiana benthamiana, and evaluated its tissue permeability and anti-inflammatory efficacy in air-liquid interface cultured human nasal epithelial cells (HNECs). Despite showing 3.67- and 17-fold lower binding affinity for IL-4Ra in surface plasmon resonance assays and cell binding assays, respectively, Dup-scFv retained most of the affinity of dupilumab, which was originally high, with a dissociation constant (KD) of 4.76 pM. In HNECs cultured at the air-liquid interface, Dup-scFv administered on the air side inhibited the inflammatory marker CCL26 in hard-to-reach basal cells more effectively than dupilumab. In addition, Dup-scFv had an overall permeability of 0.8% across cell layers compared to undetectable levels of dupilumab. These findings suggest that plant-produced Dup-scFv can be delivered non-invasively to cultured HNESc to alleviate inflammatory signaling, providing a practical approach to utilize plant-based proteins for topical therapeutic applications.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Células Epiteliales , Nicotiana , Anticuerpos de Cadena Única , Humanos , Nicotiana/metabolismo , Anticuerpos Monoclonales Humanizados/farmacología , Células Epiteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Anticuerpos de Cadena Única/farmacología , Anticuerpos de Cadena Única/genética , Quimiocinas CC/metabolismo , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Células Cultivadas , Mucosa Nasal/metabolismo , Mucosa Nasal/citología , Mucosa Nasal/inmunología
7.
PLoS One ; 19(5): e0285655, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38753593

RESUMEN

BACKGROUND: Chronic rhinosinusitis (CRS) is an inflammatory disease affecting the sinuses or nose. Persistent inflammatory responses can lead to tissue remodeling, which is a pathological characteristics of CRS. Activation of fibroblasts in the nasal mucosal stroma, differentiation and collagen deposition, and subepithelial fibrosis have been associated with CRS. OBJECTIVES: We aimed to assess the inhibitory effects of doxycycline and deoxycholic acid-polyethyleneimine conjugate (DA3-Doxy) on myofibroblast differentiation and extracellular matrix (ECM) production in nasal fibroblasts stimulated with TGF-ß1. METHODS: To enhance efficacy, we prepared DA3-Doxy using a conjugate of low-molecular-weight polyethyleneimine (PEI) (MW 1800) and deoxycholic acid (DA) and Doxy. The synthesis of the DA3-Doxy polymer was confirmed using nuclear magnetic resonance, and the critical micelle concentration required for cationic micelle formation through self-assembly was determined. Subsequently, the Doxy loading efficiency of DA3 was assessed. The cytotoxicity of Doxy, DA3, PEI, and DA-Doxy in nasal fibroblasts was evaluated using the WST-1 assay. The anti-tissue remodeling and anti-inflammatory effects of DA3-Doxy and DA3 were examined using real-time polymerase chain reaction (Real-time PCR), immunocytochemistry, western blot, and Sircol assay. RESULTS: Both DA3 and DA3-Doxy exhibited cytotoxicity at 10 µg/ml in nasal fibroblasts. Doxy partially inhibited α-smooth muscle actin, collagen types I and III, and fibronectin. However, DA3-Doxy significantly inhibited α-SMA, collagen types I and III, and fibronectin at 5 µg/ml. DA3-Doxy also modulated TGF-ß1-induced changes in the expression of MMP 1, 2, and 9. Nonetheless, TGF-ß1-induced expression of MMP3 was further increased by DA3-Doxy. The expression of TIMP 1 and 2 was partially reduced with 5 µg/ml DA3-Doxy. CONCLUSIONS: Although initially developed for the delivery of genetic materials or drugs, DA3 exhibits inhibitory effects on myofibroblast differentiation and ECM production. Therefore, it holds therapeutic potential for CRS, and a synergistic effect can be expected when loaded with CRS treatment drugs.


Asunto(s)
Diferenciación Celular , Ácido Desoxicólico , Doxiciclina , Fibroblastos , Polietileneimina , Humanos , Polietileneimina/química , Polietileneimina/farmacología , Ácido Desoxicólico/química , Ácido Desoxicólico/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Diferenciación Celular/efectos de los fármacos , Doxiciclina/farmacología , Doxiciclina/química , Matriz Extracelular/metabolismo , Matriz Extracelular/efectos de los fármacos , Factor de Crecimiento Transformador beta1/metabolismo , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/citología , Actinas/metabolismo
8.
Artículo en Chino | MEDLINE | ID: mdl-38686485

RESUMEN

Allergic rhinitis is a chronic nasal mucosal inflammation characterized by upper airway hyperresponsiveness, involving a variety of immune cells and inflammatory mediators. Drugs, immunotherapy, and surgical operation are the principal treatments at present. The study found that mesenchymal stem cells have the ability of immune regulation and have a promising clinical application in the treatment of allergic rhinitis. In this review, the action mechanism of mesenchymal stem cells, the immunomodulatory effect of mesenchymal stem cells on the key cells of allergic rhinitis, and the challenges of clinical application are reviewed, to provide new directions for the treatment of allergic rhinitis.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Mucosa Nasal , Rinitis Alérgica , Humanos , Células Madre Mesenquimatosas/citología , Rinitis Alérgica/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Mucosa Nasal/citología
9.
J Virol ; 98(2): e0149423, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38294251

RESUMEN

Influenza B viruses (IBV) cocirculate with influenza A viruses (IAV) and cause periodic epidemics of disease, yet antibody and cellular responses following IBV infection are less well understood. Using the ferret model for antisera generation for influenza surveillance purposes, IAV resulted in robust antibody responses following infection, whereas IBV required an additional booster dose, over 85% of the time, to generate equivalent antibody titers. In this study, we utilized primary differentiated ferret nasal epithelial cells (FNECs) which were inoculated with IAV and IBV to study differences in innate immune responses which may result in differences in adaptive immune responses in the host. FNECs were inoculated with IAV (H1N1pdm09 and H3N2 subtypes) or IBV (B/Victoria and B/Yamagata lineages) and assessed for 72 h. Cells were analyzed for gene expression by quantitative real-time PCR, and apical and basolateral supernatants were assessed for virus kinetics and interferon (IFN), respectively. Similar virus kinetics were observed with IAV and IBV in FNECs. A comparison of gene expression and protein secretion profiles demonstrated that IBV-inoculated FNEC expressed delayed type-I/II IFN responses and reduced type-III IFN secretion compared to IAV-inoculated cells. Concurrently, gene expression of Thymic Stromal Lymphopoietin (TSLP), a type-III IFN-induced gene that enhances adaptive immune responses, was significantly downregulated in IBV-inoculated FNECs. Significant differences in other proinflammatory and adaptive genes were suppressed and delayed following IBV inoculation. Following IBV infection, ex vivo cell cultures derived from the ferret upper respiratory tract exhibited reduced and delayed innate responses which may contribute to reduced antibody responses in vivo.IMPORTANCEInfluenza B viruses (IBV) represent nearly one-quarter of all human influenza cases and are responsible for significant clinical and socioeconomic impacts but do not pose the same pandemic risks as influenza A viruses (IAV) and have thus received much less attention. IBV accounts for greater severity and deaths in children, and vaccine efficacy remains low. The ferret can be readily infected with human clinical isolates and demonstrates a similar course of disease and immune responses. IBV, however, generates lower antibodies in ferrets than IAV following the challenge. To determine whether differences in initial innate responses following infection may affect the development of robust adaptive immune responses, ferret respiratory tract cells were isolated, infected with IAV/IBV, and compared. Understanding the differences in the initial innate immune responses to IAV and IBV may be important in the development of more effective vaccines and interventions to generate more robust protective immune responses.


Asunto(s)
Inmunidad Adaptativa , Células Epiteliales , Hurones , Inmunidad Innata , Virus de la Influenza A , Virus de la Influenza B , Interferones , Mucosa Nasal , Animales , Niño , Humanos , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/virología , Hurones/inmunología , Hurones/virología , Virus de la Influenza A/clasificación , Virus de la Influenza A/crecimiento & desarrollo , Virus de la Influenza A/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Virus de la Influenza B/clasificación , Virus de la Influenza B/crecimiento & desarrollo , Virus de la Influenza B/inmunología , Vacunas contra la Influenza , Gripe Humana/virología , Interferones/inmunología , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Linfopoyetina del Estroma Tímico/genética , Linfopoyetina del Estroma Tímico/inmunología , Células Cultivadas
10.
Int Forum Allergy Rhinol ; 14(7): 1147-1162, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38197521

RESUMEN

BACKGROUND: The role of Akt in nasal immunity is unstudied. Akt phosphorylates and activates endothelial nitric oxide synthase (eNOS) expressed in epithelial ciliated cells. Nitric oxide (NO) production by ciliated cells can have antibacterial and antiviral effects. Increasing nasal NO may be a useful antipathogen strategy in chronic rhinosinusitis (CRS). We previously showed that small-molecule Akt activator SC79 induces nasal cell NO production and suppresses IL-8 via the transcription factor Nrf-2. We hypothesized that SC79 NO production may additionally have antibacterial effects. METHODS: NO production was measured using fluorescent dye DAF-FM. We tested effects of SC79 during co-culture of Pseudomonas aeruginosa with primary nasal epithelial cells, using CFU counting and live-dead staining to quantify bacterial killing. Pharmacology determined the mechanism of SC79-induced NO production and tested dependence on Akt. RESULTS: SC79 induced dose-dependent, Akt-dependent NO production in nasal epithelial cells. The NO production required eNOS and Akt. The NO released into the airway surface liquid killed P. aeruginosa. No toxicity (LDH release) or inflammatory effects (IL8 transcription) were observed over 24 h. CONCLUSIONS: Together, these data suggest multiple immune pathways are stimulated by SC79, with antipathogen effects. This in vitro pilot study suggests that a small-molecule Akt activator may have clinical utility in CRS or respiratory other infection settings, warranting future in vivo studies.


Asunto(s)
Células Epiteliales , Mucosa Nasal , Óxido Nítrico , Proteínas Proto-Oncogénicas c-akt , Pseudomonas aeruginosa , Humanos , Óxido Nítrico/metabolismo , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Mucosa Nasal/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Células Cultivadas , Óxido Nítrico Sintasa de Tipo III/metabolismo , Antibacterianos/farmacología , Rinitis/inmunología , Rinitis/microbiología , Rinitis/tratamiento farmacológico
11.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055020

RESUMEN

The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.


Asunto(s)
COVID-19/virología , Mucosa Nasal/citología , Mucosa Nasal/virología , Técnicas de Cultivo de Tejidos/métodos , Adolescente , Adulto , Enzima Convertidora de Angiotensina 2/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , SARS-CoV-2 , Internalización del Virus
12.
Anat Rec (Hoboken) ; 305(8): 1871-1891, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34545690

RESUMEN

Our knowledge of nasal cavity anatomy has grown considerably with the advent of micro-computed tomography (CT). More recently, a technique called diffusible iodine-based contrast-enhanced CT (diceCT) has rendered it possible to study nasal soft tissues. Using diceCT and histology, we aim to (a) explore the utility of these techniques for inferring the presence of venous sinuses that typify respiratory mucosa and (b) inquire whether distribution of vascular mucosa may relate to specialization for derived functions of the nasal cavity (i.e., nasal-emission of echolocation sounds) in bats. Matching histology and diceCT data indicate that diceCT can detect venous sinuses as either darkened, "empty" spaces, or radio-opaque islands when blood cells are present. Thus, we show that diceCT provides reliable information on vascular distribution in the mucosa of the nasal airways. Among the bats studied, a nonecholocating pteropodid (Cynopterus sphinx) and an oral-emitter of echolocation sounds (Eptesicus fuscus) possess venous sinus networks that drain into the sphenopalatine vein rostral to the nasopharynx. In contrast, nasopharyngeal passageways of nasal-emitting hipposiderids are notably packed with venous sinuses. The mucosae of the nasopharyngeal passageways are far less vascular in nasal-emitting phyllostomids, in which vascular mucosae are more widely distributed in the nasal cavity, and in some nectar-feeding species, a particularly large venous sinus is adjacent to the vomeronasal organ. Therefore, we do not find a common pattern of venous sinus distribution associated with nasal emission of sounds in phyllostomids and hipposiderids. Instead, vascular mucosa is more likely critical for air-conditioning and sometimes vomeronasal function in all bats.


Asunto(s)
Quirópteros , Cavidad Nasal , Mucosa Nasal , Venas , Microtomografía por Rayos X , Animales , Quirópteros/anatomía & histología , Quirópteros/fisiología , Ecolocación/fisiología , Cavidad Nasal/anatomía & histología , Cavidad Nasal/irrigación sanguínea , Cavidad Nasal/citología , Cavidad Nasal/diagnóstico por imagen , Mucosa Nasal/anatomía & histología , Mucosa Nasal/irrigación sanguínea , Mucosa Nasal/citología , Mucosa Nasal/diagnóstico por imagen , Venas/anatomía & histología , Venas/citología , Venas/diagnóstico por imagen
13.
Nat Commun ; 12(1): 7092, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876592

RESUMEN

The nasal epithelium is a plausible entry point for SARS-CoV-2, a site of pathogenesis and transmission, and may initiate the host response to SARS-CoV-2. Antiviral interferon (IFN) responses are critical to outcome of SARS-CoV-2. Yet little is known about the interaction between SARS-CoV-2 and innate immunity in this tissue. Here we apply single-cell RNA sequencing and proteomics to a primary cell model of human nasal epithelium differentiated at air-liquid interface. SARS-CoV-2 demonstrates widespread tropism for nasal epithelial cell types. The host response is dominated by type I and III IFNs and interferon-stimulated gene products. This response is notably delayed in onset relative to viral gene expression and compared to other respiratory viruses. Nevertheless, once established, the paracrine IFN response begins to impact on SARS-CoV-2 replication. When provided prior to infection, recombinant IFNß or IFNλ1 induces an efficient antiviral state that potently restricts SARS-CoV-2 viral replication, preserving epithelial barrier integrity. These data imply that the IFN-I/III response to SARS-CoV-2 initiates in the nasal airway and suggest nasal delivery of recombinant IFNs to be a potential chemoprophylactic strategy.


Asunto(s)
Células Epiteliales/virología , Interferón Tipo I/inmunología , Interferones/inmunología , Mucosa Nasal/virología , SARS-CoV-2/fisiología , Antivirales/inmunología , Antivirales/farmacología , COVID-19/inmunología , COVID-19/virología , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/inmunología , Humanos , Inmunidad Innata , Cinética , Mucosa Nasal/citología , Mucosa Nasal/inmunología , SARS-CoV-2/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tropismo Viral , Replicación Viral/efectos de los fármacos , Interferón lambda
14.
Physiol Rep ; 9(20): e15075, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34676696

RESUMEN

Exercise has substantial health benefits, but the effects of exercise on immune status and susceptibility to respiratory infections are less clear. Furthermore, there is limited research examining the effects of prolonged exercise on local respiratory immunity and antiviral activity. To assess the upper respiratory tract in response to exercise, we collected nasal lavage fluid (NALF) from human subjects (1) at rest, (2) after 45 min of moderate-intensity exercise, and (3) after 180 min of moderate-intensity exercise. To assess immune responses of the lower respiratory tract, we utilized a murine model to examine the effect of exercise duration on bronchoalveolar lavage (BAL) fluid immune cell content and lung gene expression. NALF cell counts did not change after 45 min of exercise, whereas 180 min significantly increased total cells and leukocytes in NALF. Importantly, fold change in NALF leukocytes correlated with the post-exercise fatigue rating in the 180-min exercise condition. The acellular portion of NALF contained strong antiviral activity against Influenza A in both resting and exercise paradigms. In mice undergoing moderate-intensity exercise, BAL total cells and neutrophils decreased in response to 45 or 90 min of exercise. In lung lobes, increased expression of heat shock proteins suggested that cellular stress occurred in response to exercise. However, a broad upregulation of inflammatory genes was not observed, even at 180 min of exercise. This work demonstrates that exercise duration differentially alters the cellularity of respiratory tract fluids, antiviral activity, and gene expression. These changes in local mucosal immunity may influence resistance to respiratory viruses, including influenza or possibly other pathogens in which nasal mucosa plays a protective role, such as rhinovirus or SARS-CoV-2.


Asunto(s)
Ejercicio Físico/fisiología , Virus de la Influenza A/inmunología , Leucocitos/inmunología , Pulmón/inmunología , Líquido del Lavado Nasal/inmunología , Neutrófilos/inmunología , Adolescente , Adulto , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Expresión Génica , Humanos , Leucocitos/metabolismo , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Lavado Nasal (Proceso)/métodos , Líquido del Lavado Nasal/citología , Mucosa Nasal/citología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Neutrófilos/metabolismo , Factores de Tiempo , Adulto Joven
15.
Bioengineered ; 12(1): 8583-8593, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607526

RESUMEN

Interleukin (IL)-13-associated inflammatory response is important for the pathogenesis of allergic rhinitis (AR). Apremilast is a phosphodiesterase-4 (PDE4) inhibitor approved for psoriasis treatment. Here, we investigated the potential effects of Apremilast against IL-13-induced injury in human nasal epithelial cells (hNECs). Firstly, Apremilast ameliorated oxidative stress in IL-13-challenged cells by decreasing the levels of reactive oxygen species (ROS) and the production of malondialdehyde (MDA). Secondly, Apremilast inhibited the expressions of IL-6 and IL-8. Moreover, Apremilast inhibited the expressions of the chemokines colony-stimulating factor 2 (CSF2) and chemokine ligand 11 (CCL11). Interestingly, exposure to IL-13 increased the expressions of mucin 4 and mucin 5AC (MUC5AC), which was ameliorated by treatment with Apremilast. Interestingly, we found that Apremilast inhibited the phosphorylation of c-Jun-N-terminal kinase (JNK). Importantly, Apremilast reduced the levels of c-fos and c-Jun, the two AP-1 subfamilies. The luciferase reporter assay demonstrates that Apremilast reduced the transcriptional activity of activator protein 1 (AP-1). Lastly, we found that Apremilast prevented the activation of nuclear factor kappa-B (NF-κB) by decreasing the levels of nuclear NF-κB p65 and the luciferase activity of the NF-κB reporter. In summary, we conclude that Apremilast possesses a protective effect against IL-13-induced inflammatory response and mucin production in hNECs by inhibiting the activity of AP-1 and NF-κB.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Interleucina-13/metabolismo , Mucinas/metabolismo , Mucosa Nasal/citología , Talidomida/análogos & derivados , Células Cultivadas , Humanos , Inflamación/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Talidomida/farmacología
16.
Food Chem Toxicol ; 157: 112606, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34653555

RESUMEN

Evaluating the safety of previously fabricated and effective green synthetized colloidal silver (GSCS) on the mucosal barrier structure and function is essential prior to conduct human trials. The GSCS was applied to primary human nasal epithelial cells (HNECs) grown in an air-liquid interface (ALI) culture. Epithelial barrier integrity was evaluated by measuring the transepithelial electrical resistance (TEER) and fluorescein isothiocyanate (FITC)-dextran paracellular permeability. Ciliary beat frequency (CBF) was quantified. Effects of the GSCS on cell viability and inflammation were examined through lactate dehydrogenase, the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide viability assay and interleukin 6 (IL-6) enzyme linked immunosorbent assay. The localization and transportation of GSCS within HNECs and their HNEC-ALI cultures was assessed by transmission electron microscopy and inductively coupled plasma-mass-spectrometry, respectively. Application of GSCS to HNECs-ALI cultures for up to 2 h caused a significant reduction in the TEER values, however, it did not drop within the first 10 and 20 min for CRS and non-CRS control HNECs. The paracellular permeability, cell viability, IL-6 secretion and CBF remained unchanged. No GSCS was observed within or transported across HNECs. In conclusion, application of GSCS to HNECs is devoid of toxic effects.


Asunto(s)
Nanopartículas del Metal/toxicidad , Mucosa Nasal/efectos de los fármacos , Plata/toxicidad , Permeabilidad de la Membrana Celular , Células Cultivadas , Cilios/efectos de los fármacos , Dextranos/farmacocinética , Impedancia Eléctrica , Ensayo de Inmunoadsorción Enzimática/métodos , Fluoresceína-5-Isotiocianato/análogos & derivados , Fluoresceína-5-Isotiocianato/farmacocinética , Tecnología Química Verde/métodos , Humanos , Mucosa Nasal/citología , Plata/química
17.
ACS Appl Mater Interfaces ; 13(41): 48365-48377, 2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34633177

RESUMEN

It is critical to obtain an anti-inflammatory microenvironment when curing spinal cord injury (SCI). On the basis of this, we prepared Lycium barbarum oligosaccharide (LBO)-nasal mucosa-derived mesenchymal stem cells (EMSCs) fibronectin hydrogel for SCI restoration via inflammatory license effect and M2 polarization of microglias. LBO exhibited remarkable M2 polarization potential for microglia. However, EMSCs primed by LBO generated enhanced paracrine effects through the inflammatory license-like process. The observed dual function is likely based on the TNFR2 pathway. In addition, LBO-EMSC hydrogel possesses a synergistic effect on M2 polarization of microglia through the PI3K-Akt-mTOR signaling pathway. The obtained findings provide a simple approach for MSC-based therapies for SCI and shed more light on the role of TNFR2 on bidirectional regulation in tissue regeneration.


Asunto(s)
Fibrina/farmacología , Hidrogeles/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Polisacáridos/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Andamios del Tejido/química , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Línea Celular , Fibrina/química , Humanos , Hidrogeles/química , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lycium/química , Masculino , Células Madre Mesenquimatosas/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Mucosa Nasal/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Polisacáridos/química , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Recuperación de la Función/efectos de los fármacos , Remielinización/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
18.
STAR Protoc ; 2(4): 100782, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34585152

RESUMEN

This protocol is intended as a guide for implementing or refining the usage of the air-liquid interface (ALI) model system to generate airway mucociliary tissue in vitro. We present a streamlined protocol for isolating the stem cells from inferior nasal turbinates of donors, allowing for a simple and low-cost supply of primary cells for research. We also provide our detailed protocols for ALI tissue processing and immunofluorescence to aid in the standardization of these techniques between research groups. For complete details on the use and execution of this protocol, please refer to Hussain et al., (2014)Yang et al., (2016)Im et al., (2019).


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Epiteliales , Técnicas de Preparación Histocitológica/métodos , Mucosa Nasal/citología , Adulto , Diferenciación Celular , Células Cultivadas , Células Epiteliales/química , Células Epiteliales/citología , Humanos , Masculino
19.
Bioengineered ; 12(1): 6045-6056, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34482800

RESUMEN

Allergic rhinitis (AR) is a symptomatic allergic disease that leads to severe inflammation. Astragaloside IV (AS-IV) is a primary active component of Astragalus membranaceus and exerts immune-regulation and anti-inflammatory effects. However, the pharmacological effect of AS-IV in the nasal epithelial cells (NECs) has not been reported. The present study aimed to assess the effect of AS-IV on inflammatory cytokines and mucin 5 subtype AC (MUC5AC) overproduction in histamine (His)-stimulated NECs and its underlying mechanism. NECs were stimulated with or without His for 24 h in the absence or presence of AS-IV. The levels of inflammatory cytokines including IL-6, IL-8, MCP-1, IL-1ß, granulocyte-macrophage colony-stimulating factor (GM-CSF), eotaxin, and MUC5AC were assayed. Our findings indicated that AS-IV inhibited His-evoked release and expression of inflammatory cytokines and MUC5AC in NECs. RNA-seq analyses indicated the significant changes in expression levels involved in inflammation genes upon treatment of His-induced NECs with AS-IV. Our findings indicated that AS-IV inhibited His-evoked inflammatory cytokines secretion and MUC5AC overproduction in NECs, which were partly mediated by regulation of inflammation-related genes. Therefore, our findings provided a scientific basis for the development of AS-IV as an effective agent for clinical therapeutic strategy in the treatment of AR.


Asunto(s)
Antagonistas de los Receptores Histamínicos/farmacología , Inflamación/genética , Mucina 5AC/metabolismo , Saponinas/farmacología , Triterpenos/farmacología , Células Cultivadas , Citocinas/metabolismo , Células Epiteliales , Histamina/metabolismo , Humanos , Inflamación/metabolismo , Mucosa Nasal/citología , Rinitis Alérgica
20.
Sci Rep ; 11(1): 15927, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34354210

RESUMEN

Previous studies focusing on the age disparity in COVID-19 severity have suggested that younger individuals mount a more robust innate immune response in the nasal mucosa after infection with SARS-CoV-2. However, it is unclear if this reflects increased immune activation or increased immune residence in the nasal mucosa. We hypothesized that immune residency in the nasal mucosa of healthy individuals may differ across the age range. We applied single-cell RNA-sequencing and measured the cellular composition and transcriptional profile of the nasal mucosa in 35 SARS-CoV-2 negative children and adults, ranging in age from 4 months to 65 years. We analyzed in total of ~ 30,000 immune and epithelial cells and found that age and immune cell proportion in the nasal mucosa are inversely correlated, with little evidence for structural changes in the transcriptional state of a given cell type across the age range. Orthogonal validation by epigenome sequencing indicate that it is especially cells of the innate immune system that underlie the age-association. Additionally, we characterize the predominate immune cell type in the nasal mucosa: a resident T cell like population with potent antiviral properties. These results demonstrate fundamental changes in the immune cell makeup of the uninfected nasal mucosa over the lifespan. The resource we generate here is an asset for future studies focusing on respiratory infection and immunization strategies.


Asunto(s)
COVID-19/inmunología , Mucosa Nasal/inmunología , SARS-CoV-2/inmunología , Adolescente , Adulto , COVID-19/genética , Niño , Preescolar , Femenino , Humanos , Inmunidad Celular , Inmunidad Innata , Lactante , Masculino , Persona de Mediana Edad , Mucosa Nasal/citología , Mucosa Nasal/metabolismo , Índice de Severidad de la Enfermedad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transcriptoma , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA