Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.773
Filtrar
1.
Nano Lett ; 24(33): 10380-10387, 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39120059

RESUMEN

The advancement of effective nasal mucoadhesive delivery faces challenges due to rapid mucociliary clearance (MCC). Conventional studies have employed mucoadhesive materials, mainly forming spherical nanoparticles, but these offer limited adhesion to the nasal mucosa. This study hypothesizes that a 2D nanoscale structure utilizing adhesive polyphenols can provide a superior strategy for countering MCC, aligning with the planar mucosal layers. We explore the use of tannic acid (TA), a polyphenolic molecule known for its adhesive properties and ability to form complexes with biomolecules. Our study introduces an unprecedented 2D nanopatch, assembled through the interaction of TA with green fluorescent protein (GFP), and cell-penetrating peptide (CPP). This 2D nanopatch demonstrates robust adhesion to nasal mucosa and significantly enhances immunoglobulin A secretions, suggesting its potential for enhancing nasal vaccine delivery. The promise of a polyphenol-enabled adhesive 2D nanopatch signifies a pivotal shift from conventional spherical nanoparticles, opening new pathways for delivery strategies through respiratory mucoadhesion.


Asunto(s)
Mucosa Nasal , Polifenoles , Taninos , Taninos/química , Polifenoles/química , Polifenoles/administración & dosificación , Mucosa Nasal/metabolismo , Mucosa Nasal/inmunología , Animales , Nanopartículas/química , Humanos , Péptidos de Penetración Celular/química , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Fluorescentes Verdes/química , Adhesivos/química , Depuración Mucociliar/efectos de los fármacos , Inmunoglobulina A , Ratones
2.
J Exp Med ; 221(9)2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-38949639

RESUMEN

Children resist COVID-19, and previous studies reported increased innate immunity in their upper airways. A new paper by Watkins et al. (https://doi.org/10.1084/jem.20230911) shows that the nasal mucosa of children is characterized by often asymptomatic viral and/or bacterial infections that dynamically regulate distinct innate immune programs.


Asunto(s)
COVID-19 , Inmunidad Innata , Mucosa Nasal , SARS-CoV-2 , Humanos , COVID-19/inmunología , Niño , Inmunidad Innata/inmunología , SARS-CoV-2/inmunología , Mucosa Nasal/virología , Mucosa Nasal/inmunología
3.
J Exp Med ; 221(9)2024 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-38949638

RESUMEN

Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1ß and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.


Asunto(s)
COVID-19 , Inmunidad Innata , SARS-CoV-2 , Humanos , Inmunidad Innata/inmunología , Preescolar , Lactante , COVID-19/inmunología , COVID-19/virología , Niño , SARS-CoV-2/inmunología , Femenino , Masculino , Nasofaringe/inmunología , Nasofaringe/virología , Nasofaringe/microbiología , Carga Viral , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Mucosa Nasal/microbiología , Citocinas/metabolismo , Citocinas/inmunología , Interacciones Huésped-Patógeno/inmunología , Adolescente , Nariz/inmunología , Nariz/virología , Nariz/microbiología , Coinfección/inmunología , Coinfección/virología
4.
Artículo en Chino | MEDLINE | ID: mdl-38965843

RESUMEN

Objective: To analyze the cellular composition characteristics of the nasal tissue immune microenvironment in patients with control, chronic rhinosinusitis without nasal polyps (CRSsNP), non-eosinophilic chronic rhinosinusitis with nasal polyps (neCRSwNP), and eosinophilic chronic rhinosinusitis with nasal polyps (eCRSwNP) using mass cytometry flow technology. Methods: Thirteen CRS patients who underwent endoscopic nasal surgery at the Department of Otorhinolaryngology Head and Neck Surgery of Peking Union Medical College Hospital from March to December 2022 were recruited, including 8 males and 5 females, aged 22.3 to 58.3 years. Three control mucosae were obtained from normal ethmoid or sphenoid sinuses of patients with benign tumors of the temporal fossa or non-functional pituitary adenomas who underwent endoscopic surgery, excluding allergic rhinitis and sinusitis. Sixteen clinical tissue samples (3 of control, 3 of CRSsNP, 4 of neCRSwNP, and 6 of eCRSwNP) were prepared into single-cell suspensions. Mass cytometry flow detection was performed using a combination of 42 molecular markers to analyze the differences in cell subpopulations among the groups. Data were analyzed using GraphPad Prism 9. Results: Based on the mass cytometry flow results, cells from control, CRSsNP, neCRSwNP, and eCRSwNP were divided into seven main cell subgroups, with detailed subgrouping of T/NK cells and myeloid cells. In T/NK cells, compared with the control group, the number of NK CD56bright cells increased in the CRSsNP group, while NK CD56dim cells decreased; compared with the CRSsNP group, the eCRSwNP group showed a decrease in NKT cells and CD4+Tem cells; compared with the CRSsNP group, the eCRSwNP group showed a significant increase in CD25 expression within Treg cells; compared with the CRSsNP group, the eCRSwNP group showed a significant decrease in Tbet expression in CD8+Teff cells and CD8+TRM cells; in eCRSwNP, the expression of CD103 in CD8+TRM cells was significantly lower than in CRSsNP. In myeloid cells, compared with the other three groups, the eCRSwNP group showed a significant increase in macrophages and a significant decrease in cDC1 and monocytes; compared with the control group and CRSsNP, the eCRSwNP group also showed a significant decrease in resting state macrophages; compared with the CRSsNP group, the eCRSwNP group showed a significant decrease in the level of CX3CR1 within cDC2 and monocytes; the expression levels of NLRP3 in cDC2 and macrophages in the eCRSwNP group were significantly higher than in the other three groups; compared with the control group, the expression levels of Gata3 in cDC2 and macrophages in the eCRSwNP group were also significantly increased; additionally, the expression of CCR2 within monocytes in the eCRSwNP group was lower than in the CRSsNP group. In ILC, compared with the control group, the expression of CCR6 decreased in the eCRSwNP group. Conclusions: Compared with the control group, CRSsNP, and neCRSwNP, eCRSwNP shows an increase in macrophage number, a decrease in cDC1 and resting state macrophages, and depletion of protective cells CD103+CD8+TRM. Additionally, the expression levels of CCR2 and CX3CR1 in monocytes of eCRSwNP are decreased.


Asunto(s)
Pólipos Nasales , Rinosinusitis , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Microambiente Celular , Enfermedad Crónica , Eosinófilos/metabolismo , Citometría de Flujo , Espectrometría de Masas , Mucosa Nasal/metabolismo , Mucosa Nasal/inmunología , Pólipos Nasales/inmunología , Pólipos Nasales/metabolismo , Rinosinusitis/inmunología , Rinosinusitis/metabolismo
6.
Pathol Res Pract ; 260: 155468, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39018929

RESUMEN

BACKGROUND: Sinonasal mucosal melanoma (SNMM) is a rare but aggressive tumor with a poor prognosis. The co-inhibitory receptors T cell immunoglobulin and mucinodomain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3) and T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) are promising new targets in anti-cancer immunotherapy. The expression profiles of these immune checkpoint molecules (ICMs) and potential prognostic implications have not been characterized in SNMM yet. METHODS: Immunohistochemical staining for TIGIT, LAG-3 and TIM-3 was performed on tumor tissue samples from 27 patients with primary SNMM. Associations between ICM expression and demographic parameters, AJCC tumor stage, overall survival, and recurrence-free survival were retrospectively analyzed. RESULTS: SNMM patients with low numbers of TIGIT+ and TIM-3+ tumor infiltrating lymphocytes (TILs) in the primary tumor survived significantly longer than patients with a high degree of TIGIT+ and TIM-3+ TILs. High infiltration with TIM-3+ or TIGIT+ lymphocytes was associated with the higher T4 stage and decreased 5-year survival. CONCLUSION: We identified high densities of TIM-3+ and TIGIT+ TILs as strong negative prognostic biomarkers in SNMM. This suggests that TIM-3 and TIGIT contribute to immunosuppression in SNMM and provides a rationale for novel treatment strategies based on this next generation of immune checkpoint inhibitors. Prospective studies with larger case numbers are warranted to confirm our findings and their implications for immunotherapy.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A , Linfocitos Infiltrantes de Tumor , Melanoma , Receptores Inmunológicos , Humanos , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Masculino , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/análisis , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Femenino , Persona de Mediana Edad , Melanoma/patología , Melanoma/inmunología , Melanoma/mortalidad , Melanoma/metabolismo , Anciano , Pronóstico , Adulto , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Estudios Retrospectivos , Anciano de 80 o más Años , Neoplasias de los Senos Paranasales/patología , Neoplasias de los Senos Paranasales/inmunología , Neoplasias de los Senos Paranasales/metabolismo , Neoplasias de los Senos Paranasales/mortalidad , Mucosa Nasal/patología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo
7.
Immun Inflamm Dis ; 12(7): e1337, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39023421

RESUMEN

OBJECTIVE: To investigate the effect of nasal mucosa-derived ectodermal mesenchymal stem cells (NM-EMSCs) on the inflammatory state of rats with chronic rhinosinusitis (CRS) and the underlying therapeutic mechanism. METHODS: NM-EMSCs were isolated and extracted to construct a rat model of CRS. Fifteen Sprague‒Dawley (SD) rats were randomly divided into three groups: CK + NS group rats were injected locally with saline in the nasal mucosa; CRS + NS group rats were injected locally with saline in the nasal mucosa; and CRS + EMSCs group rats were injected locally with NM-EMSCs in the nasal mucosa. One rat from the CRS + EMSCs group was randomly euthanized at 2, 4, and 6 days after injection, and the nasal mucosa tissues were collected for HE staining, Masson's trichrome staining, and periodic acid-Schiff staining. RESULTS: NM-EMSCs specifically expressing CD73, CD105, and CD90 were successfully isolated from the nasal mucosa of rats and were able to differentiate into adipocytes, osteoblasts, and chondrocytes. After saline and NM-EMSC injection, compared with those in the blank control CK + NS group, the nasal mucosa in the CRS + NS and CRS + EMSC groups exhibited obvious thickening, a large amount of inflammatory cell infiltration, and increased collagen and mucin distribution. Four days post-NM-EMSC injection, the thickening of the nasal mucosa in the CRS group was gradually alleviated, the inflammatory cell infiltration gradually decreased, and the distribution of collagen and mucin and the collagen-positive area gradually decreased. Moreover, only a small number of inflammatory cells were visible, and the distribution of mucins was limited to 6 days post-NM-EMSC injection. CONCLUSION: NM-EMSCs effectively attenuated inflammation in the nasal mucosa of CRS model rats.


Asunto(s)
Diferenciación Celular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Mucosa Nasal , Ratas Sprague-Dawley , Rinitis , Sinusitis , Animales , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Mucosa Nasal/inmunología , Sinusitis/terapia , Sinusitis/inmunología , Sinusitis/patología , Ratas , Células Madre Mesenquimatosas/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Enfermedad Crónica , Rinitis/terapia , Rinitis/inmunología , Rinitis/patología , Modelos Animales de Enfermedad , Células Cultivadas , Masculino , Rinosinusitis
8.
Front Immunol ; 15: 1429442, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39040099

RESUMEN

Introduction: Allergic rhinitis (AR) is an upper airway inflammatory disease of the nasal mucosa. Conventional treatments such as symptomatic pharmacotherapy and allergen-specific immunotherapy have considerable limitations and drawbacks. As an emerging therapy with regenerative potential and immunomodulatory effect, mesenchymal stem cell-derived exosomes (MSC-Exos) have recently been trialed for the treatment of various inflammatory and autoimmune diseases. Methods: In order to achieve sustained and protected release of MSC-Exos for intranasal administration, we fabricated Poly(lactic-co-glycolic acid) (PLGA) micro and nanoparticles-encapsulated MSC-Exos (PLGA-Exos) using mechanical double emulsion for local treatment of AR. Preclinical in vivo imaging, ELISA, qPCR, flow cytometry, immunohistochemical staining, and multiomics sequencing were used for phenotypic and mechanistic evaluation of the therapeutic effect of PLGA-Exos in vitro and in vivo. Results: The results showed that our PLGA platform could efficiently encapsulate and release the exosomes in a sustained manner. At protein level, PLGA-Exos treatment upregulated IL-2, IL-10 and IFN-γ, and downregulated IL-4, IL-17 and antigen-specific IgE in ovalbumin (OVA)-induced AR mice. At cellular level, exosomes treatment reduced Th2 cells, increased Tregs, and reestablished Th1/Th2 balance. At tissue level, PLGA-Exos significantly attenuated the infiltration of immune cells (e.g., eosinophils and goblet cells) in nasal mucosa. Finally, multiomics analysis discovered several signaling cascades, e.g., peroxisome proliferator-activated receptor (PPAR) pathway and glycolysis pathway, that might mechanistically support the immunomodulatory effect of PLGA-Exos. Discussion: For the first time, we present a biomaterial-facilitated local delivery system for stem cell-derived exosomes as a novel and promising strategy for AR treatment.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Rinitis Alérgica , Exosomas/inmunología , Exosomas/metabolismo , Animales , Rinitis Alérgica/terapia , Rinitis Alérgica/inmunología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Inmunomodulación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Administración Intranasal
9.
Signal Transduct Target Ther ; 9(1): 190, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39039046

RESUMEN

The upper respiratory tract is the initial site of SARS-CoV-2 infection. Nasal spike-specific secretory immunoglobulin A (sIgA) correlates with protection against Omicron breakthrough infection. We report that intranasal vaccination using human adenovirus serotype 5 (Ad5) vectored Omicron spike in people who previously vaccinated with ancestral vaccine could induce robust neutralizing sIgA in the nasal passage. Nasal sIgA was predominantly present in dimeric and multimeric forms and accounted for nearly 40% of total proteins in nasal mucosal lining fluids (NMLFs). A low-level IgG could also be detected in NMLFs but not IgM, IgD, and IgE. After a complete nasal wash, sIgA in the nasal passage could be replenished rapidly within a few hours. A comparison of purified paired serum IgA, serum IgG, and nasal sIgA from the same individuals showed that sIgA was up to 3-logs more potent than serum antibodies in binding to spikes and in neutralizing Omicron subvariants. Serum IgG and IgA failed to neutralize XBB and BA.2.86, while nasal sIgA retained potent neutralization against these newly emerged variants. Further analysis showed that sIgA was more effective than IgG or IgA in blocking spike-mediated cell-to-cell transmission and protecting hACE2 mice from XBB challenge. Using a sIgA monoclonal antibody as a reference, we estimated that the total nasal sIgA contains about 2.6-3.9% spike-specific sIgA in NMLFs collected approximately one month after intranasal vaccination. Our study provided insights for developing intranasal vaccines that can induce sIgA to build an effective and mutation-resistant first-line immune barrier against constantly emerging variants.


Asunto(s)
Administración Intranasal , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacunas contra la COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Humanos , SARS-CoV-2/inmunología , SARS-CoV-2/genética , COVID-19/prevención & control , COVID-19/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Animales , Ratones , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/administración & dosificación , Inmunoglobulina A/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/genética , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Femenino , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Inmunoglobulina A Secretora/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Masculino
10.
J Infect Dis ; 230(1): e17-e29, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052740

RESUMEN

BACKGROUND: While inflammatory and immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in peripheral blood are extensively described, responses at the upper respiratory mucosal site of initial infection are relatively poorly defined. We sought to identify mucosal cytokine/chemokine signatures that distinguished coronavirus disease 2019 (COVID-19) severity categories, and relate these to disease progression and peripheral inflammation. METHODS: We measured 35 cytokines and chemokines in nasal samples from 274 patients hospitalized with COVID-19. Analysis considered the timing of sampling during disease, as either the early (0-5 days after symptom onset) or late (6-20 days after symptom onset) phase. RESULTS: Patients that survived severe COVID-19 showed interferon (IFN)-dominated mucosal immune responses (IFN-γ, CXCL10, and CXCL13) early in infection. These early mucosal responses were absent in patients who would progress to fatal disease despite equivalent SARS-CoV-2 viral load. Mucosal inflammation in later disease was dominated by interleukin 2 (IL-2), IL-10, IFN-γ, and IL-12p70, which scaled with severity but did not differentiate patients who would survive or succumb to disease. Cytokines and chemokines in the mucosa showed distinctions from responses evident in the peripheral blood, particularly during fatal disease. CONCLUSIONS: Defective early mucosal antiviral responses anticipate fatal COVID-19 but are not associated with viral load. Early mucosal immune responses may define the trajectory of severe COVID-19.


Asunto(s)
COVID-19 , Citocinas , Inflamación , SARS-CoV-2 , Humanos , COVID-19/inmunología , COVID-19/mortalidad , Masculino , Femenino , Persona de Mediana Edad , SARS-CoV-2/inmunología , Citocinas/sangre , Anciano , Inflamación/inmunología , Inmunidad Mucosa , Carga Viral , Adulto , Quimiocinas/sangre , Índice de Severidad de la Enfermedad , Mucosa Nasal/inmunología , Mucosa Nasal/virología
11.
Front Immunol ; 15: 1409458, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39015572

RESUMEN

Current treatments of eosinophilic chronic rhinosinusitis (ECRS) involve corticosteroids with various adverse effects and costly therapies such as dupilumab, highlighting the need for improved treatments. However, because of the lack of a proper mouse ECRS model that recapitulates human ECRS, molecular mechanisms underlying this disease are incompletely understood. ECRS is often associated with aspirin-induced asthma, suggesting that dysregulation of lipid mediators in the nasal mucosa may underlie ECRS pathology. We herein found that the expression of microsomal PGE synthase-1 (encoded by PTGES) was significantly lower in the nasal mucosa of ECRS patients than that of non-ECRS subjects. Histological, transcriptional, and lipidomics analyses of Ptges-deficient mice revealed that defective PGE2 biosynthesis facilitated eosinophil recruitment into the nasal mucosa, elevated expression of type-2 cytokines and chemokines, and increased pro-allergic and decreased anti-allergic lipid mediators following challenges with Aspergillus protease and ovalbumin. A nasal spray containing agonists for the PGE2 receptor EP2 or EP4, including omidenepag isopropyl that has been clinically used for treatment of glaucoma, markedly reduced intranasal eosinophil infiltration in Ptges-deficient mice. These results suggest that the present model using Ptges-deficient mice is more relevant to human ECRS than are previously reported models and that eosinophilic inflammation in the nasal mucosa can be efficiently blocked by activation of the PGE2-EP2 pathway. Furthermore, our findings suggest that drug repositioning of omidenepag isopropyl may be useful for treatment of patients with ECRS.


Asunto(s)
Dinoprostona , Eosinofilia , Ratones Noqueados , Mucosa Nasal , Subtipo EP2 de Receptores de Prostaglandina E , Rinitis , Sinusitis , Animales , Sinusitis/tratamiento farmacológico , Sinusitis/metabolismo , Sinusitis/inmunología , Humanos , Ratones , Rinitis/tratamiento farmacológico , Rinitis/metabolismo , Rinitis/inmunología , Dinoprostona/metabolismo , Mucosa Nasal/metabolismo , Mucosa Nasal/inmunología , Mucosa Nasal/efectos de los fármacos , Eosinofilia/tratamiento farmacológico , Eosinofilia/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Modelos Animales de Enfermedad , Masculino , Transducción de Señal/efectos de los fármacos , Prostaglandina-E Sintasas/genética , Prostaglandina-E Sintasas/metabolismo , Eosinófilos/inmunología , Eosinófilos/metabolismo , Eosinófilos/efectos de los fármacos , Femenino , Enfermedad Crónica , Ratones Endogámicos C57BL , Rinosinusitis
12.
Front Immunol ; 15: 1344995, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011034

RESUMEN

Background: Allergic rhinitis (AR) is a complex disease in which gene-environment interactions contribute to its pathogenesis. Epigenetic modifications, such as N6-methyladenosine (m6A) modification of mRNA, play important roles in regulating gene expression in multiple physiological and pathological processes. However, the function of m6A modification in AR and the inflammatory response is poorly understood. Methods: We used the ovalbumin (OVA) and aluminum hydroxide to induce an AR mouse model. Nasal symptoms, histopathology, and serum cytokines were examined. We performed combined m6A and RNA sequencing to analyze changes in m6A modification profiles. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and methylated RNA immunoprecipitation sequencing qPCR (MeRIP-qPCR) were used to verify differential methylation of mRNAs and the m6A methylation level. Knockdown or inhibition of Alkbh5 in nasal mucosa of mice was mediated by lentiviral infection or IOX1 treatment. Results: We showed that m6A was enriched in a group of genes involved in MAPK signaling pathway. Moreover, we identified a MAPK pathway involving Map3k8, Erk2, and Nfκb1 that may play a role in the disrupted inflammatory response associated with nasal inflammation. The m6A eraser, Alkbh5, was highly expressed in the nasal mucosa of AR model mice. Furthermore, knockdown of Alkbh5 expression by lentiviral infection resulted in high MAPK pathway activity and a significant nasal mucosa inflammatory response. Our findings indicate that ALKBH5-mediated m6A dysregulation likely contributes to a nasal inflammatory response via the MAPK pathway. Conclusion: Together, our data show that m6A dysregulation mediated by ALKBH5, is likely to contribute to inflammation of the nasal mucosa via the MAPK signaling pathway, suggesting that ALKBH5 is a potential biomarker for AR treatment.


Asunto(s)
Adenosina , Desmetilasa de ARN, Homólogo 5 de AlkB , Modelos Animales de Enfermedad , Sistema de Señalización de MAP Quinasas , Mucosa Nasal , ARN Mensajero , Rinitis Alérgica , Animales , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Rinitis Alérgica/inmunología , Rinitis Alérgica/metabolismo , Rinitis Alérgica/genética , Ratones , Adenosina/análogos & derivados , Adenosina/metabolismo , Metilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Femenino , Ratones Endogámicos BALB C , Inflamación/genética , Inflamación/inmunología , Citocinas/metabolismo
13.
PLoS Pathog ; 20(7): e1012282, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38990812

RESUMEN

Haemophilus influenzae is a human respiratory pathogen and inhabits the human respiratory tract as its only niche. Despite this, the molecular mechanisms that allow H. influenzae to establish persistent infections of human epithelia are not well understood. Here, we have investigated how H. influenzae adapts to the host environment and triggers the host immune response using a human primary cell-based infection model that closely resembles human nasal epithelia (NHNE). Physiological assays combined with dualRNAseq revealed that NHNE from five healthy donors all responded to H. influenzae infection with an initial, 'unproductive' inflammatory response that included a strong hypoxia signature but did not produce pro-inflammatory cytokines. Subsequently, an apparent tolerance to large extracellular and intraepithelial burdens of H. influenzae developed, with NHNE transcriptional profiles resembling the pre-infection state. This occurred in parallel with the development of intraepithelial bacterial populations, and appears to involve interruption of NFκB signalling. This is the first time that large-scale, persistence-promoting immunomodulatory effects of H. influenzae during infection have been observed, and we were able to demonstrate that only infections with live, but not heat-killed H. influenzae led to immunomodulation and reduced expression of NFκB-controlled cytokines such as IL-1ß, IL-36γ and TNFα. Interestingly, NHNE were able to re-activate pro-inflammatory responses towards the end of the 14-day infection, resulting in release of IL-8 and TNFα. In addition to providing first molecular insights into mechanisms enabling persistence of H. influenzae in the host, our data further indicate the presence of infection stage-specific gene expression modules, highlighting fundamental similarities between immune responses in NHNE and canonical immune cells, which merit further investigation.


Asunto(s)
Células Epiteliales , Infecciones por Haemophilus , Haemophilus influenzae , Humanos , Haemophilus influenzae/inmunología , Infecciones por Haemophilus/inmunología , Infecciones por Haemophilus/microbiología , Células Epiteliales/microbiología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Mucosa Nasal/microbiología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Tolerancia Inmunológica , Células Cultivadas , Citocinas/metabolismo
14.
HNO ; 72(9): 633-638, 2024 Sep.
Artículo en Alemán | MEDLINE | ID: mdl-39031180

RESUMEN

Diagnosis of allergic disease is primarily verified by IgE blood serum analysis. Determination in nasal secretions is technically more difficult, particularly due to a low specimen volume and the method of sample collection. Nasal secretions are frequently collected by lavage, which allows qualitative diagnostics, whereas swabs with defined amounts of mucus enable quantitative analyses. In the case of negative skin and serum tests, detection of IgE in nasal mucus combined with nasal provocation testing aids differentiation between local allergic and nonallergic rhinitis.


Asunto(s)
Inmunoglobulina E , Mucosa Nasal , Humanos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo
15.
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
16.
Nature ; 632(8025): 630-636, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39085605

RESUMEN

The upper airway is an important site of infection, but immune memory in the human upper airway is poorly understood, with implications for COVID-19 and many other human diseases1-4. Here we demonstrate that nasal and nasopharyngeal swabs can be used to obtain insights into these challenging problems, and define distinct immune cell populations, including antigen-specific memory B cells and T cells, in two adjacent anatomical sites in the upper airway. Upper airway immune cell populations seemed stable over time in healthy adults undergoing monthly swabs for more than 1 year, and prominent tissue resident memory T (TRM) cell and B (BRM) cell populations were defined. Unexpectedly, germinal centre cells were identified consistently in many nasopharyngeal swabs. In subjects with SARS-CoV-2 breakthrough infections, local virus-specific BRM cells, plasma cells and germinal centre B cells were identified, with evidence of local priming and an enrichment of IgA+ memory B cells in upper airway compartments compared with blood. Local plasma cell populations were identified with transcriptional profiles of longevity. Local virus-specific memory CD4+ TRM cells and CD8+ TRM cells were identified, with diverse additional virus-specific T cells. Age-dependent upper airway immunological shifts were observed. These findings provide new understanding of immune memory at a principal mucosal barrier tissue in humans.


Asunto(s)
Memoria Inmunológica , Células B de Memoria , Células T de Memoria , Mucosa Nasal , Nasofaringe , SARS-CoV-2 , Adulto , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/citología , COVID-19/inmunología , COVID-19/virología , Centro Germinal/inmunología , Centro Germinal/citología , Inmunoglobulina A/inmunología , Memoria Inmunológica/inmunología , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Nasofaringe/virología , Nasofaringe/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/citología , SARS-CoV-2/inmunología
17.
Immunity ; 57(8): 1955-1974.e8, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-38964332

RESUMEN

The nasal mucosa is often the initial site of respiratory viral infection, replication, and transmission. Understanding how infection shapes tissue-scale primary and memory responses is critical for designing mucosal therapeutics and vaccines. We generated a single-cell RNA-sequencing atlas of the murine nasal mucosa, sampling three regions during primary influenza infection and rechallenge. Compositional analysis revealed restricted infection to the respiratory mucosa with stepwise changes in immune and epithelial cell subsets and states. We identified and characterized a rare subset of Krt13+ nasal immune-interacting floor epithelial (KNIIFE) cells, which concurrently increased with tissue-resident memory T (TRM)-like cells. Proportionality analysis, cell-cell communication inference, and microscopy underscored the CXCL16-CXCR6 axis between KNIIFE and TRM cells. Secondary influenza challenge induced accelerated and coordinated myeloid and lymphoid responses without epithelial proliferation. Together, this atlas serves as a reference for viral infection in the upper respiratory tract and highlights the efficacy of local coordinated memory responses.


Asunto(s)
Memoria Inmunológica , Células T de Memoria , Mucosa Nasal , Infecciones por Orthomyxoviridae , Animales , Memoria Inmunológica/inmunología , Ratones , Mucosa Nasal/virología , Mucosa Nasal/inmunología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Células T de Memoria/inmunología , Células Epiteliales/inmunología , Células Epiteliales/virología , Ratones Endogámicos C57BL , Humanos , Análisis de la Célula Individual , Gripe Humana/inmunología , Gripe Humana/virología , Femenino , Receptores CXCR6/metabolismo , Receptores CXCR6/inmunología , Virus de la Influenza A/inmunología , Virus de la Influenza A/fisiología
18.
Discov Med ; 36(185): 1260-1267, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38926112

RESUMEN

BACKGROUND: The incidence rate of allergic rhinitis (AR) is on the rise, which seriously affects the quality of life, work efficiency, mental state of patients, and sleeping in AR sleep. This experiment aimed to investigate the changes in Treg/Th17 cells in the nasal mucosa of an AR mouse model and the intervention effect of an Anti-IL-17 antibody. METHODS: A mouse model of AR was induced by intraperitoneal ovalbumin (OVA) injection for sensitization and stimulation with nasal drops. The times of rubbing, sneezing, and symptomatology scores were counted and analyzed. Pathological damage to the nasal mucosa was observed by Hematoxylin-Eosin (HE) staining. Peripheral blood CD4+CD25+CD127- Treg cell levels and the content of Th17 cells were measured by flow cytometry (FCM). ELISA kits were used to detect the levels of relevant cytokines (IL-10 and TGF-ß1) secreted by Treg cells. The intervention effect of Anti-IL-17 antibody was observed by giving Anti-IL-17 antibody treatment. RESULTS: The times of rubbing, sneezing, and symptomatology scores were significantly higher in mice in the OVA group than in the Control group (p < 0.001). The percentage of CD4+CD25+CD127- Treg cells in CD4+CD25+ T cells (p < 0.05) and the levels of IL-10 (p < 0.001) and TGF-ß1 (p < 0.001) were significantly decreased. After OVA induction, the continuity of the nasal mucosa of mice was interrupted, the percentage of Th17 cells, IL-17, and IL-4 levels were increased, and IFN-γ levels were significantly reduced (p < 0.001). And protein expression of RORγt was significantly upregulated (p < 0.001). In addition, all of these results were reversed by Anti-IL-17 antibody treatment, significantly improving AR-related symptoms (p < 0.05). CONCLUSION: Anti-IL-17 antibodies may regulate the body's immune response by promoting CD4+CD25+CD127- Treg cell differentiation, thereby ameliorating the symptoms associated with AR.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-17 , Ratones Endogámicos BALB C , Mucosa Nasal , Rinitis Alérgica , Linfocitos T Reguladores , Células Th17 , Animales , Linfocitos T Reguladores/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/patología , Rinitis Alérgica/inmunología , Rinitis Alérgica/patología , Rinitis Alérgica/sangre , Células Th17/inmunología , Ratones , Interleucina-17/inmunología , Interleucina-17/metabolismo , Femenino , Ovalbúmina/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Anticuerpos/inmunología
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA