Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Appl Mater Interfaces ; 16(9): 11147-11158, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38407048

RESUMEN

The interferon-λ (IFN-λ)-regulated innate immune responses in the airway expand our understanding toward antiviral strategies against influenza A virus (IAV). The application of IFN-λ as mucosal antiviral therapeutic is still challenging, and advanced research will be necessary to achieve more efficient delivery of recombinant IFN-λs to the damaged respiratory mucosa. In this study, we examine the capability of IFN-λ to stimulate the innate immune response, promoting the swift elimination of IAV in the lungs. Additionally, we develop IFN-λ-loaded nanoparticles incorporated into pulmonary surfactant for inhalation therapy aimed at treating lung infections caused by IAV. We found that inhaled delivery of IFNλ-PSNPs significantly restricted IAV replication in the lungs from 3 days after infection (dpi), and IAV-caused lung histopathologic findings were completely improved in response to IFNλ-PSNPs. More significant and rapid attenuation of viral RNA was observed in the lung of mice with inhaled delivery of IFNλ-PSNPs compared to mice with recombinant IFN-λs. Inhalation treatment of IFNλ-PSNPs to IAV-infected mice can result in the increase of monocyte frequency in concert with restoration of T and B cells composition. Furthermore, the transcriptional profiles of monocytes shifted toward heightened IFN responses following IFNλ-PSNP treatment. These results imply that IFN-λ could serve as a robust inducer of innate immunity in the lungs against IAV infection, and inhalation of IFN-λs encapsulated in PSNPs effectively resolves lung infections caused by IAV through rapid viral clearance. PSNPs facilitated improved delivery of IFN-λs to the lungs, triggering potent antiviral immune responses upon IAV infection onset.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Surfactantes Pulmonares , Animales , Ratones , Humanos , Interferón lambda , Inmunidad Innata/genética , Pulmón/patología
3.
BMC Microbiol ; 23(1): 154, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237381

RESUMEN

BACKGROUND: Allergic rhinitis (AR) is characterized by airway inflammation in nasal mucosa from inhaled allergens and interleukin (IL)-33 is the potent inducer of Th2 inflammation in allergic nasal epithelium. Staphylococcus epidermidis is one of the most abundant colonizers of the healthy human nasal mucosa and might impact the allergen-induced inflammatory responses in the nasal epithelium. Thus, we sought to characterize the mechanism of S. epidermidis regulating Th2 inflammation and IL-33 production in AR nasal mucosa. RESULTS: The AR symptoms were alleviated and eosinophilic infiltration, serum IgE levels, and Th2 cytokines were significantly decreased in OVA-sensitized AR mice in response to human nasal commensal S. epidermidis. The inoculation of S. epidermidis to normal human nasal epithelial cells reduced IL-33 and GATA3 transcriptions and also reduced IL-33 and GATA3 expression in AR nasal epithelial (ARNE) cells and the nasal mucosa of AR mice. Our data exhibited that the cellular necroptosis of ARNE cells might be involved in IL-33 production and inoculation of S. epidermidis decreased the phosphorylation of necroptosis enzymes in ARNE cells, which was related to the reduction of IL-33 production. CONCLUSIONS: We present that human nasal commensal S. epidermidis reduces allergic inflammation by suppressing IL-33 production in nasal epithelium. Our findings indicate that S. epidermidis serves a role in blocking allergen-induced cellular necroptosis in allergic nasal epithelium which might be a key mechanism of reduction of IL-33 and Th2 inflammation.


Asunto(s)
Rinitis Alérgica , Staphylococcus epidermidis , Humanos , Animales , Ratones , Interleucina-33 , Necroptosis , Inmunoglobulina E/metabolismo , Células Th2 , Mucosa Nasal , Alérgenos , Inflamación , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos BALB C
4.
Front Immunol ; 13: 1009424, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36524125

RESUMEN

Introduction: The innate immune responses of upper airway could further our understanding toward antiviral strategies against SARS-CoV-2. We characterize the potential of interferon (IFN)-λ as an innate immune inducer for the rapid clearance of SARS-CoV-2 in the lung and the therapeutic efficacy of intranasal inoculation of IFN-λ to resolve acute lung infection. Methods: Syrian golden hamsters were infected with SARS-CoV-2 and the dynamics of SARS-CoV-2 infection depending on IFN-λ inoculation were tested. Results: SARS-CoV-2-infected Syrian golden hamsters exhibited a significant decrease in body weight and high viral mRNA level at 3 days post-infection (dpi). Although viral replication was reduced completely from 7 dpi, the pathologic findings remained prominent until 14 dpi in the lung of hamsters. The transcription of IFN-λ was significantly induced in response to SARS-CoV-2 infection with the increase of IFN-stimulated genes. Intranasal inoculation of IFN-λ restricted SARS-CoV-2 replication in the lungs of infected completely from 3 dpi with markedly reduction of inflammatory cytokines. The transcriptional phenotypes were altered to the direction of damage repair and tissue remodeling in the lungs of SARS-CoV-2-infected hamsters following intranasal inoculation of IFN-λ, which improved SARS-CoV-2-caused lung damage. Conclusion: Collectively, our findings suggest that IFN-λ might be a potent innate immune inducer in the lung and intranasal inoculation of IFN-λ resolves SARS-CoV-2 infection with rapid viral clearance and improvement of lung damage.


Asunto(s)
COVID-19 , SARS-CoV-2 , Cricetinae , Animales , COVID-19/patología , Interferón lambda , Carga Viral , Mesocricetus , Pulmón
5.
Int Immunopharmacol ; 112: 109180, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36030690

RESUMEN

Remarkable progress has recently been achieved to identify the biological function and potential value of novel therapeutic targets for the effective control of allergic asthma. Interferon (IFN)-λ has been suggested to restrict chronic inflammation in the lungs of asthmatic mice and we sought to determine the contribution of IFN-λ as an asthma therapeutic. We show that inhaled IFN-λ can restrict Th2 and Th17 inflammation in the lungs of asthmatic mice, accompanied with alteration of IL-10 secretion. BALB/C mice were used for an asthmatic mouse model with OVA. Recombinant IFN-λs (IFN-λ2: 2 µg, IFN-λ3: 2 µg) were inoculated into asthmatic mice after OVA challenge by intranasal delivery. Lungs of asthmatic mice were severely inflamed, with extensive inflammatory cell infiltration and increased goblet cell metaplasia with higher total lung resistance. Transcription of IL-4, IL-5, IL-13, and IL-17A was significantly higher until five days after the final OVA challenge. Asthmatic mice were administered recombinant IFN-λ via inhalation three times after the last challenge and the asthmatic mice showed improvement in lung histopathologic findings, and total lung resistance was maintained under normal range. IFN-λ inhalation exhibited significant decreases in Th2 and Th17 cytokine levels, and the populations of Th2 and Th17 cells were recovered from the lungs of asthmatic mice. Additionally, increase in IL-10 secretion from CD4 + Th cells population was observed in response to inhaled delivery of IFN-λ along with alterations in Th2 and Th17 cell-derived inflammation. Our findings show that inhaled delivery of IFN-λ can restrict airway inflammation in the lungs of asthmatic mice by controlling Th2- and Th17-mediated responses accompanied by regulation of IL-10 secretion even after asthma development.


Asunto(s)
Asma , Células Th17 , Ratones , Animales , Interleucina-17 , Interleucina-13 , Interleucina-10/uso terapéutico , Interleucina-5 , Interleucina-4 , Ovalbúmina , Ratones Endogámicos BALB C , Líquido del Lavado Bronquioalveolar , Asma/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Citocinas , Modelos Animales de Enfermedad , Interferones/uso terapéutico , Inmunidad , Células Th2
6.
NPJ Biofilms Microbiomes ; 8(1): 26, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35418111

RESUMEN

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


Asunto(s)
Virus de la Influenza A , Mucosa Nasal , Staphylococcus epidermidis , Internalización del Virus , Humanos , Virus de la Influenza A/fisiología , Interferones , Mucosa Nasal/microbiología , Mucosa Nasal/virología , Serina Proteasas
8.
iScience ; 24(10): 103172, 2021 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-34604720

RESUMEN

Emerging evidence indicates that severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) is transmitted through the human nasal mucosa via the principal entry factors angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2), which are highly expressed in the nasal epithelium. Therefore, the biologics targeting host entry factors on human nasal mucosa will be necessary for complete control of SARS-CoV-2. Our data reveal that ACE2 was more abundant in human nasal mucosa than lung tissue. Both ACE2 and TMPRSS2 transcriptions significantly decreased in nasal epithelium in response to S. epidermidis and were relatively lower in human nasal mucus with large numbers of S. epidermidis. ACE2 transcription was also reduced in nasal epithelium in response to nasal symbiont S. aureus. This study proposes that Staphylococcus species nasal commensals might potentially restrict SARS-CoV-2 entry to the nasal epithelium via down regulation of cellular receptors coupled with reduction of principal host protease.

9.
BMC Microbiol ; 20(1): 301, 2020 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-33028252

RESUMEN

BACKGROUND: The host-microbial commensalism can shape the innate immune responses in respiratory mucosa and nasal microbiome also modulates front-line immune mechanism in the nasal mucosa. Inhaled allergens encounter the host immune system first in the nasal mucosa, and microbial characteristics of nasal mucus directly impact the mechanisms of initial allergic responses in nasal epithelium. However, the roles of the nasal microbiome in allergic nasal mucosa remain uncertain. We sought to determine the distribution of nasal microbiomes in allergic nasal mucosa and elucidate the interplay between nasal microbiome Staphylococcus species and Th2 cytokines in allergic rhinitis (AR) models. RESULTS: Staphylococcus aureus (AR-SA) and S. epidermidis (AR-SE) were isolated from the nasal mucosa of patients with AR. The influence of nasal microbiome Staphylococcus species on allergic nasal mucosa was also tested with in vitro and in vivo AR models. Pyrosequencing data showed that colonization by S. epidermidis and S. aureus was more dominant in nasal mucus of AR subjects. The mRNA and protein levels of IL-33 and TSLP were significantly higher in AR nasal epithelial (ARNE) cells which were cultured from nasal mucosa of AR subjects, and exposure of ARNE cells to AR-SA reduced IL-33 mRNA and secreted protein levels. Particularly, ovalbumin-driven AR mice inoculated with AR-SA by intranasal delivery exhibited significantly reduced IL-33 in their nasal mucosa. In the context of these results, allergic symptoms and Th2 cytokine levels were significantly downregulated after intranasal inoculation of AR-SA in vivo AR mice. CONCLUSION: Colonization by Staphylococcus species was more dominant in allergic nasal mucosa, and nasal commensal S. aureus from subjects with AR mediates anti-allergic effects by modulating IL-33-dependent Th2 inflammation. The results demonstrate the role of host-bacterial commensalism in shaping human allergic inflammation.


Asunto(s)
Inmunidad Innata , Mucosa Nasal/inmunología , Rinitis Alérgica/inmunología , Staphylococcus aureus/inmunología , Staphylococcus epidermidis/inmunología , Simbiosis/inmunología , Animales , Corynebacterium/crecimiento & desarrollo , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Enterobacter aerogenes/crecimiento & desarrollo , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Femenino , Expresión Génica , Humanos , Interleucina-33/genética , Interleucina-33/inmunología , Ratones Endogámicos BALB C , Micrococcus luteus/crecimiento & desarrollo , Moco/inmunología , Moco/microbiología , Mucosa Nasal/microbiología , Ovalbúmina/administración & dosificación , Cultivo Primario de Células , ARN Mensajero/genética , ARN Mensajero/inmunología , Rinitis Alérgica/inducido químicamente , Rinitis Alérgica/microbiología , Rinitis Alérgica/patología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus epidermidis/crecimiento & desarrollo
10.
Antiviral Res ; 180: 104860, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32565134

RESUMEN

Middle East Respiratory Syndrome Coronavirus (MERS-CoV) causes severe respiratory in human with high mortality and it has been a challenge to determine optimum treatment for MERS-CoV-induced respiratory infection. Here, we observed the distribution of MERS-CoV receptors using human respiratory mucosa and also evaluated the contribution of interferon-lambdas (IFN-λs) in response to MERS-CoV infection using in vitro normal human nasal epithelial (NHNE) and bronchial epithelial (NHBE) cells. We found that the gene and protein expression of DPPIV, MERS-CoV receptor, were more dominantly located in nasal and bronchial epithelium although human nasal mucosa exhibited relatively lower DPPIV expression than lung parenchymal tissues. The quantitative mRNA level of the MERS-CoV envelope (upE) gene was significantly induced in MERS-CoV-infected cultured NHNE and NHBE cells until 3 days after infection. The induction of IFNs was identified in NHNE and NHBE cells after MERS-CoV infection and IFN-λs were predominantly increased in MERS-CoV-infected respiratory epithelial cells. Inoculation of IFN-λs to NHNE and NHBE cells suppressed MERS-CoV replication and in particular, IFN-λ4 showed a strong therapeutic effect in reducing MERS-CoV infection with higher induction of IFN-stimulated genes. Thus, IFN-λ has a decisive function in the respiratory epithelium that greatly limits MERS-CoV replication, and may be a key cytokine for better therapeutic outcomes against MERS-CoV infection in respiratory tract.


Asunto(s)
Antivirales/uso terapéutico , Interferones/uso terapéutico , Interleucinas/uso terapéutico , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Mucosa Respiratoria/virología , Replicación Viral/efectos de los fármacos , Bronquios/virología , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Citocinas/metabolismo , Células Epiteliales/virología , Regulación Viral de la Expresión Génica , Humanos , Inmunidad Innata/inmunología , Interferones/biosíntesis , Interleucinas/biosíntesis , Mucosa Laríngea/virología , Pulmón/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Reacción en Cadena de la Polimerasa , Mucosa Respiratoria/efectos de los fármacos
11.
Cytokine ; 119: 32-36, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30861490

RESUMEN

The possibility has been suggested that interferon (IFN)-λs can be induced rapidly for restricting respiratory viral infection in asthmatic mice and may modulate Th2-related immune responses that underlie the pathogenesis of asthma. We sought to determine the in vivo contribution of IFN-λs on decrease of Th2 cytokines in the respiratory tract of in vivo asthma. Lungs of asthmatic mice were severely inflamed, with extensive inflammatory cell infiltration and increased goblet cell metaplasia with higher total lung resistance. The mean protein levels of TSLP and IL-33 from BAL fluid of asthmatic mice were significantly higher until 7 days. Following the collection of lung tissue of 20 asthmatic mice, TSLP and IL-33 gene expressions inversely correlated with mRNA levels of IFN-λ2/3. Asthmatic mice were administered recombinant IFN-λ2/3 via the intranasal route and the mRNA levels of IFN-stimulated genes were elevated to an even greater extent in the lung tissue of the mice without intranasal IFN-λ2/3. Asthma-related histopathologic lung inflammation was significantly improved and total lung resistance was maintained within normal range in IFN-λ2/3-treated asthmatic mice. Moreover, IFN-λ2/3-treated asthmatic mice exhibited significant decrease of secreted protein levels of TSLP and IL-33 in the BAL fluid until 7 days after IFN administration. The current data provide compelling evidence that the compensation of IFN-λs can restrict the secretion of epithelial-derived Th2 cytokines, accompanied with reduced asthmatic immunopathology and IFN-λs are critical for limiting Th2-mediated allergic responses in allergic asthma.


Asunto(s)
Asma/inmunología , Citocinas/inmunología , Células Epiteliales/inmunología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Células Th2/inmunología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Interleucina-33/inmunología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología
12.
Sci Rep ; 6: 31169, 2016 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-27484958

RESUMEN

We investigated whether cilostazol, an activator of cyclic adenosine monophosphate (cAMP)-dependent intracellular signaling, could inhibit ultraviolet B (UVB) irradiation-induced photoaging in HR-1 hairless mice. Cilostazol decreased wrinkle formation and skin thickness in UVB-irradiated mice, as well as increased staining of collagen fibers and inhibition of reactive oxygen species (ROS) formation in the skin. Moreover, the proteolytic activities of gelatinase matrix metalloproteinase (MMP)-9 and collagenase MMP-3 were significantly decreased in UVB-irradiated mice treated with cilostazol. Western blotting showed that UVB-induced activation of p38 mitogen-activated protein kinases (MAPK) and nuclear factor (NF)-κB was significantly inhibited by cilostazol, whereas the activation of Akt was significantly enhanced by cilostazol. Confirmation of localized protein expression in the skin revealed marked p38 MAPK and NF-κB activation that was mainly detected in the dermis. Marked Akt activation was mainly detected in the epidermis. Our results suggest that cilostazol may have anti-photoaging effects on UVB-induced wrinkle formation by maintaining the extracellular matrix density in the dermis, which occurs via regulation of ROS and related p38 MAPK and NF-κB signaling, and subsequent down-regulation of MMPs. Therefore, cilostazol may protect against photoaging-induced wrinkle formation.


Asunto(s)
Inhibidores de Fosfodiesterasa 3/administración & dosificación , Envejecimiento de la Piel/efectos de la radiación , Tetrazoles/administración & dosificación , Rayos Ultravioleta , Animales , Cilostazol , Colágeno/análisis , Metaloproteinasa 3 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Ratones Pelados , Modelos Animales , Especies Reactivas de Oxígeno/análisis , Piel/patología , Envejecimiento de la Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...