Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.269
Filtrar
1.
Basic Clin Pharmacol Toxicol ; 135(1): 98-108, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38784974

RESUMEN

Adenosine, lidocaine and Mg2+ (ALM) solution is an emerging therapy that reduces secondary injury after intravenous administration in experimental models of traumatic brain injury (TBI). Intranasal delivery of ALM may offer an alternative route for rapid, point-of-care management of TBI. As a preliminary safety screen, we evaluated whether ALM exerts cytotoxic or inflammatory effects on primary human nasal epithelial cells (pHNEC) in vitro. Submerged monolayers and air-liquid interface cultures of pHNEC were exposed to media only, normal saline only, therapeutic ALM or supratherapeutic ALM for 15 or 60 min. Safety was measured through viability, cytotoxicity, apoptosis, cellular and mitochondrial stress, and inflammatory mediator secretion assays. No differences were found in viability or cytotoxicity in cultures exposed to saline or ALM for up to 60 min, with no evidence of apoptosis after exposure to supratherapeutic ALM concentrations. Despite comparable inflammatory cytokine secretion profiles and mitochondrial activity, cellular stress responses were significantly lower in cultures exposed to ALM than saline. In summary, data show ALM therapy has neither adverse toxic nor inflammatory effects on human nasal epithelial cells, setting the stage for in vivo toxicity studies and possible clinical translation of intranasal ALM therapy for TBI treatment.


Asunto(s)
Adenosina , Administración Intranasal , Apoptosis , Supervivencia Celular , Células Epiteliales , Lidocaína , Mucosa Nasal , Humanos , Lidocaína/administración & dosificación , Lidocaína/toxicidad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Adenosina/administración & dosificación , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Magnesio/administración & dosificación , Citocinas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo
2.
Am J Vet Res ; 85(6)2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38697189

RESUMEN

OBJECTIVE: To examine the potential of galangin in a mouse model of ovalbumin (OVA)-induced allergic rhinitis (AR), as chronic AR, induced by immunoglobulin-E (IgE), leads to histamine release and nasal inflammation, and although galangin exhibits antiasthmatic and anti-inflammatory potential, its effect on AR is yet to be investigated. ANIMALS: 126 BALB/c mice. METHODS: AR induction involved sensitizing female mice with OVA (5%, 500 µL, IP) for 14 days. Post OVA challenge, the mice were divided into 7 groups (n = 18/group), including normal, AR control, montelukast (10 mg/kg), galangin (5, 10, and 20 mg/kg), and per se (galangin [20 mg/kg] treatment. Various outcomes were evaluated, including nasal symptoms, histopathology, biochemistry, and nasal lavage fluid inflammatory cytokines and signaling pathways in nasal mucosal to assess galangin potential in AR. RESULTS: In AR mice, galangin (10 and 20 mg/kg) significantly (P < .05) reduced sneezing, rubbing, and nasal discharge post-OVA challenge. Galangin treatment attenuated (P < .05) elevated serum histamine, ß-hexosaminidase, IgE, and Immunoglobulin G1 levels in AR control mice. Additionally, galangin significantly (P < .05) decreased OVA-induced alterations in IL-4, IL-6, IL-13, and interferon-γ levels in nasal lavage fluid compared to AR control mice. Western blot analysis demonstrated that galangin lowered OVA-induced AR by significantly (P < .05) downregulating the phosphorylated protein kinase B and mammalian target of rapamycin-protein expressions while markedly (P < .05) upregulating the glycogen synthase kinase-3ß protein expressions in nasal mucosal. Galangin also significantly ameliorated (P < .05) the OVA-induced histological aberrations in the nasal mucosa, reflected by reduced eosinophil infiltration, hyperplasia, and edema. CLINICAL RELEVANCE: Galangin exhibits antihistaminic and anti-inflammatory effects in AR mice by regulating IgE-mediated histamine and inflammatory release and modulating the phosphatidylinositol 3-kinase/Ak strain transforming/mammalian target of rapamycin pathways.


Asunto(s)
Flavonoides , Ratones Endogámicos BALB C , Ovalbúmina , Rinitis Alérgica , Animales , Flavonoides/farmacología , Flavonoides/uso terapéutico , Ratones , Femenino , Rinitis Alérgica/tratamiento farmacológico , Rinitis Alérgica/inducido químicamente , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Modelos Animales de Enfermedad , Quinolinas/farmacología , Quinolinas/uso terapéutico , Citocinas/metabolismo , Mucosa Nasal/efectos de los fármacos , Inmunoglobulina E/sangre , Acetatos , Ciclopropanos , Sulfuros
3.
Environ Toxicol Pharmacol ; 108: 104459, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38685369

RESUMEN

Non-smokers exposed to second-hand smoke (SHS) present risk of developing tobacco smoke-associated pathologies. To investigate the airway molecular response to SHS exposure that could be used in health risk assessment, comparative shotgun proteomics was performed on nasal epithelium from a group of healthy restaurant workers, non-smokers (never and former) exposed and not exposed to SHS in the workplace. HIF1α-glycolytic targets (GAPDH, TPI) and proteins related to xenobiotic metabolism, cell proliferation and differentiation leading to cancer (ADH1C, TUBB4B, EEF2) showed significant modulation in non-smokers exposed. In never smokers exposed, enrichment of glutathione metabolism pathway and EEF2-regulating protein synthesis in genotoxic response were increased, while in former smokers exposed, proteins (LYZ, ATP1A1, SERPINB3) associated with tissue damage/regeneration, apoptosis inhibition and inflammation that may lead to asthma, COPD or cancer, were upregulated. The identified proteins are potential response and susceptibility/risk biomarkers for SHS exposure.


Asunto(s)
Mucosa Nasal , Exposición Profesional , Proteómica , Contaminación por Humo de Tabaco , Humanos , Contaminación por Humo de Tabaco/efectos adversos , Exposición Profesional/efectos adversos , Mucosa Nasal/metabolismo , Mucosa Nasal/efectos de los fármacos , Adulto , Masculino , Restaurantes , Femenino , Persona de Mediana Edad
4.
Braz J Otorhinolaryngol ; 90(3): 101399, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38442638

RESUMEN

OBJECTIVES: Montelukast is a well-known leukotriene receptor antagonist commonly used in treating allergic rhinitis and asthma. Omega-3 fatty acid is also known as an antiallergic and immunomodulator molecule. This study aimed to elucidate the efficacy of systemic montelukast and omega-3 fatty acid treatment in allergic rhinitis models in Wistar Hannover rats. METHODS: This research was conducted on 28 healthy Wistar Hannover rats weighing 250-350 g. After establishing the allergic rhinitis model, nasal symptoms were observed and scored, and the nasal mucosa of all rats was investigated histologically. Light microscopy was utilized to evaluate the degree of ciliary loss, goblet cell hyperplasia, vascular congestion, vascular proliferation, inflammatory cell infiltration, eosinophil infiltration, and hypertrophy in chondrocytes. RESULTS: As a result of the analysis of the data obtained from the study, it was determined that typical allergic rhinitis symptoms such as nasal scratching and sneezing were significantly reduced in the rats in the montelukast and omega-3 treated group, and these symptoms did not increase after repeated intranasal OVA-protease applications. Histological examinations after fish oil treatment did not reveal typical inflammatory changes in allergic rhinitis. None of the rats in the montelukast and omega-3 groups had any increase in goblet cells, whereas 14.3% of the rats in the control group and 28.6% of the rats in the allergic rhinitis group had mild increase. Last but not least, 71.4% of rats in the allergic rhinitis group had a moderate increase. The difference between the groups was statistically significant (p < 0.001). CONCLUSION: Regarding the outcomes of this research, it was observed that w-3 fatty acids had antiallergic effects, both histopathological and clinical, in the allergic rhinitis model. We believe that further randomized controlled trials incorporating larger cohorts are warranted to verify the use of omega-3 fatty acids in treating allergic rhinitis. The level of evidence of this article is Level 2.


Asunto(s)
Acetatos , Ciclopropanos , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3 , Aceites de Pescado , Antagonistas de Leucotrieno , Ovalbúmina , Quinolinas , Ratas Wistar , Rinitis Alérgica , Sulfuros , Animales , Ciclopropanos/uso terapéutico , Sulfuros/uso terapéutico , Acetatos/uso terapéutico , Quinolinas/uso terapéutico , Ácidos Grasos Omega-3/uso terapéutico , Rinitis Alérgica/tratamiento farmacológico , Rinitis Alérgica/patología , Ratas , Antagonistas de Leucotrieno/uso terapéutico , Aceites de Pescado/uso terapéutico , Masculino , Resultado del Tratamiento , Mucosa Nasal/patología , Mucosa Nasal/efectos de los fármacos
5.
J Laryngol Otol ; 138(5): 520-526, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38380493

RESUMEN

OBJECTIVE: To investigate epidermal growth factor, transforming growth factor-α and interleukin-8 production in nasal mucosa irrigated with hypertonic 2.3 per cent solution with algae extracts, in comparison to 0.9 per cent NaCl during the first two weeks after surgery for nasal polyposis, in relation to symptoms and local findings. METHODS: This prospective study included 20 nasal polyposis patients postoperatively irrigated with hypertonic solution and 20 nasal polyposis patients postoperatively irrigated with isotonic solution. We evaluated nasal symptom score, endoscopic score and mediator levels in nasal secretions before and after irrigation. RESULTS: Following treatment, nasal symptom score and endoscopic score were significantly lower in the hypertonic solution group (p = 0.023; p < 0.001, respectively). The increase in the epidermal growth factor and the decrease in the transforming growth factor-α and interleukin-8 concentration were higher in the hypertonic group (p < 0.001 for all mediators). CONCLUSION: Irrigation with a hypertonic solution was found to be more effective than an isotonic solution in nasal mucosa reparation.


Asunto(s)
Factor de Crecimiento Epidérmico , Interleucina-8 , Lavado Nasal (Proceso) , Mucosa Nasal , Pólipos Nasales , Agua de Mar , Factor de Crecimiento Transformador alfa , Humanos , Pólipos Nasales/cirugía , Pólipos Nasales/metabolismo , Masculino , Femenino , Estudios Prospectivos , Interleucina-8/metabolismo , Interleucina-8/análisis , Adulto , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Mucosa Nasal/efectos de los fármacos , Lavado Nasal (Proceso)/métodos , Factor de Crecimiento Epidérmico/análisis , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Transformador alfa/metabolismo , Factor de Crecimiento Transformador alfa/análisis , Endoscopía/métodos , Soluciones Hipertónicas , Anciano , Adulto Joven
6.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35074895

RESUMEN

The development of small-molecules targeting different components of SARS-CoV-2 is a key strategy to complement antibody-based treatments and vaccination campaigns in managing the COVID-19 pandemic. Here, we show that two thiol-based chemical probes that act as reducing agents, P2119 and P2165, inhibit infection by human coronaviruses, including SARS-CoV-2, and decrease the binding of spike glycoprotein to its receptor, the angiotensin-converting enzyme 2 (ACE2). Proteomics and reactive cysteine profiling link the antiviral activity to the reduction of key disulfides, specifically by disruption of the Cys379-Cys432 and Cys391-Cys525 pairs distal to the receptor binding motif in the receptor binding domain (RBD) of the spike glycoprotein. Computational analyses provide insight into conformation changes that occur when these disulfides break or form, consistent with an allosteric role, and indicate that P2119/P2165 target a conserved hydrophobic binding pocket in the RBD with the benzyl thiol-reducing moiety pointed directly toward Cys432. These collective findings establish the vulnerability of human coronaviruses to thiol-based chemical probes and lay the groundwork for developing compounds of this class, as a strategy to inhibit the SARS-CoV-2 infection by shifting the spike glycoprotein redox scaffold.


Asunto(s)
Amino Alcoholes/farmacología , Enzima Convertidora de Angiotensina 2/química , Antivirales/farmacología , Éteres Fenílicos/farmacología , Receptores Virales/química , SARS-CoV-2/efectos de los fármacos , Glicoproteína de la Espiga del Coronavirus/química , Compuestos de Sulfhidrilo/farmacología , Regulación Alostérica , Amino Alcoholes/química , Enzima Convertidora de Angiotensina 2/antagonistas & inhibidores , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/química , Sitios de Unión , COVID-19/virología , Línea Celular , Disulfuros/antagonistas & inhibidores , Disulfuros/química , Disulfuros/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/virología , Oxidación-Reducción , Éteres Fenílicos/química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Receptores Virales/antagonistas & inhibidores , Receptores Virales/genética , Receptores Virales/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína de la Espiga del Coronavirus/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Compuestos de Sulfhidrilo/química , Tratamiento Farmacológico de COVID-19
7.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34768968

RESUMEN

Tissue remodeling contributes to ongoing inflammation and refractoriness of chronic rhinosinusitis (CRS). During this process, epithelial-mesenchymal transition (EMT) plays an important role in dysregulated remodeling and both microRNA (miR)-29b and heat shock protein 47 (HSP47) may be engaged in the pathophysiology of CRS. This study aimed to determine the role of miR-29b and HSP47 in modulating transforming growth factor (TGF)-ß1-induced EMT and migration in airway epithelial cells. Expression levels of miR-29b, HSP47, E-cadherin, α-smooth muscle actin (α-SMA), vimentin and fibronectin were assessed through real-time PCR, Western blotting, and immunofluorescence staining. Small interfering RNA (siRNA) targeted against miR-29b and HSP47 were transfected to regulate the expression of EMT-related markers. Cell migration was evaluated with wound scratch and transwell migration assay. miR-29b mimic significantly inhibited the expression of HSP47 and TGF-ß1-induced EMT-related markers in A549 cells. However, the miR-29b inhibitor more greatly induced the expression of them. HSP47 knockout suppressed TGF-ß1-induced EMT marker levels. Functional studies indicated that TGF-ß1-induced EMT was regulated by miR-29b and HSP47 in A549 cells. These findings were further verified in primary nasal epithelial cells. miR-29b modulated TGF-ß1-induced EMT-related markers and migration via HSP47 expression modulation in A549 and primary nasal epithelial cells. These results suggested the importance of miR-29b and HSP47 in pathologic tissue remodeling progression in CRS.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Proteínas del Choque Térmico HSP47/antagonistas & inhibidores , Proteínas del Choque Térmico HSP47/genética , Factor de Crecimiento Transformador beta1/metabolismo , Células A549 , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/fisiología , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , MicroARNs/metabolismo , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Rinitis/genética , Rinitis/metabolismo , Sinusitis/genética , Sinusitis/metabolismo , Sinusitis/patología , Factor de Crecimiento Transformador beta1/administración & dosificación , Factor de Crecimiento Transformador beta1/genética
8.
Mol Med Rep ; 24(6)2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34590155

RESUMEN

Formononetin has proven to be anti­inflammatory and able to alleviate symptoms of certain allergic diseases. The present study aimed to determine and elucidate the potential effects of formononetin in allergic rhinitis. JME/CF15 cells were pretreated with formononetin at different doses, followed by stimulation with IL­13. Cell Counting Kit­8 assay was performed to determine the cytotoxicity of formononetin. The expression levels of inflammation­related proteins, histamine, IgE, TNF­α, IL­1ß, IL­6, granulocyte­macrophage colony­stimulating factor and eotaxin in IL­13­stimulated JME/CF15 cells were detected using ELISAs. The expression levels of phosphorylated­NF­κB p65, NF­κB p65 and cyclooxygenase­2 (Cox­2) were analyzed using western blotting. Reverse transcription­quantitative PCR, western blotting and immunofluorescence were performed to measure the levels of mucin 5AC oligomeric mucus/gel­forming. Expression levels of sirtuin 1 (SIRT1) and nuclear erythroid factor 2­related factor 2 (Nrf2) proteins were also measured using western blotting. The results of the present study revealed that formononetin exerted no cytotoxic effect on the viability of JME/CF15 cells. Following stimulation of JME/CF15 cells with IL­13, formononetin suppressed the upregulated expression levels of proinflammatory cytokines. IL­13­induced formation of mucus was also attenuated by formononetin treatment. Furthermore, it was found that the SIRT1/Nrf2 signaling pathway was activated in formononetin­treated JME/CF15 cells, whereas treatment with the SIRT1 inhibitor, EX527, reversed the effects of formononetin on IL­13­induced inflammation and mucus formation in JME/CF15 cells. In conclusion, the findings of the current study indicated that formononetin may activate the SIRT1/Nrf2 signaling pathway, thereby inhibiting IL­13­induced inflammation and mucus formation in JME/CF15 cells. These results suggested that formononetin may represent a promising agent for the treatment of allergic rhinitis.


Asunto(s)
Inflamación/tratamiento farmacológico , Isoflavonas/farmacología , Moco/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Sirtuina 1/metabolismo , Antiinflamatorios/farmacología , Citocinas/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Interleucina-13 , Rinitis Alérgica/tratamiento farmacológico , Transducción de Señal
9.
Tohoku J Exp Med ; 255(1): 19-25, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34497164

RESUMEN

Airborne fine particulate matter with an aerodynamic diameter equal to or smaller than 2.5 µm (abbreviated as PM2.5) increases the risk of nasal lesions, but the underlying molecular mechanism has not been fully elucidated. In the atmosphere, the composition of PM2.5 collected varies in physical and chemical properties, which affects its damage to human health. Thus, we constructed artificial PM2.5 particles based on actual PM2.5 and investigated the in vivo effects of artificial PM2.5 exposure on the oxidative stress, inflammatory response, and nasal mucosa morphology of rats. The results showed that artificial PM2.5 is comparable in composition ratio, size, and morphology to actual PM2.5. This in vivo study indicated that artificial PM2.5 exposure reduces total superoxide dismutase and glutathione peroxidase activities, elevates malondialdehyde content in the nasal mucosa, and induces increased levels of pro-inflammatory mediators, including interleukin-1, interleukin-6 and tumor necrosis factor-α. Our data shows that artificial PM2.5 particles could be used for experimental study of PM2.5 toxicology, ensuring that the physical and chemical properties of experimental PM2.5 are relatively constant and allowing for repeatability of this research. Oxidative damage and inflammatory response may be the toxic mechanisms that cause nasal lesions after exposure to artificial PM2.5.


Asunto(s)
Inflamación/etiología , Estrés Oxidativo/efectos de los fármacos , Material Particulado/toxicidad , Contaminantes Atmosféricos/química , Contaminantes Atmosféricos/toxicidad , Animales , Femenino , Humanos , Inflamación/patología , Mediadores de Inflamación/metabolismo , Exposición por Inhalación , Malondialdehído/metabolismo , Modelos Animales , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Tamaño de la Partícula , Material Particulado/química , Ratas , Ratas Sprague-Dawley
11.
Sci Rep ; 11(1): 13259, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34168212

RESUMEN

Urban particulate matter (UPM) is an important trigger of airway inflammation. The cross-talk between the external and internal matrix in the respiratory tract occurs due to the transepithelial network of macrophages/dendritic cells. This study characterized the immune processes induced by the epithelium after UPM exposure in special regard to interactions with monocyte-derived dendritic cells (moDCs) and monocyte-derived macrophages (moMφs) in obstructive lung diseases. A triple-cell co-culture model (8 controls, 10 asthma, and 8 patients with COPD) utilized nasal epithelial cells, along with moMφs, and moDCs was exposed to UPM for 24 h. The inflammatory response of nasal epithelial cells to UPM stimulation is affected differently by cell-cell interactions in healthy people, asthma or COPD patients of which the interactions with DCs had the strongest impact on the inflammatory reaction of epithelial cells after UPM exposure. The epithelial remodeling and DCs dysfunction might accelerate the inflammation after air pollution exposure in asthma and COPD.


Asunto(s)
Asma/inducido químicamente , Células Dendríticas/efectos de los fármacos , Inflamación/inducido químicamente , Macrófagos/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Material Particulado/efectos adversos , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Adulto , Anciano , Estudios de Casos y Controles , Estudios Transversales , Citocinas/metabolismo , Citometría de Flujo , Humanos , Masculino , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Estudios Prospectivos
12.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L119-L129, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34009038

RESUMEN

In vitro biomarkers to assess cystic fibrosis transmembrane conductance regulator activity are desirable for precision modulator selection and as a tool for clinical trials. Here, we describe an organoid swelling assay derived from human nasal epithelia using commercially available reagents and equipment and an automated imaging process. Cells were collected in nasal brush biopsies, expanded in vitro, and cultured as spherical organoids or as monolayers. Organoids were used in a functional swelling assay with automated measurements and analysis, whereas monolayers were used for short-circuit current measurements to assess ion channel activity. Clinical data were collected from patients on modulators. Relationships between swelling data and short-circuit current, as well as between swelling data and clinical outcome measures, were assessed. The organoid assay measurements correlated with short-circuit current measurements for ion channel activity. The functional organoid assay distinguished individual responses as well as differences between groups. The organoid assay distinguished incremental drug responses to modulator monotherapy with ivacaftor and combination therapy with ivacaftor, tezacaftor, and elexacaftor. The swelling activity paralleled the clinical response. In conclusion, an in vitro biomarker derived from patients' cells can be used to predict responses to drugs and is likely to be useful as a preclinical tool to aid in the development of novel treatments and as a clinical trial outcome measure for a variety of applications, including gene therapy or editing.


Asunto(s)
Aminofenoles/farmacología , Benzodioxoles/farmacología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Indoles/farmacología , Mucosa Nasal/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Pirrolidinas/farmacología , Quinolonas/farmacología , Estudios de Casos y Controles , Agonistas de los Canales de Cloruro/farmacología , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Transporte Iónico , Mutación , Mucosa Nasal/efectos de los fármacos , Organoides/efectos de los fármacos , Organoides/metabolismo
13.
mBio ; 12(2)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33906927

RESUMEN

SARS-CoV-2 infection causing the COVID-19 pandemic calls for immediate interventions to avoid viral transmission, disease progression, and subsequent excessive inflammation and tissue destruction. Primary normal human bronchial epithelial cells are among the first targets of SARS-CoV-2 infection. Here, we show that ColdZyme medical device mouth spray efficiently protected against virus entry, excessive inflammation, and tissue damage. Applying ColdZyme to fully differentiated, polarized human epithelium cultured at an air-liquid interphase (ALI) completely blocked binding of SARS-CoV-2 and increased local complement activation mediated by the virus as well as productive infection of the tissue model. While SARS-CoV-2 infection resulted in exaggerated intracellular complement activation immediately following infection and a drop in transepithelial resistance, these parameters were bypassed by single pretreatment of the tissues with ColdZyme mouth spray. Crucially, our study highlights the importance of testing already evaluated and safe drugs such as ColdZyme mouth spray for maintaining epithelial integrity and hindering SARS-CoV-2 entry within standardized three-dimensional (3D) in vitro models mimicking the in vivo human airway epithelium.IMPORTANCE Although our understanding of COVID-19 continuously progresses, essential questions regarding prophylaxis and treatment remain open. A hallmark of severe SARS-CoV-2 infection is a hitherto-undescribed mechanism leading to excessive inflammation and tissue destruction associated with enhanced pathogenicity and mortality. To tackle the problem at the source, transfer of SARS-CoV-2, subsequent binding, infection, and inflammatory responses have to be avoided. In this study, we used fully differentiated, mucus-producing, and ciliated human airway epithelial cultures to test the efficacy of ColdZyme medical device mouth spray in terms of protection from SARS-CoV-2 infection. Importantly, we found that pretreatment of the in vitro airway cultures using ColdZyme mouth spray resulted in significantly shielding the epithelial integrity, hindering virus binding and infection, and blocking excessive intrinsic complement activation within the airway cultures. Our in vitro data suggest that ColdZyme mouth spray may have an impact in prevention of COVID-19.


Asunto(s)
Antivirales/farmacología , Mucosa Respiratoria/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , Bronquios/citología , COVID-19/prevención & control , COVID-19/virología , Complemento C3/inmunología , Células Epiteliales , Humanos , Inmunidad Innata/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Vaporizadores Orales , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/virología , SARS-CoV-2/fisiología , Acoplamiento Viral/efectos de los fármacos
14.
Biochem Biophys Res Commun ; 554: 1-6, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33770685

RESUMEN

Exposure to fine particulate matter (PM2.5) increases the incidence of allergic rhinitis (AR). microRNA (miRNA) can regulate cell proliferation, invasion and apoptosis. However, the mechanism of miR-338-3p in mediating PM2.5-induced autophagy in AR animal models remains unknown. To explore the mechanism of miR-338-3p in PM2.5-induced autophagy in AR, the human nasal epithelium cells and AR model exposed to PM2.5 were deployed. The results showed that miR-338-3p was down-regulated in both nasal mucosa of PM2.5-exacerbated AR rat models and PM2.5-treated RPMI-2650 cells. Forced expression of miR-338-3p could inhibit autophagy in vitro. miR-338-3p specifically bound to UBE2Q1 3'-untranslated region (3' UTR) and negatively regulated its expression. Overexpression of UBE2Q1 attenuated the inhibitory effects of miR-338-3p on PM2.5-induced autophagy of RPMI-2650 cells through AKT/mTOR pathway. Moreover, our in vivo study found that after administration of agomiR-338-3p in AR rats model, the expression of autophagy-related proteins decreased and nasal symptoms alleviated. In conclusion, this study revealed that miR-338-3p acts as an autophagy suppressor in PM2.5-exacerbated AR by directly targeting UBE2Q1 and affecting AKT/mTOR pathway.


Asunto(s)
MicroARNs/genética , Mucosa Nasal/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Rinitis Alérgica/prevención & control , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Contaminantes Atmosféricos/análisis , Animales , Autofagia/fisiología , Línea Celular , Modelos Animales de Enfermedad , Humanos , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/patología , Material Particulado/administración & dosificación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Rinitis Alérgica/etiología , Rinitis Alérgica/genética , Rinitis Alérgica/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
15.
Sci Rep ; 11(1): 6351, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737566

RESUMEN

Perillyl alcohol (POH) has been extensively studied for the treatment of peripheral and primary brain tumors. The intranasal route of administration has been preferred for dosing POH in early-stage clinical trials associated with promising outcomes in primary brain cancer. However, it is unclear how intranasal POH targets brain tumors in these patients. Multiple studies indicate that intranasally applied large molecules may enter the brain and cerebrospinal fluid (CSF) through direct olfactory and trigeminal nerve-associated pathways originating in the nasal mucosa that bypass the blood-brain barrier. It is unknown whether POH, a small molecule subject to extensive nasal metabolism and systemic absorption, may also undergo direct transport to brain or CSF from the nasal mucosa. Here, we compared CSF and plasma concentrations of POH and its metabolite, perillic acid (PA), following intranasal or intravascular POH application. Samples were collected over 70 min and assayed by high-performance liquid chromatography. Intranasal administration resulted in tenfold higher CSF-to-plasma ratios for POH and tenfold higher CSF levels for PA compared to equal dose intravascular administration. Our preclinical results demonstrate POH undergoes direct transport from the nasal mucosa to the CSF, a finding with potential significance for its efficacy as an intranasal chemotherapeutic for brain cancer.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Monoterpenos/farmacología , Mucosa Nasal/efectos de los fármacos , Administración Intranasal , Animales , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/patología , Neoplasias Encefálicas/líquido cefalorraquídeo , Neoplasias Encefálicas/patología , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Humanos , Ratas , Nervio Trigémino/efectos de los fármacos
16.
Sci Rep ; 11(1): 6393, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737583

RESUMEN

Cystic fibrosis (CF) is characterized by an airway obstruction caused by a thick mucus due to a malfunctioning Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein. The sticky mucus restricts drugs in reaching target cells limiting the efficiency of treatments. The development of new approaches to enhance drug delivery to the lungs represents CF treatment's main challenge. In this work, we report the production and characterization of hybrid core-shell nanoparticles (hNPs) comprising a PLGA core and a dipalmitoylphosphatidylcholine (DPPC) shell engineered for inhalation. We loaded hNPs with a 7-mer peptide nucleic acid (PNA) previously considered for its ability to modulate the post-transcriptional regulation of the CFTR gene. We also investigated the in vitro release kinetics of hNPs and their efficacy in PNA delivery across the human epithelial airway barrier using an ex vivo model based on human primary nasal epithelial cells (HNEC) from CF patients. Confocal analyses and hNPs transport assay demonstrated the ability of hNPs to overcome the mucus barrier and release their PNA cargo within the cytoplasm, where it can exert its biological function.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/tratamiento farmacológico , Nanopartículas/química , Ácidos Nucleicos de Péptidos/farmacología , 1,2-Dipalmitoilfosfatidilcolina/química , 1,2-Dipalmitoilfosfatidilcolina/farmacología , Obstrucción de las Vías Aéreas/tratamiento farmacológico , Obstrucción de las Vías Aéreas/genética , Obstrucción de las Vías Aéreas/patología , Fibrosis Quística/genética , Fibrosis Quística/patología , Sistemas de Liberación de Medicamentos , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Moco/efectos de los fármacos , Mucosa Nasal/efectos de los fármacos , Ácidos Nucleicos de Péptidos/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/farmacología
17.
Sci Rep ; 11(1): 6206, 2021 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-33737667

RESUMEN

High mobility group box 1 (HMGB1) has been known to involve in the pathogenesis of many inflammatory diseases. The aim of this study was to establish animal model of acute rhinosinusitis (ARS), and determine whether ethyl pyruvate (EP) attenuate inflammatory response of sinonasal mucosa by inhibiting HMGB1 in ARS animals. Thirty-six Sprague Dawley (SD) rat were used as follows: six normal controls without intervention (group 1); thirty rats were used for establishment of ARS rats model by nasal insertion of Merocel sponge, and model rats without any treatments (group 2), treated with nasal drops of sterile saline (group 3), 10 µl EP (group 4), and 20 µl EP (group 5), twice a day for 5 days, respectively. Bacterial culture was done regularly and the main bacterial strains were identified using matrix-assisted laser desorption/ionization time of flight mass spectrometry. HMGB1 expression in sinonasal mucosa was detected by immunohistochemistry and RT-PCR. Serum levels of HMGB1, IL-6, and TNF-α were determined by ELISA. Data from 29 of 36 rats that had completed research were analyzed. Bacterial colony formation unit (CFU) of nasal secretion was significantly higher in each group of ARS rats compared with controls (p < 0.001). ARS rats treated with EP had only slightly decreased CFU, but significantly attenuated inflammatory response of sinonasal mucosa and decreased HMGB1 expression compared to those treated with saline alone (p < 0.001). Serum levels of HMGB1, IL-6 and TNF-α were significantly higher in ARS rats compared to controls, and decreased by EP treatments (p < 0.001). Nasal sponge packing led to acute inflammatory response of nasal sinus in rats, and increased the expression of HMGB1, IL-6, and TNF-α. Nasal drops with EP could attenuate the inflammation of sinonasal mucosa through inhibiting the expression of HMGB1, IL-6 and TNF-α in ARS rats.


Asunto(s)
Antiinflamatorios/farmacología , Proteína HMGB1/genética , Mucosa Nasal/efectos de los fármacos , Piruvatos/farmacología , Rinitis/tratamiento farmacológico , Sinusitis/tratamiento farmacológico , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Formaldehído/administración & dosificación , Formaldehído/antagonistas & inhibidores , Regulación de la Expresión Génica , Proteína HMGB1/antagonistas & inhibidores , Proteína HMGB1/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/genética , Interleucina-6/metabolismo , Masculino , Mucosa Nasal/metabolismo , Mucosa Nasal/patología , Alcohol Polivinílico/administración & dosificación , Ratas , Ratas Sprague-Dawley , Rinitis/inducido químicamente , Rinitis/genética , Rinitis/patología , Sinusitis/inducido químicamente , Sinusitis/genética , Sinusitis/patología , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
19.
HNO ; 69(12): 952-960, 2021 Dec.
Artículo en Alemán | MEDLINE | ID: mdl-33586050

RESUMEN

BACKGROUND: The e­cigarette has become increasingly popular in recent years. However, the question of toxicity is not yet clear and there is global uncertainty regarding the use of e­cigarettes. This is intensified by the fact that there is a lack of declaration of the liquid ingredients. OBJECTIVE: The present paper investigates propylene glycol, a major component of the liquids, for possible acute inflammatory reactions as well as cytotoxic and genotoxic effects on human nasal mucosa cells. MATERIALS AND METHODS: The nasal mucosa cells from 10 volunteers were cultivated at the air-liquid interface and then exposed to different concentrations of propylene glycol. The analysis was carried out using the trypan blue test, comet assay, micronucleus test, and interleukin (IL)-6 and IL­8 sandwich ELISA. RESULTS: The trypan blue test showed no reduction in vitality. No increase in IL­6 and IL­8 concentrations were detected in the sandwich ELISA. In the comet assay, the Olive tail moment showed a dose-dependent increase in DNA fragmentation compared to the negative control at all examined concentrations. A difference between the pure substance and the negative control was shown in the micronucleus test. CONCLUSION: Possibly repairable dose-dependent DNA fragmentation and profound DNA alterations at high concentrations of propylene glycol warrant enhanced genotoxicological studies. These should include long-term exposure studies and assessment of further ingredients of the liquids. Consequently, the manufacturers need to be forced to declare the latter.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Mucosa Nasal/efectos de los fármacos , Glicoles de Propileno/toxicidad , Vapeo/efectos adversos , Células Cultivadas , Ensayo Cometa , Humanos , Inflamación , Pruebas de Micronúcleos , Mucosa Nasal/citología
20.
Drug Deliv ; 28(1): 229-239, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33501873

RESUMEN

PURPOSE: The work aimed to develop a co-loaded loratadine and sulpiride nasal nanoemulsion for allergic rhinitis management. METHODS: Compatibility studies were conducted adopting differential scanning calorimetry and Fourier transform infrared spectroscopy. Nanoemulsion formulations were prepared using soybean lecithin, olive oil and tween 80. Sodium cholate and glycerol were employed as co-surfactants. Nanoemulsions were assessed for viscosity, pH, droplet size, polydispersity index, zeta potential, electrical conductivity, entrapment, In vitro drug release and corresponding kinetics. Stability of the selected formulation was investigated. The biological effectiveness was evaluated in rabbit models of ovalbumin-induced allergic rhinitis by measuring TNF-α, TGF-ß and IL-1. RESULTS: Compatibility studies revealed absence of drug/drug interactions. Nanoemulsions exhibited > 90% entrapment efficiency. The selected nanoemulsion demonstrated small droplet size (85.2 ± 0.2 nm), low PDI (0.35 ± 0.0) and appropriate Zeta Potential (-23.3 ± 0.2) and stability. It also displayed enhanced in vitro drug release following the Higuashi Diffusion and Baker-Lonsdale models. The mean relative mRNA expression of TNF-α, IL-1 and TGF-ß significantly decreased from 9.59 ± 1.06, 4.15 ± 0.02 and 4.15 ± 0.02 to 1.28 ± 0.02, 1.93 ± 0.06 and 1.56 ± 0.02 respectively after treatment with the selected nanoemulsion formulation. CONCLUSION: The results reflected a promising potent effect of the combined loratadine and sulpiride nasal nanoemulsion in managing the symptoms of allergic rhinitis.


Asunto(s)
Antagonistas de Dopamina/administración & dosificación , Emulsiones , Antagonistas de los Receptores Histamínicos H1 no Sedantes/administración & dosificación , Loratadina/administración & dosificación , Mucosa Nasal/efectos de los fármacos , Rinitis Alérgica/metabolismo , Sulpirida/administración & dosificación , Tensoactivos , Administración Intranasal , Animales , Rastreo Diferencial de Calorimetría , Modelos Animales de Enfermedad , Antagonistas de Dopamina/farmacología , Combinación de Medicamentos , Liberación de Fármacos , Glicerol , Antagonistas de los Receptores Histamínicos H1 no Sedantes/farmacología , Técnicas In Vitro , Interleucina-1/metabolismo , Lecitinas , Loratadina/farmacología , Nanoestructuras , Mucosa Nasal/metabolismo , Aceite de Oliva , Ovalbúmina , Senos Paranasales/efectos de los fármacos , Senos Paranasales/metabolismo , Polisorbatos , Conejos , Rinitis Alérgica/inducido químicamente , Colato de Sodio , Glycine max , Espectroscopía Infrarroja por Transformada de Fourier , Sulpirida/farmacología , Factor de Crecimiento Transformador beta/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA