Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Pharmacology ; 109(1): 22-33, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37980896

RESUMEN

INTRODUCTION: We investigated the potential of LPS (10-300 µg/rat) administered intratracheally (i.t.) to induce reproducible features of acute lung injury (ALI) and compared the pharmacological efficacy of anti-inflammatory glucocorticoids and antifibrotic drugs to reduce the disease. Additionally, we studied the time-dependent progression of ALI in this LPS rat model. METHODS: We conducted (1) dose effect studies of LPS administered i.t. at 10, 30, 100, and 300 µg/rat on ALI at 4 h timepoint; (2) pharmacological interventions using i.t. fluticasone (100 and 300 µg/rat), i.t. pirfenidone (4,000 µg/rat), and peroral dexamethasone (1 mg/kg) at 4 h timepoint; (3) kinetic studies at 0, 2, 4, 6, 8, 10, and 24 h post-LPS challenge. Phenotype or pharmacological efficacy was assessed using predetermined ALI features such as pulmonary inflammation, edema, and inflammatory mediators. RESULTS: All LPS doses induced a similar increase of inflammation, edema, and inflammatory mediators, e.g., IL6, IL1ß, TNFα, and CINC-1. In pharmacological intervention studies, we showed fluticasone and dexamethasone ameliorated ALI by inhibiting inflammation (>60-80%), edema (>70-100%), and the increase of cytokines IL6, IL1ß, and TNFα (≥70-90%). We also noticed some inhibition of CINC-1 (25-35%) and TIMP1 (57%) increase with fluticasone and dexamethasone. Conversely, pirfenidone failed to inhibit inflammation, edema, and mediators of inflammation. Last, in ALI kinetic studies, we observed progressive pulmonary inflammation and TIMP1 levels, which peaked at 6 h and remained elevated up to 24 h. Progressive pulmonary edema started between 2 and 4 h and was sustained at later timepoints. On average, levels of IL6 (peak at 6-8 h), IL1ß (peak at 2-10 h), TNFα (peak at 2 h), CINC-1 (peak at 2-6 h), and TGFß1 (peak at 8 h) were elevated between 2 and 10 h and declined toward 24 h post-LPS challenge. CONCLUSION: Our data show that 10 µg/rat LPS achieved a robust, profound, and reproducible experimental ALI phenotype. Glucocorticoids ameliorated key ALI features at the 4-h timepoint, but the antifibrotic pirfenidone failed. Progressive inflammation and sustained pulmonary edema were present up to 24 h, whereas levels of inflammatory mediators were dynamic during ALI progression. This study's data might be helpful in designing appropriate experiments to test the potential of new therapeutics to cure ALI.


Asunto(s)
Lesión Pulmonar Aguda , Neumonía , Edema Pulmonar , Piridonas , Ratas , Animales , Lipopolisacáridos/toxicidad , Fluticasona/uso terapéutico , Fluticasona/farmacología , Factor de Necrosis Tumoral alfa/genética , Interleucina-6 , Cinética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/tratamiento farmacológico , Pulmón , Neumonía/inducido químicamente , Inflamación , Mediadores de Inflamación , Dexametasona/farmacología , Dexametasona/uso terapéutico , Edema
2.
EMBO Mol Med ; 15(9): e17748, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37538042

RESUMEN

Hematopoietic cell transplantation (HCT) treats many blood conditions but remains underused due to complications such as graft-versus-host disease (GvHD). In GvHD, donor immune cells attack the patient, requiring powerful immunosuppressive drugs like glucocorticoids (GCs) to prevent death. In this study, we tested the hypothesis that donor cell conditioning with the glucocorticoid fluticasone propionate (FLU) prior to transplantation could increase hematopoietic stem cell (HSC) engraftment and reduce GvHD. Murine HSCs treated with FLU had increased HSC engraftment and reduced severity and incidence of GvHD after transplantation into allogeneic hosts. While most T cells died upon FLU treatment, donor T cells repopulated in the hosts and appeared less inflammatory and alloreactive. Regulatory T cells (Tregs) are immunomodulatory and survived FLU treatment, resulting in an increased ratio of Tregs to conventional T cells. Our results implicate an important role for Tregs in maintaining allogeneic tolerance in FLU-treated grafts and suggest a therapeutic strategy of pre-treating donor cells (and not the patients directly) with GCs to simultaneously enhance engraftment and reduce GvHD upon allogeneic HCT.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Ratones , Animales , Fluticasona/farmacología , Fluticasona/uso terapéutico , Trasplante Homólogo/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Enfermedad Injerto contra Huésped/prevención & control , Inmunosupresores
3.
Colloids Surf B Biointerfaces ; 228: 113364, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37290201

RESUMEN

The nano/micron sized-fluticasone propionate inhalable suspension (FPs) is used for asthma treatment, and this study aimed to elucidate the effects of particle size on the absorption of FPs by various pulmonary cells and the subsequent therapeutic efficacy for asthma. FPs of 727, 1136 and 1612 nm were prepared, and an increase in diameter diminished the endocytosis and macropinocytosis of FPs by alveolar epithelial cells (A549 and Calu-3 cells) but facilitated their uptake by M2-like macrophages; results about the transport across Calu-3 monolayer showed the mucus layer was the main rate-limiting step for the uptake of FPs by epithelial cells; the animal tests showed that although a decrease in diameter improved the pulmonary absorption of FPs, the particle size did not affect the lung distribution of FPs; a further detection revealed that larger FPs were taken more effectively by alveolar macrophages and lymphocytes and exerted a better therapeutic effect on asthma than the smaller ones. This study showed that the particle size of FPs had a significant impact on their absorption, elimination and cellular distribution in the lung after inhalation and further on their effectiveness in asthma treatment, and the particle size of the nano/micron sized-FPs should be designed and optimized for asthma treatment on the premise of meeting the requirements of inhalation preparations.


Asunto(s)
Androstadienos , Asma , Animales , Fluticasona/farmacología , Fluticasona/uso terapéutico , Tamaño de la Partícula , Androstadienos/uso terapéutico , Asma/tratamiento farmacológico , Pulmón , Administración por Inhalación
4.
BMC Nephrol ; 24(1): 63, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36949400

RESUMEN

BACKGROUND: Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulopathy worldwide, and lacks the effective treatment. The study was aimed to investigate the clinical efficacy of fluticasone propionate aerosol combined with angiotensin converting enzyme inhibitor / angiotensin receptor blocker (ACEI/ARB) in the treatment of IgAN. METHODS: 142 patients with biopsy-proven IgAN at Shenzhen People?s hospital from June 2018 to June 2020 were enrolled. The patients were randomly divided into the supportive care plus fluticasone group and the supportive care group. The patients of the supportive care plus fluticasone group were treated with fluticasone propionate aerosol (250 ?g Bid) combined with ACEI/ARB, while the supportive care group was merely treated with ACEI/ARB. The patients were followed up at 3, 6 and 9 months after enrollment. Primary outcomes include changes in proteinuria and estimated glomerular filtration rate (eGFR). RESULTS: The level of proteinuria in the supportive care plus fluticasone group was significantly lower compared with the supportive care group at 0, 3, 6 and 9 months. Meanwhile, during the follow-up period, no serious adverse events were recorded during the study in either group. However, fluticasone treatment did not alleviate the decline in eGFR. CONCLUSION: Fluticasone propionate aerosol combined with ACEI/ARB can reduce the level of proteinuria in thetreatment of IgAN, and has no significant effects on renal function.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina , Fluticasona , Glomerulonefritis por IGA , Humanos , Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Fluticasona/uso terapéutico , Fluticasona/farmacología , Tasa de Filtración Glomerular , Glomerulonefritis por IGA/tratamiento farmacológico , Proteinuria/tratamiento farmacológico , Resultado del Tratamiento
5.
Rhinology ; 61(2): 161-169, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-36479866

RESUMEN

BACKGROUND: Disruption of the nasal epithelial barrier is believed to play a role in Coronavirus Disease-2019 (COVID-19) outcomes. Fluticasone propionate has been shown to restore the nasal epithelial barrier in allergic rhinitis to the level of healthy controls. The therapeutic potential of nasal steroid sprays in COVID-19 has recently been reported. However, further insight into the mode of action is warranted. OBJECTIVES: To explore the in vitro mechanisms of the preventive potential of fluticasone propionate in SARS-CoV-2 infection. METHODS: Human air liquid interface cultures of Calu-3 cells and primary nasal epithelial cells isolated from healthy donors were used to investigate the preventive effect of fluticasone propionate on SARS-CoV-2 induced barrier disruption, virus replication and ACE2 expression. RESULTS: 48 hours pre-treatment with fluticasone propionate prevented the SARS-CoV-2 induced increase in fluorescein isothiocyanate-dextran 4 kDa permeability and reduced infection with SARS-CoV-2. Pre-treatment with fluticasone propionate also decreased ACE2 expression in SARS-CoV-2 infected Calu-3 cells. CONCLUSION: Fluticasone propionate pre-treatment prevented SARS-CoV-2 increased epithelial permeability, reduced ACE2 expression and SARS-CoV-2 infection, underscoring the therapeutic potential of fluticasone propionate in the context of COVID-19.


Asunto(s)
Antiinflamatorios , COVID-19 , Humanos , Fluticasona/farmacología , Fluticasona/uso terapéutico , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2 , Androstadienos/farmacología , Androstadienos/uso terapéutico , Rociadores Nasales , Permeabilidad
6.
Pulm Pharmacol Ther ; 77: 102171, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36243386

RESUMEN

AIMS: To investigate the pharmacokinetics and effects on the hypothalamic-pituitary-adrenal (HPA) axis of mometasone furoate (MF), fluticasone propionate (FP) and fluticasone furoate (FF). METHODS: Study 1: Fourteen healthy participants received inhaled and intravenous MF (inhaled dose via Twisthaler) and FP (inhaled dose via Diskus), both given at 400 µg, using a randomised, single-dose, four-way crossover design. Study 2: Twenty-seven participants with mild to moderate asthma, who discontinued their corticosteroid medication for 5 days to obtain a baseline 24 h serum cortisol, received inhaled MF Twisthaler and FP Diskus, both given at 400 µg twice daily (BID), using a randomised, 14-day repeat dose, two-way crossover design. Study 3: Forty-four healthy participants were randomised to a double-blind, placebo-controlled, five-period crossover study where the following treatments were administered via the inhaled route for 7 days: FP Diskus (250, 500, 1000 µg BID), FF Diskus (100, 200, 400, 800, 1600 µg once daily [QD]) or placebo Diskus. In each study, 24-h serial blood samples were collected and assayed to assess concentrations of MF, 6ß-hydroxy mometasone, mometasone, FP, FF and cortisol. Pharmacokinetic and serum cortisol parameters were estimated as geometric means and 95% confidence intervals (CI). RESULTS: Study 1: For intravenous MF and FP, respectively: absolute bioavailability was 11.4% (95% CI: 7.5, 17.6) and 7.8% (6.3, 9.6); plasma clearance was 47 L/h (41, 52) and 60 L/h (52, 69); half-life was 7.4 h (6.9, 8.0) and 7.2 h (6.5, 8.0); and volume of distribution was 499 L (439, 567) and 623 L (557, 698). Inhalation of single dose MF or FP did not significantly affect serum cortisol (<10% reduction from baseline), whereas intravenous administration of MF or FP each changed serum cortisol by approximately -50% from baseline. Study 2: For MF and FP, respectively: area under the curve up to the last measurable concentration on Day 1 was 421 pg h/mL (270, 659) and 248 pg h/mL (154, 400), and on Day 14 was 1092 pg h/mL (939, 1269) and 591 pg h/mL (501, 696); absolute bioavailability was 12.8% (11.2, 14.2) and 8.9% (7.7, 10.2). On Day 14, 24-h serum cortisol change from baseline was -35% (-44%, -26%) and -18% (-28%, -5%) for MF and FP, respectively; the reduction was significantly greater for MF than FP (ratio for geometric adjusted mean serum cortisol concentration: 1.28 [1.04, 1.56]). Low plasma concentrations of 6ß-hydroxy mometasone were detected after intravenous dosing (Study 1) and after multiple inhaled dosing (Study 2); mometasone was not detected in any samples. Study 3: Inhaled FP and FF had similar systemic bioavailability estimates (12.0% [11.0, 13.2] and 15.0% [12.0, 17.3], respectively), but a differential effect on the HPA axis which was in agreement with the known 1.7-fold higher glucocorticoid receptor-binding affinity of FF versus FP. However, for FP 250 µg BID and FF 100, 200 and 400 µg QD, reduction in serum cortisol was not significantly different from placebo. For higher doses, FP 500 and 1000 µg BID, and FF 800 and 1600 µg QD, changes in serum cortisol concentration relative to placebo were -30%, -70%, -41% and -90%, respectively. Repeat inhaled dosing of FP 1000 µg/day (within the therapeutic dose range) resulted in comparable cortisol suppression to MF in the therapeutic range (30% reduction); whereas for FF this occurred at more than 3-fold above the therapeutic dose range (644 µg/day). CONCLUSIONS: Single inhaled and intravenous doses of MF and FP (400 µg) resulted in similar bioavailability and reductions in serum cortisol. Repeat dosing of inhaled MF and FP in the therapeutic range (800 µg/day) resulted in greater systemic exposure for MF, and a 35% reduction in serum cortisol that was 2-fold greater than for FP. The higher glucocorticoid receptor-binding affinity and bioavailability, lower clearance and the presence of active metabolites may contribute to the greater systemic exposure and effect on cortisol for MF. Repeat dosing of inhaled FP and FF resulted in similar systemic bioavailability but differed in terms of the dose required for comparable cortisol suppression to MF in the therapeutic range. Unlike FP and FF, MF has active metabolites that may contribute to its systemic effects, while device/formulation performance differences also exist between MF-containing products.


Asunto(s)
Sistema Hipotálamo-Hipofisario , Receptores de Glucocorticoides , Humanos , Fluticasona/farmacología , Furoato de Mometasona/farmacología , Estudios Cruzados , Sistema Hipófiso-Suprarrenal , Androstadienos/farmacología , Administración por Inhalación , Hidrocortisona/farmacología , Método Doble Ciego
7.
Sci Rep ; 12(1): 12334, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35853985

RESUMEN

The most classic treatment recommended in the current chronic obstructive pulmonary disease (COPD) guidelines is glucocorticoid and ß2 receptor agonist combination, such as salmeterol xinafoate and fluticasone propionate (Sal/Flu), causing many adverse reactions due to hormones. Magnesium isoglycyrrhizinate (MgIG) is an anti-inflammatory glycyrrhizic acid preparation for treating chronic inflammation, contributing to its structure is similar to steroidal anti-inflammatory drugs. In this study, we successfully established COPD rat model by endotracheal-atomized lipopolysaccharide exposure and cigarette smoke induction, as characterized by lung function decline. We discovered that salmeterol xinafoate/MgIG combination could alleviated lung inflammation infiltration, airway wall thickness (AWT) and the secretion of bronchial mucin MUC5AC of COPD rats more than salmeterol xinafoate, MgIG, or salmeterol xinafoate and fluticasone propionate treatment did, as well as reduced inflammatory cells (white blood cells, neutrophils and lymphocytes) accumulation in bronchoalveolar lavage fluid and decreased TNF-α, IL-6 and IL-1ß production in the serum of COPD rats. Finally, we found that Moreover, the mechanism involved might be related to the suppression of JAK/STAT signaling pathway. Overall, our studies suggested that MgIG might be a potential alternative adjuvant drug for fluticasone propionate for the clinical treatment of patients with COPD.


Asunto(s)
Broncodilatadores , Enfermedad Pulmonar Obstructiva Crónica , Administración por Inhalación , Albuterol/uso terapéutico , Androstadienos/farmacología , Animales , Antiinflamatorios/uso terapéutico , Broncodilatadores/efectos adversos , Combinación de Medicamentos , Fluticasona/farmacología , Fluticasona/uso terapéutico , Ratas , Xinafoato de Salmeterol/farmacología , Xinafoato de Salmeterol/uso terapéutico , Saponinas , Triterpenos
8.
Sci Transl Med ; 14(641): eabl8146, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35442706

RESUMEN

Asthma and inflammatory airway diseases restrict airflow in the lung, compromising gas exchange and lung function. Inhaled corticosteroids (ICSs) can reduce inflammation, control symptoms, and improve lung function; however, a growing number of patients with severe asthma do not benefit from ICS. Using bronchial airway epithelial brushings from patients with severe asthma or primary human cells, we delineated a corticosteroid-driven fibroblast growth factor (FGF)-dependent inflammatory axis, with FGF-responsive fibroblasts promoting downstream granulocyte colony-stimulating factor (G-CSF) production, hyaluronan secretion, and neutrophilic inflammation. Allergen challenge studies in mice demonstrate that the ICS, fluticasone propionate, inhibited type 2-driven eosinophilia but induced a concomitant increase in FGFs, G-CSF, hyaluronan, and neutrophil infiltration. We developed a model of steroid-induced neutrophilic inflammation mediated, in part, by induction of an FGF-dependent epithelial-mesenchymal axis, which may explain why some individuals do not benefit from ICS. In further proof-of-concept experiments, we found that combination therapy with pan-FGF receptor inhibitors and corticosteroids prevented both eosinophilic and steroid-induced neutrophilic inflammation. Together, these results establish FGFs as therapeutic targets for severe asthma patients who do not benefit from ICS.


Asunto(s)
Asma , Factores de Crecimiento de Fibroblastos , Corticoesteroides/farmacología , Corticoesteroides/uso terapéutico , Animales , Fluticasona/farmacología , Fluticasona/uso terapéutico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Ácido Hialurónico , Inflamación/tratamiento farmacológico , Ratones
9.
Sci Rep ; 12(1): 5132, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35332239

RESUMEN

Non-T2 severe asthma and chronic obstructive pulmonary disease (COPD) are airway chronic inflammatory disorders with a poor response to corticosteroids. LAS194046, a novel pan-Janus kinase (JAK) inhibitor, shows inhibitory effects on T2 allergic lung inflammation in rats. In this work we analyze the effects of LAS194046, fluticasone propionate and their combination in neutrophils from non-T2 severe asthma and COPD patients in vitro. Neutrophils from 23 healthy subjects, 23 COPD and 21 non-T2 severe asthma patients were incubated with LAS194046 (0.01 nM-1 µM), fluticasone propionate (0.1 nM-1 µM) or their combination and stimulated with lipopolysaccharide (LPS 1 µM). LAS194046 shows similar maximal % inhibition and potency inhibiting IL-8, MMP-9 and superoxide anion release in neutrophils from healthy, COPD and asthma. Fluticasone propionate suppresses mediator release only in neutrophils from healthy patients. The combination of LAS194046 with fluticasone propionate shows synergistic anti-inflammatory and anti-oxidant effects. The mechanisms involved in the synergistic effects of this combination include the increase of MKP1 expression, decrease of PI3Kδ, the induction of glucocorticoid response element and the decrease of ERK1/2, P38 and JAK2/STAT3 phosphorylation compared with monotherapies. In summary, LAS194046 shows anti-inflammatory effects in neutrophils from COPD and severe non-T2 asthma patients and induces synergistic anti-inflammatory effects when combined with fluticasone propionate.


Asunto(s)
Asma , Inhibidores de las Cinasas Janus , Enfermedad Pulmonar Obstructiva Crónica , Administración por Inhalación , Androstadienos/farmacología , Animales , Antiinflamatorios/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Fluticasona/farmacología , Fluticasona/uso terapéutico , Humanos , Activación Neutrófila , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Ratas
10.
Expert Rev Pharmacoecon Outcomes Res ; 22(5): 743-751, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35176212

RESUMEN

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is an inflammatory lung disease characterized by long-term breathing problems and airflow limitations. International guidelines recommend using bronchodilators like long-acting beta- and muscarinic antagonists, and inhalational corticosteroids. OBJECTIVES: The cost-effectiveness of single-inhaler triple therapy containing fluticasone furoate/umeclidinium/vilanterol (FF/UMEC/VI) was compared to the treatments Fluticasone Furoate/Vilanterol (FF/VI), Umeclidinio/Vilanterol (UMEC/VI) and Fluticasone Propionate 250 mcg/Salmeterol 25mcg + Tiotropio 18 mcg (FP/SAL/TIO) for patients with COPD from the Chilean public health system perspective. METHODS: A cost-effectiveness analysis was performed, including a deterministic and probabilistic sensitivity analysis over a 25-year time horizon. Two scenarios were assessed to study the effect of a 3%-discount for costs and outcomes on FF/UMEC/VI. RESULTS: The incremental cost-effectiveness (ICER) of FF/UMEC/VI versus FF/VI was $10,076/QALY, being a cost-effective alternative to a threshold of one Gross Domestic Product per capita (GDPpc), while versus FP/SAL/TIO the ICER increased to $50,288/QALY, showing to be a non-cost effective alternative to 1 GDPpc, but at a threshold of 3 GDPpc. CONCLUSION: FF/UMEC/VI appears to be a cost-effective intervention for treating COPD compared to FF/VI. However, FF/UMEC/VI compared to FP/SAL/TIO showed an ICER above the threshold of 1 GDPpc, but, in comparison with lower price, the ICER was below 3 GDPpc.


Asunto(s)
Salud Pública , Enfermedad Pulmonar Obstructiva Crónica , Androstadienos/farmacología , Androstadienos/uso terapéutico , Alcoholes Bencílicos , Broncodilatadores/uso terapéutico , Chile , Clorobencenos , Análisis Costo-Beneficio , Método Doble Ciego , Combinación de Medicamentos , Fluticasona/farmacología , Fluticasona/uso terapéutico , Volumen Espiratorio Forzado , Humanos , Quinuclidinas , Resultado del Tratamiento
12.
Sci Rep ; 11(1): 21603, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732778

RESUMEN

Ovarian chronic inflammation has been created and extended in the laying hen mainly via increasing laying frequency and microbial infection, especially during the late stage of production period. This study was aimed to evaluate glucocorticoid (GC) Fluticasone as an anti-inflammatory agent on the gene expression of the ovarian pro-and anti-inflammatory mediators (follicular cyclooxygenases COX 1, 2, and cytokines), inflammatory responses of the immune system, ovarian functions (ovulation rate and follicular growths), and hormones in the commercial-aged laying hens. White Leghorn hens aged 92-weeks were used for four weeks to be supplemented by 2 ppm Fluticasone as an optimum dose obtained in a pre-trial base on ovulation rate. As compared to control, Fluticasone resulted in a significant decrease in the mRNA expression of COX-1 and pro-and anti-inflammatory cytokines, and increase in COX-2 mRNA expression and heterophil to lymphocyte ratio (P < 0.001). A significant reduction was observed in the ovulation rate, follicular size (P < 0.001), ovarian hormones, immunoglobulins, body weight, and food consummation (P ≤ 0.05) by administering GC Fluticasone. Although a relative anti-inflammatory improvement was created by Fluticasone in the ovarian condition, the administration of this glucocorticoid resulted in a considerable reduction in ovarian hormones and functions of commercial aged laying hens.


Asunto(s)
Antiinflamatorios/farmacología , Fluticasona/farmacología , Hormonas/metabolismo , Sistema Inmunológico , Inflamación/inmunología , Ovario/inmunología , Animales , Pollos , Femenino , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ovario/efectos de los fármacos , Ovario/metabolismo , Ovulación
13.
J Microencapsul ; 38(3): 177-191, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33583315

RESUMEN

This work aimed to elaborate an optimised fluticasone propionate (FP)-loaded solid lipid nanoparticles (SLNs) to enhance FP effectiveness for topical inflammatory remediation. The influences of drug amount, lipid, and surfactant ratios, on drug release pattern and stability were investigated utilising Box-Behnken design. Elaboration, characterisation, and pharmacodynamic evaluation in comparison with the marketed formulation (Cutivate® cream, 0.05%w/w FP), were conducted for the optimised SLNs. The optimised SLNs with a size of 248.3 ± 1.89 nm (PDI = 0.275) and -32.4 ± 2.85 mV zeta potential were evidenced good stability physiognomies. The optimised SLNs pre-treated rats exhibited non-significant difference in paw volume from that of the control group and showed a significant reduction in both PGE2 and TNF-α levels by 51.5 and 61%, respectively, in comparison with the Carrageenan group. The optimised FP-loaded SLNs maximised the efficacy of FP towards inflammation alleviation that increase its potential as efficient implement in inflammatory skin diseases remediation.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/farmacología , Fluticasona/administración & dosificación , Fluticasona/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Carragenina , Dinoprostona/metabolismo , Composición de Medicamentos , Liberación de Fármacos , Estabilidad de Medicamentos , Fluticasona/farmacocinética , Pie/patología , Inflamación/inducido químicamente , Inflamación/prevención & control , Lípidos/química , Masculino , Nanopartículas , Tamaño de la Partícula , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Breath Res ; 15(1): 016008, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33045700

RESUMEN

Fractional exhaled nitric oxide (FENO) is a marker of airway inflammation. Measuring FENO at multiple flow rates enables calculation of NO parameters: bronchial NO output (J awNO), bronchial wall (C awNO) and alveolar (C ANO) NO concentrations, and bronchial diffusion factor of NO (D awNO). FENO is known to rapidly reduce after the commencement of inhaled corticosteroid (ICS) treatment. However, little is known on the effect of ICS on the other NO parameters. We assessed (1) the onset of action of ICS treatment on the NO parameters and (2) whether the changes in bronchial NO output are due to changes in bronchial wall NO concentration or diffusion factor. FENO and other NO parameters were measured at baseline and after 1, 3 and 7 d of treatment with inhaled fluticasone propionate 250 µg b.i.d. in 23 allergic children with a history of asthma-like symptoms. There was a decrease in J awNO (from 680 (244/1791) (median (1st/3rd quartile)) to 357 (165/753) pl s-1, p < 0.001) and FENO50( from 13.8 (7.5/35) to 8.3 (5.36/17.0) ppb, p < 0.001) in 3 d from the first dose of ICS. Also, C awNO seemed to reduce after 3 d (from 171 (89/328) to 79 (54/157) ppb, p = 0.041), while D awNO remained unchanged. Furthermore, C ANO reduced during the 7 d treatment (from 3.0 (2.0/5.0) to 2.3 (1.9/2.6) ppb, p = 0.004). ICS treatment reduced FENO50 and J awNO rapidly and the decline was caused by decreased bronchial wall NO concentration while bronchial NO diffusion factor remained unchanged. These findings suggest that C awNO could be a more specific marker of airway inflammation and treatment response than J awNO or FENO50, which are both determined also by D awNO that seems to be resistant to the treatment with ICS.


Asunto(s)
Bronquios/metabolismo , Glucocorticoides/administración & dosificación , Glucocorticoides/farmacología , Óxido Nítrico/metabolismo , Alveolos Pulmonares/metabolismo , Administración por Inhalación , Corticoesteroides/administración & dosificación , Corticoesteroides/farmacología , Pruebas Respiratorias , Bronquios/efectos de los fármacos , Niño , Espiración , Fluticasona/administración & dosificación , Fluticasona/farmacología , Humanos , Masculino , Alveolos Pulmonares/efectos de los fármacos
15.
Molecules ; 25(17)2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32859128

RESUMEN

Chitosan (CS) is a natural polysaccharide, widely studied in the past due to its unique properties such as biocompatibility, biodegradability and non-toxicity. Chemical modification of CS is an effective pathway to prepare new matrices with additional functional groups and improved properties, such as increment of hydrophilicity and swelling rate, for drug delivery purposes. In the present study, four derivatives of CS with trans-aconitic acid (t-Acon), succinic anhydride (Succ), 2-hydroxyethyl acrylate (2-HEA) and acrylic acid (AA) were prepared, and their successful grafting was confirmed by FTIR and 1H-NMR spectroscopies. Neat chitosan and its grafted derivatives were fabricated for the encapsulation of fluticasone propionate (FLU) and salmeterol xinafoate (SX) drugs, used for chronic obstructive pulmonary disease (COPD), via the ionotropic gelation technique. Scanning electron microscopy (SEM) micrographs demonstrated that round-shaped microparticles (MPs) were effectively prepared with average sizes ranging between 0.4 and 2.2 µm, as were measured by dynamic light scattering (DLS), while zeta potential verified in all cases their positive charged surface. FTIR spectroscopy showed that some interactions take place between the drugs and the polymeric matrices, while X-ray diffraction (XRD) patterns exhibited that both drugs were encapsulated in MPs' interior with a lower degree of crystallinity than the neat drugs. In vitro release studies of FLU and SX exposed a great amelioration in the drugs' dissolution profile from all modified CS's MPs, in comparison to those of neat drugs. The latter fact is attributed to the reduction in crystallinity of the active substances in the MPs' interior.


Asunto(s)
Fluticasona , Xinafoato de Salmeterol , Administración por Inhalación , Cápsulas , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Fluticasona/química , Fluticasona/farmacocinética , Fluticasona/farmacología , Humanos , Tamaño de la Partícula , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Xinafoato de Salmeterol/química , Xinafoato de Salmeterol/farmacocinética , Xinafoato de Salmeterol/farmacología
16.
Artículo en Inglés | MEDLINE | ID: mdl-32637364

RESUMEN

Human rhinoviruses (HRVs) are associated with acute exacerbations in patients with chronic obstructive pulmonary disease (COPD) and asthma, which are accompanied by mucus hypersecretion. Whereas, various studies have shown that HRVs increase epithelial mucin production and thus may directly contribute to mucus hypersecretion. The effects of drugs used in the treatment of COPD and asthma on HRV-induced mucin production in epithelial cell cultures have not been studied. In the present study, we assessed effects of HRVs on mucin production and secretion in well-differentiated primary human bronchial epithelial cells (PBEC) and studied the effect of the inhaled corticosteroid fluticasone propionate and the long-acting muscarinic antagonist tiotropium bromide on this process. Differentiated PBEC that were cultured at the air-liquid interface (ALI-PBEC) were infected with HRV-A16 and HRV-1B. Quantitative PCR, immunofluorescence staining, ELISA, periodic acid-Schiff (PAS) staining and immunostaining assays were used to assess the effects of HRV infection. Here we demonstrate that both HRV-A16 and HRV-1B increased mucin (MUC5AC and MUC5B) gene expression and protein release. When exploring this in more detail in HRV-A16-infected epithelial cells, mucin expression was found to be accompanied by increases in expression of SAM-pointed domain-containing Ets-like factor (SPDEF) and SPDEF-regulated genes known to be involved in the regulation of mucin production. We also found that pre-treatment with the purinergic P2R antagonist suramin inhibits HRV-enhanced MUC5AC expression and protein release, implicating involvement of purinergic signaling by extracellular ATP. We furthermore found that both fluticasone and tiotropium decreased HRV-induced mucin production without affecting viral replication, and obtained evidence to suggest that the inhibitory effect of fluticasone involved modulation of SPDEF-regulated genes and extracellular ATP release. These data show that both tiotropium and fluticasone inhibit HRV-induced epithelial mucin production independent of viral clearance, and thus provide insight into the mechanisms underlying beneficial effects of tiotropium and fluticasone in the treatment of COPD, asthma and accompanying exacerbations in these patients. Furthermore, our findings provide additional insight into the mechanisms by which HRV increases epithelial mucin production.


Asunto(s)
Células Epiteliales , Rhinovirus , Células Cultivadas , Fluticasona/farmacología , Humanos , Proteínas Proto-Oncogénicas c-ets , Bromuro de Tiotropio/farmacología
18.
Respirology ; 25(8): 827-835, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32026586

RESUMEN

BACKGROUND AND OBJECTIVE: Abnormal peripheral airway function is an important feature of asthma and relates to asthma symptoms and poor asthma control. We aimed to determine whether peripheral airway function, as measured by forced oscillatory impedance and multiple-breath nitrogen washout (MBNW), relates to symptom improvement in asthmatic participants with uncontrolled asthma, after stepping up to high-dose ICS/LABA treatment. METHODS: A total of 19 subjects (14 females, mean age: 29.9 ± 13.6 years) with uncontrolled asthma, as defined by an ACQ5 > 1.5, taking 500 µg/day fluticasone equivalent or less, underwent spirometry, plethysmography, fractional exhaled FeNO, forced oscillatory resistance (Rrs5Hz ) and reactance (Xrs5Hz ), and indices of MBNW ventilation heterogeneity (lung clearance index (LCI), diffusion-convection-dependent (Sacin) and convection-dependent (Scond)). Measurements were made before and after 8 weeks of treatment with fluticasone/formoterol combination inhaler 250/10 µg, 2 puffs twice daily. RESULTS: Treatment improved ACQ5 (P = 0.0002), FEV1 (P = 0.02), FVC (P = 0.04), FeNO (P = 0.0008), Xrs5Hz (P = 0.01), LCI (P = 0.0002), Sacin (P = 0.006) and Scond (P = 0.01). At baseline, ACQ5 correlated with Xrs5Hz (rs = 0.52, P = 0.03) and Rrs5Hz (rs = 0.55, P = 0.02). The improvement in ACQ5 was predicted by more abnormal baseline LCI (P = 0.03), Scond (P = 0.02) and Rrs5Hz (P = 0.006). Baseline Scond was the best predictor of a clinically meaningful improvement in asthma control (ΔACQ > 0.5, ROC-AUC = 0.91, P = 0.007). CONCLUSION: Step-up to high-dose combination treatment in uncontrolled asthma is associated with improved peripheral airway function as measured by Xrs5Hz and MBNW. Baseline MBNW and FOT parameters correlated with the improvement in symptoms and may predict a positive response to up-titration in uncontrolled asthmatic patients.


Asunto(s)
Asma/tratamiento farmacológico , Asma/prevención & control , Oscilometría , Ventilación Pulmonar , Administración por Inhalación , Corticoesteroides/administración & dosificación , Corticoesteroides/uso terapéutico , Agonistas de Receptores Adrenérgicos beta 2/uso terapéutico , Adulto , Asma/fisiopatología , Combinación de Medicamentos , Femenino , Fluticasona/administración & dosificación , Fluticasona/farmacología , Fluticasona/uso terapéutico , Fumarato de Formoterol/administración & dosificación , Fumarato de Formoterol/farmacología , Fumarato de Formoterol/uso terapéutico , Humanos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Nitrógeno/metabolismo , Ventilación Pulmonar/efectos de los fármacos , Curva ROC , Respiración , Pruebas de Función Respiratoria , Espirometría
20.
Auris Nasus Larynx ; 47(3): 415-424, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31822346

RESUMEN

OBJECTIVE: Particulate matter (PM), which contains organic compounds and toxic metals, is the major cause of air pollution. PM enters the body, causing various health problems. Although the effects of PM on the lower respiratory tract have been extensively investigated, the effects on the upper respiratory tract (including the nasal cavity) require further evaluation. To investigate the effect of fluticasone propionate (FP) on nasal fibroblasts exposed to UPM. METHODS: Samples of inferior turbinate tissue were obtained from six patients. The fibroblasts isolated from these samples were exposed to UPM and/or FP. The expression of interleukin (IL)-6, CXC chemokine ligand (CXCL) 1, IL-1ß, and tumour necrosis factor-alpha (TNF-α) in nasal fibroblasts was analysed using real-time PCR and enzyme-linked immunosorbent assays. The protein levels of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) were analysed by western blotting. RESULTS: FP reversed the UPM-induced reduction in cell viability. The mRNA and protein levels of IL-6, CXCL1, IL-1ß, and TNF-α were significantly increased by UPM. FP reversed the UPM-induced increases in the protein levels of NF-κB and phosphorylated-STAT3 in a dose-dependent manner. In addition, TNF-α, an inducer of NF-κB, reversed the FP-induced reduction in the levels of signalling molecules. CONCLUSION: UPM induces the expression of IL-6, CXCL1, IL-1ß, and TNF-α in nasal fibroblasts and this effect is reversed by FP via the STAT3 and NF-κB signalling pathways. These results suggest that FP has therapeutic potential for nasal diseases related to UPM, such as allergic and chronic rhinitis.


Asunto(s)
Citocinas/metabolismo , Fibroblastos/efectos de los fármacos , Fluticasona/farmacología , Material Particulado/farmacología , Adulto , Células Cultivadas , Quimiocina CXCL1/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Material Particulado/efectos adversos , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...