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

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda , Pneumonia , Edema Pulmonar , Piridonas , Ratos , Animais , Lipopolissacarídeos/toxicidade , Fluticasona/uso terapêutico , Fluticasona/farmacologia , Fator de Necrose Tumoral alfa/genética , Interleucina-6 , Cinética , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Pulmão , Pneumonia/induzido quimicamente , Inflamação , Mediadores da Inflamação , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Edema
2.
EMBO Mol Med ; 15(9): e17748, 2023 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-37538042

RESUMO

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.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Camundongos , Animais , Fluticasona/farmacologia , Fluticasona/uso terapêutico , Transplante Homólogo/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Doença Enxerto-Hospedeiro/prevenção & controle , Imunossupressores
3.
BMC Nephrol ; 24(1): 63, 2023 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-36949400

RESUMO

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.


Assuntos
Inibidores da Enzima Conversora de Angiotensina , Fluticasona , Glomerulonefrite por IGA , Humanos , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Fluticasona/uso terapêutico , Fluticasona/farmacologia , Taxa de Filtração Glomerular , Glomerulonefrite por IGA/tratamento farmacológico , Proteinúria/tratamento farmacológico , Resultado do Tratamento
4.
Sci Rep ; 12(1): 12334, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35853985

RESUMO

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.


Assuntos
Broncodilatadores , Doença Pulmonar Obstrutiva Crônica , Administração por Inalação , Albuterol/uso terapêutico , Androstadienos/farmacologia , Animais , Anti-Inflamatórios/uso terapêutico , Broncodilatadores/efeitos adversos , Combinação de Medicamentos , Fluticasona/farmacologia , Fluticasona/uso terapêutico , Ratos , Xinafoato de Salmeterol/farmacologia , Xinafoato de Salmeterol/uso terapêutico , Saponinas , Triterpenos
5.
J Microencapsul ; 38(3): 177-191, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33583315

RESUMO

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.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacologia , Fluticasona/administração & dosagem , Fluticasona/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacocinética , Carragenina , Dinoprostona/metabolismo , Composição de Medicamentos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Fluticasona/farmacocinética , Pé/patologia , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Lipídeos/química , Masculino , Nanopartículas , Tamanho da Partícula , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-32637364

RESUMO

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.


Assuntos
Células Epiteliais , Rhinovirus , Células Cultivadas , Fluticasona/farmacologia , Humanos , Proteínas Proto-Oncogênicas c-ets , Brometo de Tiotrópio/farmacologia
7.
Auris Nasus Larynx ; 47(3): 415-424, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31822346

RESUMO

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.


Assuntos
Citocinas/metabolismo , Fibroblastos/efeitos dos fármacos , Fluticasona/farmacologia , Material Particulado/farmacologia , Adulto , Células Cultivadas , Quimiocina CXCL1/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/metabolismo , Humanos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Material Particulado/efeitos adversos , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L345-L355, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31747297

RESUMO

The nongenomic mechanisms by which glucocorticoids modulate ß2 agonist-induced-bronchodilation remain elusive. Our studies aimed to elucidate mechanisms mediating the beneficial effects of glucocorticoids on agonist-induced bronchodilation. Utilizing human precision-cut lung slices (hPCLS), we measured bronchodilation to formoterol, prostaglandin E2 (PGE2), cholera toxin (CTX), or forskolin in the presence and absence of budesonide. Using cultured human airway smooth muscle (HASM), intracellular cAMP was measured in live cells following exposure to formoterol, PGE2, or forskolin in the presence or absence of budesonide. We showed that simultaneous budesonide administration amplified formoterol-induced bronchodilation and attenuated agonist-induced phosphorylation of myosin light chain, a necessary signaling event mediating force generation. In parallel studies, cAMP levels were augmented by simultaneous exposure of HASM cells to formoterol and budesonide. Budesonide, fluticasone, and prednisone alone rapidly increased cAMP levels, but steroids alone had little effect on bronchodilation in hPCLS. Bronchodilation induced by PGE2, CTX, or forskolin was also augmented by simultaneous exposure to budesonide in hPCLS. Furthermore, HASM cells expressed membrane-bound glucocorticoid receptors that failed to translocate with glucocorticoid stimulation and that potentially mediated the rapid effects of steroids on ß2 agonist-induced bronchodilation. Knockdown of glucocorticoid receptor-α had little effect on budesonide-induced and steroid-dependent augmentation of formoterol-induced cAMP generation in HASM. Collectively, these studies suggest that glucocorticoids amplify cAMP-dependent bronchodilation by directly increasing cAMP levels. These studies identify a molecular mechanism by which the combination of glucocorticoids and ß2 agonists may augment bronchodilation in diseases such as asthma or chronic obstructive pulmonary disease.


Assuntos
Brônquios/fisiologia , Broncodilatadores/farmacologia , Budesonida/farmacologia , AMP Cíclico/biossíntese , Músculo Liso/fisiologia , Brônquios/efeitos dos fármacos , Carbacol/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Toxina da Cólera/farmacologia , Colforsina/farmacologia , Dinoprostona/farmacologia , Fluticasona/farmacologia , Fumarato de Formoterol/farmacologia , Humanos , Músculo Liso/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Fosforilação/efeitos dos fármacos , Prednisona/farmacologia , Receptores de Glucocorticoides/metabolismo
9.
Int J Pharm ; 572: 118839, 2019 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-31715359

RESUMO

This study intended to investigate the in vivo pulmonary fate of intratracheally dosed nanosuspensions of fluticasone propionate (FP). Three FP suspensions, including a microsuspension and two nanosuspensions with different dissolution profiles, were prepared and they exhibited comparable aerodynamic performances after nebulization via a jet nebulizer. Following intratracheal administration to rats, the microsuspension underwent extensive mucociliary clearance, leading to a limited absorption time whereas the nanosuspensions decreased the mucociliary clearance and allowed dissolution rate-limiting and extended pulmonary absorption, resulting in prolonged pulmonary retention and long-acting anti-inflammatory efficacy in a lipopolysaccharide induced lung injury model. Delaying the FP dissolution of a nanosuspension by phospholipid coating increased AUC value in lung tissues to 1.72-fold of a conventional nanosuspension, but led to a decreased pharmacological efficacy. This study demonstrated that inhalable nanosuspensions were a feasible means for the sustained pulmonary delivery of FP and the local anti-inflammatory efficacy was highly dependent on the dissolution profiles.


Assuntos
Anti-Inflamatórios/administração & dosagem , Fluticasona/administração & dosagem , Lesão Pulmonar/tratamento farmacológico , Nanopartículas , Administração por Inalação , Animais , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Área Sob a Curva , Preparações de Ação Retardada , Modelos Animais de Doenças , Liberação Controlada de Fármacos , Fluticasona/farmacocinética , Fluticasona/farmacologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nebulizadores e Vaporizadores , Ratos , Ratos Wistar , Suspensões , Distribuição Tecidual
10.
Immunopharmacol Immunotoxicol ; 41(2): 327-336, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31039648

RESUMO

Objective: Hecogenin is a sapogenin found in Agave sisalana species that is used extensively for the treatment of anti-inflammatory, antifungal, hypotensive, anti-nociceptive activity and cancer. We have studied the anti-inflammatory effect of Hecogenin and its combination with Fluticasone on atopic dermatitis and airway hyper-responsiveness in Balb/c mice. Material and methods: Dermatitis was induced by repeated application of 2, 4-dinitrofluorobenzene in Balb/c mice. After a topical application of Hecogenin, Fluticasone and their combination on the skin lesions, the ear thickness, ear weight and erythema score were evaluated. Asthma was induced by sensitization and challenge of ovalbumin in Balb/c mice. Results: The topical application of Hecogenin and its combination with Fluticasone in mice effectively suppressed the ear swelling and weight. As well as the levels of pro-inflammatory cytokines were decreased by Hecogenin and its combination in-vivo. Whereas, intra-nasal administration of Hecogenin and its combination in ovalbumin induced airway hyper-responsiveness reveals a significant decrement in total cell count, differential cell count and cytokines levels. Similar observations were obtained for myeloperoxidase level in ear and lung tissue. The results were supported by histological studies of ear and lung tissue. Conclusion: These data indicate that Hecogenin has been proved as a potential therapy for allergic skin diseases and bronchial asthma treatments in combination with Fluticasone by reducing its dose from 50 to 25 µg/mice in combination to circumvent the long term side effects of Fluticasone. The beneficial effect of Hecogenin may be related to the diminution of TNF-α and IL-12 cytokines production in Balb/c mice.


Assuntos
Anti-Inflamatórios/farmacologia , Asma/tratamento farmacológico , Dermatite Atópica/tratamento farmacológico , Interleucina-12/imunologia , Sapogeninas/farmacologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Asma/induzido quimicamente , Asma/imunologia , Asma/patologia , Dermatite Atópica/induzido quimicamente , Dermatite Atópica/imunologia , Dermatite Atópica/patologia , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Fluticasona/farmacologia , Camundongos
11.
Eur Respir J ; 53(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30846474

RESUMO

The aim was to investigate whether microRNA (miRNA) expression is modulated by inhaled corticosteroid (ICS) treatmentWe performed genome-wide miRNA analysis on bronchial biopsies of 69 moderate/severe chronic obstructive pulmonary disease (COPD) patients at baseline and after 6- and 30-month treatment with the ICS fluticasone propionate or placebo. The effect of ICS on miRNA expression was validated in differentiated primary bronchial epithelial cultures, and functional studies were conducted in BEAS-2B cells. MiRNAs affected by ICS and their predicted targets were compared to an independent miRNA dataset of bronchial brushings from COPD patients and healthy controls.Treatment with ICS for both 6 and 30 months significantly altered the expression of four miRNAs, including miR-320d, which was increased during ICS treatment compared with placebo. The ICS-induced increase of miR-320d was confirmed in primary airway epithelial cells. MiR-320d negatively correlated targets were enriched for pro-inflammatory genes and were increased in the bronchial brushes of patients with lower lung function in the independent dataset. Overexpression of miR-320d in BEAS-2B cells dampened cigarette smoke extract-induced pro-inflammatory activity via inhibition of nuclear factor-κB.Collectively, we identified miR-320d as a novel mediator of ICS, regulating the pro-inflammatory response of the airway epithelium.


Assuntos
Corticosteroides/farmacologia , Fluticasona/farmacologia , MicroRNAs/biossíntese , MicroRNAs/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/genética , Transcriptoma/efeitos dos fármacos , Corticosteroides/administração & dosagem , Idoso , Estudos Transversais , Feminino , Fluticasona/administração & dosagem , Estudo de Associação Genômica Ampla , Humanos , Masculino , Pessoa de Meia-Idade
12.
PLoS One ; 14(1): e0207675, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30677037

RESUMO

RATIONALE: Patients with COPD have an increased risk for community-acquired pneumonia, which is further increased by inhaled corticosteroids. OBJECTIVE: To assess effects of the corticosteroids, budesonide and fluticasone propionate, on macrophage bacterial responses in COPD. METHODS: Monocyte-derived macrophages (MDMs) generated from blood monocytes from 10 non-smoker controls (NoS), 20 smokers without COPD (Sm), and 40 subjects with moderate to severe COPD (21 ex-smokers (COPD-ES) and 19 current smokers (COPD-S)) were pre-treated with budesonide or fluticasone (10 nM-1 µM) and challenged with live non-typeable Haemophilus influenzae (NTHI) or Streptococcus pneumoniae (SP). Cell surface bacterial recognition receptor expression (flow cytometry) and cytokine release (bead array) were analyzed. RESULTS: NTHI and SP reduced bacterial recognition receptor expression on MDMs from COPD and Sm, but not NoS (except TLR4). SR-AI and MARCO were reduced by both NTHI and SP, whereas other receptors by either NTHI or SP. Among COPD subjects, COPD-ES demonstrated a greater number of reductions as compared to COPD-S. NTHI reduced SR-AI, MARCO, CD11b, CD35 and CD206 in COPD-ES while only SR-AI and CD11b in COPD-S. SP reduced SRA-1, CD1d, TLR2 and TLR4 in both COPD-ES and COPD-S, and reduced MARCO and CD93 only in COPD-ES. All receptors reduced in COPD by NTHI and most by SP, were also reduced in Sm. Budesonide counteracted the receptor reductions induced by both NTHI (CD206 p = 0.03, MARCO p = 0.08) and SP (SR-AI p = 0.02) in COPD-ES. Fluticasone counteracted only SP-induced reductions in TLR2 (p = 0.008 COPD-ES and p = 0.04 COPD-S) and TLR4 (p = 0.02 COPD-ES). Cytokine release was equivalently reduced by both corticosteroids. CONCLUSIONS: Reduction in macrophage bacterial recognition receptors during bacterial exposure could provide a mechanism for the increased pneumonia risk in COPD. Differential effects of budesonide and fluticasone propionate on macrophage bacterial recognition receptor expression may contribute to the higher pneumonia incidence reported with fluticasone propionate.


Assuntos
Budesonida/farmacologia , Fluticasona/farmacologia , Haemophilus influenzae/metabolismo , Macrófagos/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/microbiologia , Receptores de Superfície Celular/metabolismo , Streptococcus pneumoniae/metabolismo , Administração por Inalação , Corticosteroides/farmacologia , Adulto , Idoso , Citocinas/metabolismo , Feminino , Haemophilus influenzae/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Fumar , Streptococcus pneumoniae/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
13.
Am J Physiol Lung Cell Mol Physiol ; 316(1): L71-L81, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30335498

RESUMO

Corticosteroids (CSs) are commonly used to manage wheezing and asthma in pediatric populations. Although corticosteroids are effective in alleviating airway diseases, some children with more moderate-severe asthma phenotypes show CS resistance and exhibit significant airflow obstruction, persistent inflammation, and more frequent exacerbations. Previous studies have demonstrated that Th1 cytokines, such as TNF-α and IFN-γ, promote CS resistance in adult human airway smooth muscle (ASM). In the present study, using a human fetal ASM cell model, we tested the hypothesis that TNF-α/IFN-γ induces CS resistance. In contrast to TNF-α or IFN-γ alone, the combination of TNF-α/IFN-γ blunted the ability of fluticasone propionate (FP) to reduce expression of the chemokines CCL5 and CXCL10 despite expression of key anti-inflammatory glucocorticoid receptor target genes being largely unaffected by TNF-α/IFN-γ. Expression of the NF-κB subunit p65 and phosphorylation of Stat1 were elevated in cells treated with TNF-α/IFN-γ, an effect that remained in the presence of FP. siRNA knockdown studies demonstrated the effects of TNF-α/IFN-γ on increased p65 are mediated by Stat1, a transcription factor activated by IFN-γ. Expression of TNFAIP3, a negative regulator of NF-κB activity, was not altered by TNF-α/IFN-γ. However, the effects of TNF-α/IFN-γ were partially reduced by overexpression of TNFAIP3 but did not influence p65 expression. Together, these data suggest that IFN-γ augments the effects of TNF-α on chemokines by enhancing expression of key inflammatory pathways in the presence of CS. Interactions between TNF-α- and IFN-γ-mediated pathways may promote inflammation in asthmatic children resistant to CSs.


Assuntos
Corticosteroides/farmacologia , Brônquios/imunologia , Resistência a Medicamentos/efeitos dos fármacos , Fluticasona/farmacologia , Interferon gama/imunologia , Miócitos de Músculo Liso/imunologia , Traqueia/imunologia , Fator de Necrose Tumoral alfa/imunologia , Brônquios/crescimento & desenvolvimento , Criança , Pré-Escolar , Resistência a Medicamentos/imunologia , Feminino , Humanos , Masculino , Células Th1/imunologia , Traqueia/crescimento & desenvolvimento
14.
Int J Chron Obstruct Pulmon Dis ; 13: 2883-2897, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271135

RESUMO

BACKGROUND: Inhaled corticosteroid use is associated with increased rates of pneumonia in COPD patients. The underlying mechanism is unknown, although recent data suggest that pneumonia is more frequent in patients treated with fluticasone propionate (FP) than budesonide. Macrophages and neutrophils from COPD patients are deficient in clearing bacteria, and this might explain increased bacterial colonization in COPD. Inhaled corticosteroid may further suppress this response; therefore, we examined the effect of FP and budesonide on phagocytosis of common respiratory pathogens by monocyte-derived macrophages (MDMs) and neutrophils. METHODS: MDMs from COPD patients (n=20-24) were preincubated with FP or budesonide for 1 or 18 hours, after which phagocytosis of fluorescently labeled inert beads or heat-killed Haemophilus influenzae/Streptococcus pneumoniae were measured fluorimetrically after 1 or 4 hours. Additionally, CXCL8, IL6, and TNFα concentrations in supernatants by ELISA, MDM-scavenger-receptor expression by flow cytometry, and MDM ability to kill bacteria were measured. Neutrophils from COPD patients (n=8) were preincubated with corticosteroids for 1 hour and bacteria phagocytosis measured by flow cytometry. RESULTS: After 1 hour's preincubation, neither corticosteroid altered MDM phagocytosis of beads or H. influenzae; however, budesonide (10-7 M) increased S. pneumoniae phagocytosis by 23% (P<0.05). After 18 hours' preincubation, neither corticosteroid altered MDM phagocytosis of any prey, although H. influenzae phagocytosis by budesonide was significantly greater compared to FP at 10-6 and 10-5 M (P<0.05). The 1-hour preincubation with either corticosteroid inhibited bacteria-induced CXCL8 release (at 10-7 and 10-5 M, P<0.05); however, this effect was lost at 18-hour preincubation. There was no change in receptor expression, bacterial killing, or neutrophil phagocytosis by either corticosteroid. CONCLUSION: These data suggest that dissolved FP and budesonide do not have an overall effect on MDM or neutrophil phagocytosis of bacteria.


Assuntos
Budesonida/farmacologia , Fluticasona/farmacologia , Macrófagos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/sangue , Idoso , Células Cultivadas , Estudos de Coortes , Ensaio de Imunoadsorção Enzimática , Feminino , Haemophilus influenzae/efeitos dos fármacos , Humanos , Macrófagos/fisiologia , Masculino , Neutrófilos/fisiologia , Fagocitose/fisiologia , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Sensibilidade e Especificidade , Streptococcus pneumoniae/efeitos dos fármacos , Reino Unido
15.
Pulm Pharmacol Ther ; 50: 47-56, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29627483

RESUMO

BACKGROUND: COPD patients have increased risk of pneumonia when treated with fluticasone propionate (FP), whereas this is generally not the case with budesonide (BUD) treatment. We hypothesized that BUD and FP differentially affect the expression of immune defense genes. METHODS: Human bronchial epithelial 16HBE cells and air-liquid interface (ALI)-cultured primary bronchial epithelial cells (PBECs) were pre-treated with clinically equipotent concentrations of BUD or FP (0.16-16 nM BUD and 0.1-10 nM FP), and the expression of immune defense genes was studied at baseline and after exposure to rhinovirus (RV16). RESULTS: Using microfluidic cards, we observed that both BUD and FP significantly suppressed CXCL8, IFNB1 and S100A8 mRNA expression in unstimulated 16HBE cells. Interestingly, BUD, but not FP, significantly increased lactotransferrin (LTF) expression. The difference between the effect of BUD and FP on LTF expression was statistically significant and confirmed by qPCR and at the protein level by western blotting. RV16 infection of ALI-cultured PBECs significantly increased the expression of CCL20, IFNB1 and S100A8, but not of LTF or CAMP/LL-37. In these RV16-exposed cells, LTF expression was again significantly higher upon pre-treatment with BUD than with FP. The same was observed for S100A8, but not for CCL20, IFNB1 or CAMP/LL-37 expression. CONCLUSIONS: Treatment of human bronchial epithelial cells with BUD results in significantly higher expression of specific immune defense genes than treatment with FP. The differential regulation of these immune defense genes may help to explain the clinical observation that BUD and FP treatment differ with respect to the risk of developing pneumonia in COPD.


Assuntos
Brônquios/efeitos dos fármacos , Brônquios/imunologia , Broncodilatadores/farmacologia , Budesonida/farmacologia , Citocinas/genética , Fluticasona/farmacologia , Brônquios/citologia , Linhagem Celular , Citocinas/biossíntese , Citocinas/imunologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Humanos , Lactoferrina/biossíntese , Lactoferrina/genética , Lactoferrina/imunologia , Poli I-C/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
16.
Eur Arch Otorhinolaryngol ; 275(6): 1483-1490, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29675754

RESUMO

PURPOSE: MP 29-02, which contains fluticasone propionate and azelastine hydrochloride, is used as a topical nasal application for the treatment of seasonal and perennial allergic rhinitis. Although a multitude of data is available on the clinical symptom reduction and treatment safety of MP 29-02, the effect of MP 29-02 on ciliary beat frequency (CBF) has not been evaluated thus far. METHODS: MP 29-02-containing solution was applied at concentrations of 2.5, 5, 10, and 20% to 14 healthy subjects, and nasal ciliated epithelial cells were then visualized using a phase-contrast microscope. CBF was measured after the application of MP 29-02. For a comparison, fluticasone propionate was used. CBF measurements were then performed for 15 min at 22 °C. Ringer's solution was applied as a negative control. RESULTS: MP 29-02 significantly reduced CBF at all the tested concentrations compared with that of the control group within the observation time. At a 2.5% concentration, MP 29-02 significantly reduced CBF from 6.81 Hz (SD ± 1.35 Hz) at baseline to 4.88 Hz (SD ± 1.52 Hz, p < 0.001) after 15 min. In contrast, for fluticasone propionate, a significant reduction was observed only with the 20% concentration after 5, 10, and 15 min. CONCLUSIONS: MP 29-09 significantly reduced CB, with an almost linear relationship between the MP 29-09 concentration and reduction in CBF. For fluticasone propionate, a significant reduction of CBF was observed only at the highest analyzed concentration. The findings have implications for the long-term use of the MP 29-02. Yet, further clinical studies are needed to confirm these results in vivo, especially in patients with seasonal or perennial allergic rhinits.


Assuntos
Androstadienos/farmacologia , Células Epiteliais/efeitos dos fármacos , Fluticasona/farmacologia , Ftalazinas/farmacologia , Administração Intranasal , Adulto , Combinação de Medicamentos , Feminino , Humanos , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Mucosa Nasal/citologia , Rinite Alérgica Perene/fisiopatologia
17.
Drug Des Devel Ther ; 12: 255-269, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29440875

RESUMO

BACKGROUND AND AIM: The challenges with current antimicrobial drug therapy and resistance remain a significant global health threat. Nanodrug delivery systems are playing a crucial role in overcoming these challenges and open new avenues for effective antimicrobial therapy. While fluticasone (FLU), a poorly water-soluble corticosteroid, has been reported to have potential antimicrobial activity, approaches to optimize its dissolution profile and antimicrobial activity are lacking in the literature. This study aimed to combine an experimental study with molecular modeling to design stable FLU nanopolymeric particles with enhanced dissolution rates and antimicrobial activity. METHODS: Six different polymers were used to prepare FLU nanopolymeric particles: hydroxyl propyl methylcellulose (HPMC), poly (vinylpyrrolidone) (PVP), poly (vinyl alcohol) (PVA), ethyl cellulose (EC), Eudragit (EUD), and Pluronics®. A low-energy method, nanoprecipitation, was used to prepare the polymeric nanoparticles. RESULTS AND CONCLUSION: The combination of HPMC-PVP and EUD-PVP was found most effective to produce stable FLU nanoparticles, with particle sizes of 250 nm ±2.0 and 280 nm ±4.2 and polydispersity indices of 0.15 nm ±0.01 and 0.25 nm ±0.03, respectively. The molecular modeling studies endorsed the same results, showing highest polymer drug binding free energies for HPMC-PVP-FLU (-35.22 kcal/mol ±0.79) and EUD-PVP-FLU (-25.17 kcal/mol ±1.12). In addition, it was observed that Ethocel® favored a wrapping mechanism around the drug molecules rather than a linear conformation that was witnessed for other individual polymers. The stability studies conducted for 90 days demonstrated that HPMC-PVP-FLU nanoparticles stored at 2°C-8°C and 25°C were more stable. Crystallinity of the processed FLU nanoparticles was confirmed using differential scanning calorimetry, powder X-ray diffraction analysis and TEM. The Fourier transform infrared spectroscopy (FTIR) studies showed that there was no chemical interaction between the drug and chosen polymer system. The HPMC-PVP-FLU nanoparticles also showed enhanced dissolution rate (P<0.05) compared to the unprocessed counterpart. The in vitro antibacterial studies showed that HPMC-PVP-FLU nanoparticles displayed superior effect against gram-positive bacteria compared to the unprocessed FLU and positive control.


Assuntos
Antibacterianos/farmacologia , Portadores de Fármacos , Fluticasona/farmacologia , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Nanopartículas , Polímeros/química , Antibacterianos/química , Cristalização , Composição de Medicamentos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Fluticasona/química , Derivados da Hipromelose/química , Estrutura Molecular , Nanomedicina , Ácidos Polimetacrílicos/química , Povidona/química , Solubilidade , Tecnologia Farmacêutica/métodos
18.
Allergol Immunopathol (Madr) ; 46(3): 226-234, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29191361

RESUMO

BACKGROUND: Corticosteroids are used in the treatment of asthma. The aim of this study was to determine the efficacy of anti-IgE and anti-TNF alpha as asthma treatments. METHODS: A mouse model of chronic asthma was developed. The fluticasone group was exposed to fluticasone and the anti-IgE and anti-TNF groups were administered anti-IgE or anti-TNF. IL-4, and IgE levels were measured, and histological analysis, pathological analysis and miRNA-126, miRNA-133a analyses were applied. RESULTS: The cell concentration in the BAL fluid decreased in all the treatment groups. The rate of perivascular and peribronchial cell infiltration decreased in the lung in the high-dose anti-IgE and anti-TNF groups. Smooth muscle thickness decreased in the lung tissue in the low-dose anti-IgE and anti-TNF groups. Bronchial wall thickness decreased in the lung tissue in the fluticasone+anti-IgE group. The IL-4 level in BAL fluid decreased in the high-dose anti-IgE, fluticasone+anti-IgE and anti-TNF groups. IgE levels increased in the BAL fluid in the high-dose anti-IgE and anti-TNF groups. The lymphocyte level increased in the BAL fluid in the high-dose anti-IgE group. The macrophage level decreased in the BAL fluid in the anti-TNF group. The relative expression of miRNA-126 increased in all groups. The relative expression of miRNA-133a decreased in the placebo and fluticasone groups. The relative expression of miRNA-133a increased in the low-dose anti-IgE, high-dose anti-IgE, fluticasone+anti-IgE and anti-TNF groups. CONCLUSIONS: The results showed that anti-IgE is successful as a treatment. Fluticasone+anti-IgE and anti-TNF were seen to be superior to other therapeutic modalities when used for prophylaxis.


Assuntos
Antiasmáticos/farmacologia , Anticorpos Anti-Idiotípicos/farmacologia , Asma/imunologia , Fluticasona/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Doença Crônica , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos BALB C
19.
J Pharm Pharmacol ; 69(12): 1745-1753, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28980320

RESUMO

OBJECTIVES: Loteprednol etabonate (LE) is the first, highly successful soft corticosteroid (SC) designed using the 'inactive metabolite' approach, starting with ∆1 -cortienic acid (d-CA). The next generation of SCs based on d-CA was etiprednol dicloacetate (ED). The 17α-dichloroacetyl function serves both as a unique pharmacophore and as the source of the molecule's softness. Highly potent SCs were designed based on a combination of ED and LE, introducing 6, 9 and 16 substituents in the molecule. METHODS: The new 6α, 9α, 16α and ß 17α-dichloroacetyl 17ß-esters were synthesized from the correspondingly substituted ∆1 -cortienic acids. The anti-inflammatory activity was assessed using LPS-induced TNF α-release under various conditions to determine intrinsic activity vs. systemic biological stability. In vivo anti-inflammatory activity was studied in the widely used ovalbumin-sensitized and ovalbumin-challenged Brown Norway rat model. KEY FINDINGS: The 6α or 9α-fluoro substitution produced highly potent corticosteroids, but the 17α-dichloroacetyl substituent provided 'softness' in all cases. Local application of these steroids will significantly reduce systemic activity, due to the facile hydrolytic deactivation of these molecules. CONCLUSIONS: A 17α-dichloroacetyl derivative of fluticasone (FLU) is highly potent but much safer than the currently used propionate or furoate ester.


Assuntos
Corticosteroides/farmacologia , Anti-Inflamatórios/farmacologia , Etabonato de Loteprednol/química , Fator de Necrose Tumoral alfa/metabolismo , Corticosteroides/síntese química , Corticosteroides/química , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/química , Modelos Animais de Doenças , Fluticasona/química , Fluticasona/farmacologia , Lipopolissacarídeos/administração & dosagem , Masculino , Ovalbumina/administração & dosagem , Ratos , Ratos Endogâmicos BN , Relação Estrutura-Atividade
20.
Clin Exp Allergy ; 47(12): 1534-1545, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28833774

RESUMO

BACKGROUND: Bronchial epithelial goblet cell metaplasia (GCM) with hyperplasia is a prominent feature of asthma, but the effects of treatment with corticosteroids alone or in combination with a long-acting ß2 -adrenergic receptor agonist (LABA) on GCM in the bronchial epithelium are unknown. OBJECTIVES: To determine whether corticosteroid alone or in combination with a LABA alters protein and gene expression pathways associated with IL-13-induced goblet cell metaplasia. RESULTS: We evaluated the effects of fluticasone propionate (FP) and of salmeterol (SM), on the response of well-differentiated cultured bronchial epithelial cells to interleukin-13 (IL-13). Outcome measures included gene expression of SPDEF/FOXa2, gene expression and protein production of MUC5AC/MUC5B and morphologic appearance of cultured epithelial cell sheets. We additionally analysed expression of these genes in bronchial epithelial brushings from healthy, steroid-naïve asthmatic and steroid-treated asthmatic subjects. In cultured airway epithelial cells, FP treatment inhibited IL-13-induced suppression of FOXa2 gene expression and up-regulation of SPDEF, alterations in gene and protein measures of MUC5AC and MUC5B and induction of GCM. The addition of SM synergistically modified the effects of FP modestly-only for gel-forming mucin MUC5AC. In bronchial epithelial cells recovered from asthmatic vs healthy human subjects, we found FOXa2 and MUC5B gene expression to be reduced and SPDEF and MUC5AC gene expression to be increased; these alterations were not observed in bronchial epithelial cells recovered after treatment with inhaled corticosteroids. CONCLUSION AND CLINICAL RELEVANCE: Corticosteroid treatment inhibits IL-13-induced GCM of the airways in asthma, possibly through its effects on SPDEF and FOXa2 regulation of mucin gene expression. These effects are modestly augmented by the addition of a long-acting ß-agonist. As we found evidence for drug treatment counteracting the effects of IL-13 on the epithelium, we conclude that further exploration into the mechanisms by which corticosteroids and long-acting ß2 -adrenergic agonists confer protection against pathologic airway changes is warranted.


Assuntos
Corticosteroides/efeitos adversos , Agonistas de Receptores Adrenérgicos beta 2/efeitos adversos , Células Caliciformes/efeitos dos fármacos , Células Caliciformes/patologia , Corticosteroides/metabolismo , Corticosteroides/uso terapêutico , Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Asma/complicações , Asma/tratamento farmacológico , Biomarcadores , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Fluticasona/efeitos adversos , Fluticasona/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/metabolismo , Humanos , Interleucina-13/farmacologia , Metaplasia , Mucinas/genética , Mucinas/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia , Xinafoato de Salmeterol/efeitos adversos , Xinafoato de Salmeterol/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA