Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Expert Opin Drug Deliv ; 19(1): 75-86, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35043744

RESUMO

INTRODUCTION: Lymphangioleiomyomatosis (LAM) is a rare lung disease that is characterized by smooth muscle-like cell growth in the lungs. The current available oral treatment rapamycin slows down the disease progression but does not result in a cure. Rapamycin is also limited by its low bioavailability and dose-related adverse side effects. New treatments are, therefore, underway to investigate alternative targets and combination therapies for LAM. In recent years, much focus has been on the development of therapies based on inhaled nanotechnology using carriers to deliver drugs, as it is shown to improve drug solubility, local targeted treatment, and bioavailability. AREAS COVERED: This review, therefore, focuses on future prospective treatments for LAM using nanoparticles and lipid-based nanocarriers, including liposomes, solid lipid nanoparticles, micelles, and polymeric nanoparticles. It also investigates how nanoparticles' physicochemical factors such as size and charge can affect the treatment of both pulmonary and extrapulmonary LAM. EXPERT OPINION: Advanced clinical research is still needed to demonstrate the full potential and drive future commercialization of LAM treatments delivered via inhaled lipid nanobased formulations. If successful, the resultant effects will be seen in the improvement in the life expectancy and life quality of LAM patients.


Assuntos
Linfangioleiomiomatose , Nanopartículas , Humanos , Lipídeos/uso terapêutico , Lipossomos , Linfangioleiomiomatose/tratamento farmacológico , Sirolimo/uso terapêutico
2.
Nanomedicine (Lond) ; 15(20): 1947-1963, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32812483

RESUMO

Aim: Lymphangioleiomyomatosis is characterized by smooth muscle-like cells in the lungs that spread to other organs via lymphatic vessels. Oral rapamycin is restricted by low bioavailability approximately 15%. The aim of the present study is to systematically investigate the effect of inhaled rapamycin solid lipid nanoparticles (Rapa-SLN) surface charge on efficacy and penetration into the lymphatics. Materials & methods: Rapa-SLN formulations with different charge: neutral, positive and negative, were produced and assessed for their physicochemical particle characteristics and efficacy in vitro. Results: Negative Rapa-SLNs were significantly faster at entering the lymphatic endothelium and more potent at inhibiting lymphanigiogenesis compared with neutral and positive Rapa-SLNs. Conclusion: Negative Rapa-SLNs showed efficient lymphatic access and should therefore be investigated further as a treatment for targeting extrapulmonary lymphangioleiomyomatosis.


Assuntos
Vasos Linfáticos , Nanopartículas , Administração Oral , Portadores de Fármacos , Lipídeos , Pulmão , Tamanho da Partícula , Sirolimo
3.
Drug Dev Ind Pharm ; 46(3): 427-442, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32070151

RESUMO

The combination of nanoparticles (NPs) and cell-penetrating peptide (CPP) represents a new opportunity to develop plasmid DNA (pDNA) delivery systems with desirable properties for lung delivery. In this study, poly(lactide-co-glycolide) (PLGA) NPs containing pDNA were formulated with and without CPP using a double-emulsion technique. NPs were characterized in regards of size, surface charge, release profile, pDNA encapsulation efficiency and pDNA integrity. Cellular uptake, intracellular trafficking, uptake mechanism and pDNA expression were assessed in both A549 and Beas-2B cells. Manufactured PLGA-NPs efficiently encapsulated pDNA with approximately 50% released in the first 24 h of incubation. Addition of CPP was essential to promote NP internalization in both cell lines, with 83.85 ± 1.2% and 96.76 ± 1.7% of Beas-2B and A549 cells, respectively, with internalized NP-DNA-CPP after 3 h of incubation. Internalization appears to occur mainly via clathrin-mediated endocytosis, with other pathways also being used by the different cell lines. An endosomal-escape mechanism seems to happen in both cell lines, and eGFP expression was observed in Beas-2B after 96 h of incubation. In summary, the NP-DNA-CPP delivery system efficiently encapsulated and protected pDNA structure and is being investigated as a promising tool for gene delivery to the lungs.


Assuntos
Peptídeos Penetradores de Células/química , DNA/administração & dosagem , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Células A549 , Linhagem Celular , Clatrina/metabolismo , Emulsões , Endocitose , Células Epiteliais , Técnicas de Transferência de Genes , Humanos , Pulmão/citologia , Pulmão/metabolismo , Plasmídeos
4.
Eur J Pharm Sci ; 142: 105098, 2020 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-31698038

RESUMO

Lymphangioleiomyomatosis (LAM) is a rare lung disease characterized by uncontrolled growth of smooth muscle -like cells in the lungs that can spread via the lymphatic system to other parts of the body. The current treatment for LAM, oral rapamycin, is limited by its low oral bioavailability and side effects. This study aims to develop an inhaled formulation of rapamycin solid lipid nanoparticles (Rapa-SLNs) to avoid first-pass metabolism, increase invivo half-life and facilitate entry into the lymphatic system through the lungs. Rapa-SLNs were manufactured using a hot evaporation technique and freeze-dried overnight with 5% (w/v) mannitol and before being assessed further for particle characteristics and in vitro aerosol performance and release. The formulation's ability to penetrate through bronchial epithelial layer was evaluated using a Calu-3 cell model, while its ability to interfere with the LAM intracellular cascade was evaluated using Mouse Embryonic fibroblast (MEF) cells deficient for the tuberous sclerosis complex 2 (TSC2) and compared with rapamycin solution. Results showed that the Rapa- SLNs had the appropriate size (237.5 ±â€¯1.8 nm), charge (-11.2), in vitro aerosol performance (MMAD=5.4 ±â€¯0.4 µm) and sustained release profile suitable for entry into the lymphatic system via the pulmonary route. Additionally, the nanoparticles were transported at a faster rate across the bronchial epithelial layer compared to free rapamycin solution. The formulation also showed similar mTOR (mammalian target of Rapamycin) inhibition properties compared to free rapamycin, and was able to significantly decrease the amount of proliferation in TSC2 negative MEF cells. This formulation is therefore a promising alternative treatment for LAM patients, as it could potentially reduce problems associated with low bioavailability and side effects of current oral treatment.


Assuntos
Lipídeos/administração & dosagem , Linfangioleiomiomatose/tratamento farmacológico , Nanopartículas/administração & dosagem , Sirolimo/administração & dosagem , Administração por Inalação , Aerossóis/administração & dosagem , Animais , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Linhagem Celular , Humanos , Pulmão/efeitos dos fármacos , Linfangioleiomiomatose/metabolismo , Camundongos , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Esclerose Tuberosa/metabolismo
5.
Pharm Res ; 36(1): 14, 2018 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-30443858

RESUMO

PURPOSE: In this study, a cell penetrating peptide was used as an uptake enhancer for pDNA delivery to the lungs. METHODS: Polyplexes were prepared between pDNA and CPP. Intracellular delivery of pDNA was assessed in both alveolar (A549) and bronchial (Calu-3) epithelial cells. Aerosol delivery was investigated using a mesh nebulizer. RESULTS: Efficient intracellular delivery of pDNA occurs in both A549 and Calu-3 cells when delivered as polyplexes. Protection against nucleases and endosomal escape mechanism occurs when pDNA is formulated within the polyplexes. For aerosol delivery, 1% (w/v) mannitol was able to protect naked DNA structure during nebulization with a significant increase in fine particle fraction (particles <5 µm). The structure of polyplexes when delivered via a mesh nebulizer using 1% (w/v) mannitol could partially withstand the shear forces involved in aerosolization. Although some loss in functionality occurred after nebulization, membrane-associated fluorescence was observed in A549 cells. In Calu-3 cells mucus entrapment was a limiting factor for polyplex delivery. CONCLUSIONS: The presence of CPP is essential for efficient intracellular delivery of pDNA. The polyplexes can be delivered to lung epithelial cells using mesh nebulizer. The use of different excipients is essential for further optimization of these delivery systems.


Assuntos
DNA/administração & dosagem , Administração por Inalação , Aerossóis , Células Epiteliais Alveolares/metabolismo , Transporte Biológico , Brônquios/metabolismo , Linhagem Celular , Sobrevivência Celular , Peptídeos Penetradores de Células/química , Liberação Controlada de Fármacos , Técnicas de Transferência de Genes , Humanos , Pulmão/metabolismo , Nebulizadores e Vaporizadores , Conformação de Ácido Nucleico , Tamanho da Partícula , Plasmídeos , Propriedades de Superfície
6.
Pharmacol Ther ; 158: 114-24, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26713679

RESUMO

Lymphangioleiomyomatosis (LAM) is a rare neoplastic disease affecting predominantly young women. Clinical symptoms of this progressive disease include dyspnoea, cough, recurrent pneumothorax, hemoptysis and chylothorax. LAM is generally aggressive in nature and ultimately results in respiratory failure. Important hallmark features of this metastatic disease include the formation of lesions of abnormal smooth muscle cells, cystic destruction of the lung tissue and lymphangiogenesis affecting the lungs, abdomen and lymphatics. Research over the last 10-15 years has significantly enhanced our understanding of the molecular and cellular processes associated with LAM. These processes include mutational inactivation of the tuberous sclerosis complex genes, TSC1 and TSC2, activation of the mammalian target of rapamycin (mTOR) pathway, enhanced cell proliferation and migration, lymphangiogenesis, metastatic spread through the blood and lymphatic circulations, sex steroid sensitivity and dysregulated autophagy. Despite this increased knowledge there is currently no cure for LAM and treatment options remain limited. Whilst the mTOR inhibitor rapamycin has shown some benefit in patients with LAM, with stabilisation of lung function and improved quality of life, cessation of treatment results in recurrence of the disease progression. This highlights the urgent need to identify novel targets and new treatment regimens. The focus of this review is to summarise our current understanding of the cellular and molecular processes associated with LAM and highlight emerging treatments.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linfangioleiomiomatose/tratamento farmacológico , Linfangioleiomiomatose/patologia , Animais , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Linfangioleiomiomatose/genética , Linfangioleiomiomatose/metabolismo , Mutação/genética , Qualidade de Vida , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores
7.
J Cell Mol Med ; 19(11): 2633-46, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26282580

RESUMO

Lymphangioleiomyomatosis (LAM) is associated with dysfunction of the tuberous sclerosis complex (TSC) leading to enhanced cell proliferation and migration. This study aims to examine whether doxycycline, a tetracycline antibiotic, can inhibit the enhanced migration of TSC2-deficient cells, identify signalling pathways through which doxycycline works and to assess the effectiveness of combining doxycycline with rapamycin (mammalian target of rapamycin complex 1 inhibitor) in controlling cell migration, proliferation and wound closure. TSC2-positive and TSC2-negative mouse embryonic fibroblasts (MEF), 323-TSC2-positive and 323-TSC2-null MEF and Eker rat uterine leiomyoma (ELT3) cells were treated with doxycycline or rapamycin alone, or in combination. Migration, wound closure and proliferation were assessed using a transwell migration assay, time-lapse microscopy and manual cell counts respectively. RhoA-GTPase activity, phosphorylation of p70S6 kinase (p70S6K) and focal adhesion kinase (FAK) in TSC2-negative MEF treated with doxycycline were examined using ELISA and immunoblotting techniques. The enhanced migration of TSC2-null cells was reduced by doxycycline at concentrations as low as 20 pM, while the rate of wound closure was reduced at 2-59 µM. Doxycycline decreased RhoA-GTPase activity and phosphorylation of FAK in these cells but had no effect on the phosphorylation of p70S6K, ERK1/2 or AKT. Combining doxycycline with rapamycin significantly reduced the rate of wound closure at lower concentrations than achieved with either drug alone. This study shows that doxycycline inhibits TSC2-null cell migration. Thus doxycycline has potential as an anti-migratory agent in the treatment of diseases with TSC2 dysfunction.


Assuntos
Doxiciclina/uso terapêutico , Quinase 2 de Adesão Focal/efeitos dos fármacos , Linfangioleiomiomatose/etiologia , Sirolimo/uso terapêutico , Esclerose Tuberosa/tratamento farmacológico , Quinases Associadas a rho/efeitos dos fármacos , Animais , Doxiciclina/administração & dosagem , Linfangioleiomiomatose/tratamento farmacológico , Camundongos , Ratos
8.
Eur J Pharm Biopharm ; 93: 311-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25936858

RESUMO

PURPOSE: An inhalable dry powder formulation of tranexamic acid (TA) was developed and tested in a novel high-dose Orbital® multi-breath inhaler. The formulation was specifically intended for the treatment of pulmonary haemorrhage and wound healing associated with haemoptysis. METHODS: Inhalable TA particles were prepared by spray drying and the powder characterised using laser diffraction, electron microscopy, thermal analysis, moisture sorption and X-ray powder diffraction. The aerosol performance was evaluated using cascade impaction and inline laser diffraction and interaction with epithelia cells and wound healing capacity investigated using Calu-3 air interface model. RESULTS: The spray dried TA particles were crystalline and spherical with a D0.5 of 3.35 µm. The powders were stable and had limited moisture sorption (0.307%w/w at 90%RH). The Orbital device delivered ca. 38 mg powder per 'inhalation' at 60 l · min(-1) across four sequential shots with an overall fine particle fraction (⩽ 6.4 µm) of 59.3 ± 3.5% based on the emitted mass of ca. 150 mg. The TA particles were well tolerated by Calu-3 bronchial epithelia cells across a wide range of doses (from 1 nM to 10nM) and no increase in inflammatory mediators was observed after deposition of the particles (a decrease in IL-1ß, IL-8 and INFγ was observed). Time lapse microscopy of a damaged confluent epithelia indicated that wound closure was significantly greater in TA treated cells compared to control. CONCLUSION: A stable, high performance aerosol of TA has been developed in a multi-breath DPI device that can be used for the treatment of pulmonary lesions and haemoptysis.


Assuntos
Antifibrinolíticos/administração & dosagem , Hemoptise/tratamento farmacológico , Ácido Tranexâmico/administração & dosagem , Administração por Inalação , Aerossóis , Antifibrinolíticos/química , Linhagem Celular , Química Farmacêutica , Cristalografia por Raios X , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Desenho de Equipamento , Humanos , Mediadores da Inflamação/metabolismo , Microscopia Eletrônica de Varredura , Microscopia de Vídeo , Nebulizadores e Vaporizadores , Tamanho da Partícula , Difração de Pó , Pós , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Tecnologia Farmacêutica/métodos , Termogravimetria , Fatores de Tempo , Imagem com Lapso de Tempo , Ácido Tranexâmico/química , Cicatrização/efeitos dos fármacos
10.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L703-10, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22923640

RESUMO

Recent evidence suggests that the rare and progressive lung disease lymphangioleiomyomatosis (LAM) is metastatic in nature. Dysfunction of the tumor suppressor genes tuberous sclerosis complex (TSC), in particular mutational inactivation of TSC2, enhances both cell proliferation and migration. Although substantial progress has been made in understanding the role of TSC2 in abnormal LAM cell proliferation and its pharmacological targeting, the mechanisms underlying the enhanced migratory capacity in LAM are not well understood. In this study, we examined the role of TSC2 in cell attachment, spreading, and migration, processes that contribute to the metastatic phenotype. Here we show that loss of TSC2 increased both the attachment and spreading of mouse embryonic fibroblasts to the extracellular matrix proteins collagen type I and fibronectin and that reexpression of TSC2 reduced these effects. Integrin-α1ß1 modulated cell migration with the ß1-subunit involved in cell attachment and spreading as shown by using functional blocking antibodies. Loss of TSC2 increased integrin-α1 expression, and inhibition of this integrin subunit reduced cell migration. The enhanced attachment and spreading were independent of the intracellular signaling pathways mammalian target of rapamycin complex 1 and Rho-associated kinase, as pharmacological inhibition with rapamycin or Y27632, respectively, was without effect. Together, these data demonstrate that TSC2 controls cell migration, attachment, and spreading through the α1ß1-integrin receptor and thus suggest a potential therapeutic target for the treatment of increased cell invasiveness in LAM.


Assuntos
Adesão Celular/fisiologia , Movimento Celular/fisiologia , Integrina alfa1beta1/metabolismo , Neoplasias Pulmonares/metabolismo , Linfangioleiomiomatose/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibroblastos/citologia , Leiomioma , Neoplasias Pulmonares/patologia , Linfangioleiomiomatose/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Mutantes , Complexos Multiproteicos , Invasividade Neoplásica/patologia , Proteínas/metabolismo , Ratos , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR , Proteína 2 do Complexo Esclerose Tuberosa , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/genética , Quinases Associadas a rho/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 302(3): L334-42, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22101762

RESUMO

ß(2)-Adrenergic receptor (ß2AR) agonists induce airway relaxation via cAMP. Phosphodiesterase (PDE)s degrade and regulate cAMP, and in airway smooth muscle (ASM) cells PDE4D degrades cAMP. Long-acting ß(2)-agonists are now contraindicated as monotherapy for asthma, and increased PDE4D has been speculated to contribute to this phenomenon. In this study we investigated the expression of PDE4D in asthmatic and nonasthmatic ASM cells and its regulation by formoterol and budesonide. Primary ASM cells from people with or without asthma were stimulated with transforming growth factor (TGF)-ß(1), formoterol, and/or budesonide. PDE4D mRNA was assessed by real-time PCR, or PCR to assess splice variant production. PDE4D protein was assessed by Western blotting, and we investigated the effect of formoterol on cAMP production and PDE activity. Interleukin (IL)-6 was assessed using ELISA. PDE4D mRNA was dose dependently upregulated by formoterol, with a single splice variant, PDE4D5, present. Formoterol did not induce PDE4D protein at time points between 3 to 72 h, whereas it did induce and increase IL-6 secretion. We pretreated cells with actinomycin D and a proteasome inhibitor, MG132, and found no evidence of alterations in mRNA, protein expression, or degradation of PDE4D. Finally PDE activity was not altered by formoterol. This study shows, for the first time, that PDE4D5 is predominantly expressed in human ASM cells from people with and without asthma and that formoterol does not upregulate PDE4D protein production. This leads us to speculate that continual therapy with ß2AR agonists is unlikely to cause PDE4-mediated tachyphylaxis.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Asma/patologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Etanolaminas/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos , Adulto , Idoso , Idoso de 80 Anos ou mais , Budesonida/farmacologia , Estudos de Casos e Controles , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/genética , Feminino , Fumarato de Formoterol , Glucocorticoides/farmacologia , Humanos , Interleucina-6/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Receptores Adrenérgicos beta 2/metabolismo , Taquifilaxia , Adulto Jovem
12.
J Cell Physiol ; 227(8): 3044-52, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22015454

RESUMO

Transforming growth factor (TGF) ß1 increases pro-inflammatory cytokines and contractile protein expression by human airway smooth muscle (ASM) cells, which could augment airway inflammation and hyperresponsiveness. Phosphoinositide 3' kinase (PI3K) is one of the signaling pathways implicated in TGFß1 stimulation, and may be altered in asthmatic airways. This study compared the expression of PI3K isoforms by ASM cells from donors with asthma (A), chronic obstructive pulmonary disease (COPD), or neither disease (NA), and investigated the role of PI3K isoforms in the production of TGFß1 induced pro-inflammatory cytokine and contractile proteins in ASM cells. A cells expressed higher basal levels of p110δ mRNA compared to NA and COPD cells; however COPD cells produced more p110δ protein. TGFß1 increased 110δ mRNA expression to the same extent in the three groups. Neither the p110δ inhibitor IC87114 (1, 10, 30 µM), the p110ß inhibitor TGX221 (0.1, 1, 10 µM) nor the PI3K pan inhibitor LY294002 (3, 10 µM) had any effect on basal IL-6, calponin or smooth muscle α-actin (α-SMA) expression. However, TGFß1 increased calponin and α-SMA expression was inhibited by IC87114 and LY294002 in all three groups. IC87114, TGX221, and LY294002 reduced TGFß1 induced IL-6 release in a dose related manner in all groups of ASM cells. PI3K p110δ is important for TGFß1 induced production of the contractile proteins calponin and α-SMA and the proinflammatory cytokine IL-6 in ASM cells, and may therefore be relevant as a potential therapeutic target to treat both inflammation and airway remodeling.


Assuntos
Asma/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-6/metabolismo , Pulmão/metabolismo , Músculo Liso/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Asma/patologia , Benzamidas/farmacologia , Células Cultivadas , Cromonas/farmacologia , Classe I de Fosfatidilinositol 3-Quinases , Proteínas Contráteis/metabolismo , Dioxóis/farmacologia , Feminino , Humanos , Interleucina-6/genética , Pulmão/citologia , Masculino , Pessoa de Meia-Idade , Morfolinas/farmacologia , Músculo Liso/citologia , Fosfatidilinositol 3-Quinases/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Quinazolinas/farmacologia , Doadores de Tecidos , Fator de Crescimento Transformador beta1/farmacologia
13.
PLoS One ; 6(5): e20000, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21611147

RESUMO

BACKGROUND AND OBJECTIVE: Asthma is associated with airway narrowing in response to bronchoconstricting stimuli and increased airway smooth muscle (ASM) mass. In addition, some studies have suggested impaired ß-agonist induced ASM relaxation in asthmatics, but the mechanism is not known. OBJECTIVE: To characterize the potential defect in ß-agonist induced cAMP in ASM derived from asthmatic in comparison to non-asthmatic subjects and to investigate its mechanism. METHODS: We examined ß(2)-adrenergic (ß(2)AR) receptor expression and basal ß-agonist and forskolin (direct activator of adenylyl cyclase) stimulated cAMP production in asthmatic cultured ASM (n = 15) and non-asthmatic ASM (n = 22). Based on these results, PDE activity, PDE4D expression and cell proliferation were determined. RESULTS: In the presence of IBMX, a pan PDE inhibitor, asthmatic ASM had ∼50% lower cAMP production in response to isoproterenol, albuterol, formoterol, and forskolin compared to non-asthmatic ASM. However when PDE4 was specifically inhibited, cAMP production by the agonists and forskolin was normalized in asthmatic ASM. We then measured the amount and activity of PDE4, and found ∼2-fold greater expression and activity in asthmatic ASM compared to non-asthmatic ASM. Furthermore, inhibition of PDE4 reduced asthmatic ASM proliferation but not that of non-asthmatic ASM. CONCLUSION: Decreased ß-agonist induced cAMP in ASM from asthmatics results from enhanced degradation due to increased PDE4D expression. Clinical manifestations of this dysregulation would be suboptimal ß-agonist-mediated bronchodilation and possibly reduced control over increasing ASM mass. These phenotypes appear to be "hard-wired" into ASM from asthmatics, as they do not require an inflammatory environment in culture to be observed.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Asma/patologia , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Pulmão/patologia , Músculo Liso/enzimologia , Músculo Liso/patologia , Asma/enzimologia , Proliferação de Células/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Humanos , Isoproterenol/farmacologia , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Fenótipo , Inibidores de Fosfodiesterase/farmacologia
14.
J Pharmacol Exp Ther ; 337(2): 557-66, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21349933

RESUMO

The phosphatidylinositol 3-kinase (PI3K) signal transduction pathway is implicated in the airway remodeling associated with asthma. The class IA PI3K isoforms are known to be activated by growth factors and cytokines. Because this pathway is a possible site of pharmacological intervention for treating the disease, it is important to know which isoforms contribute to this process. Therefore, we used a pharmacological approach to investigate the roles of the three class IA PI3K isoforms (p110α, p110ß, and p110δ) in airway remodeling using airway smooth muscle (ASM) cells derived from asthmatic subjects and ASM cells and lung fibroblasts from nonasthmatic subjects. These studies used the inhibitors N'-[(E)-(6-bromoimidazo[1,2-a]pyridin-3-yl)methylidene]-N,2-dimethyl-5-nitrobenzenesulfonohydrazide (PIK75) (which selectively inhibits p110α), 7-methyl-2-(4-morpholinyl)-9-[1-(phenylamino)ethyl]-4H-pyrido[1,2-a]pyrimidin-4-one (TGX221) (which selectively inhibits p110ß), and 2-[(6-amino-9H-purin-9-yl)methyl]-5-methyl-3-(2-methylphenyl)-4(3H)-quinazolinone (IC87114) (which selectively inhibits p110δ). Cells were stimulated with transforming growth factor-ß (TGFß) and/or 10% fetal bovine serum in the presence or absence of inhibitor or vehicle control (dimethyl sulfoxide). PIK75, but not TGX221 or IC87114, attenuated TGFß-induced fibronectin deposition in all cell types tested. PIK75 and TGX221 each decreased secretion of vascular endothelial growth factor and interleukin-6 in nonasthmatic ASM cells and lung fibroblasts, whereas TGX221 was not as effective in asthmatic ASM cells. In addition, PIK75 decreased cell survival in TGFß-stimulated asthmatic, but not nonasthmatic, ASM cells. In conclusion, specific PI3K isoforms may play a role in pathophysiological events relevant to airway wall remodeling.


Assuntos
Células-Tronco Mesenquimais/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Sistema Respiratório/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Western Blotting , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/enzimologia , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Humanos , Hidrazonas/farmacologia , Interleucina-6/metabolismo , Isoenzimas/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Morfolinas/farmacologia , Músculo Liso/citologia , Músculo Liso/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Pirimidinonas/farmacologia , Quinazolinas/farmacologia , Sistema Respiratório/citologia , Especificidade por Substrato , Sulfonamidas/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
J Cell Physiol ; 225(3): 846-54, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20607798

RESUMO

Human bronchial epithelial (HBE) cells contribute to asthmatic airway inflammation by secreting cytokines, chemokines, and growth factors, including interleukin (IL)-6, IL-8 and transforming growth factor (TGF) ß1, all of which are elevated in asthmatic airways. This study examines the signaling pathways leading to TGFß1 induced IL-6 and IL-8 in primary HBE cells from asthmatic and non-asthmatic volunteers. HBE cells were stimulated with TGFß1 in the presence or absence of signaling inhibitors. IL-6 and IL-8 protein and mRNA were measured by ELISA and real-time PCR respectively, and cell signaling kinases by Western blot. TGFß1 increased IL-6, but inhibited IL-8 production in both asthmatic and non-asthmatic cells; however, TGF induced significantly more IL-6 in asthmatic cells. Inhibition of JNK MAP kinase partially reduced TGFß1 induced IL-6 in both cell groups. TGFß1 induced Smad2 phosphorylation, and blockade of Smad2/3 prevented both the TGFß1 modulated IL-6 increase and the decrease in IL-8 production in asthmatic and non-asthmatic cells. Inhibition of Smad2/3 also increased basal IL-8 release in asthmatic cells but not in non-asthmatic cells. Using CHIP assays we demonstrated that activated Smad2 bound to the IL-6, but not the IL-8 promoter region. We conclude that the Smad2/3 pathway is the predominant TGFß1 signaling pathway in HBE cells, and this is altered in asthmatic bronchial epithelial cells. Understanding the mechanism of aberrant pro-inflammatory cytokine production in asthmatic airways will allow the development of alternative ways to control airway inflammation.


Assuntos
Asma/imunologia , Brônquios/imunologia , Células Epiteliais/imunologia , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Mucosa Respiratória/imunologia , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Adolescente , Adulto , Idoso , Asma/metabolismo , Sítios de Ligação , Western Blotting , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Imunoprecipitação da Cromatina , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Interleucina-6/genética , Interleucina-8/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Pessoa de Meia-Idade , Fosforilação , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Transdução de Sinais , Proteína Smad2/antagonistas & inibidores , Proteína Smad3/antagonistas & inibidores , Regulação para Cima , Adulto Jovem
16.
Respirology ; 15(2): 303-12, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20070588

RESUMO

UNLABELLED: PPARgamma levels in asthma- and non-asthma-derived airway smooth muscle cells and PPARgamma activation-induced cell proliferation were investigated. In the presence of FBS, PPARgamma levels were higher in subconfluent asthma-derived cells but lower in confluent cells compared with non-asthma-derived. However, PPARgamma activation did not alter cell proliferation. BACKGROUND AND OBJECTIVE: Airway remodelling involves thickening of the airway smooth muscle (ASM) bulk. Proliferation of asthma-derived ASM cells is increased in vitro, but underlying mechanisms remain unknown. Peroxisome proliferators activated receptor-gamma (PPARgamma) regulates the cell cycle. It is suggested that PPARgamma agonists have anti-inflammatory effects, which may be valuable in the treatment of asthma, but information regarding their antiproliferative properties in ASM is lacking. Although corticosteroids reduce airway inflammation, in vitro they inhibit proliferation in only non-asthma ASM cells by reducing cyclin D1. We therefore investigated the effects of mitogenic stimulation (foetal bovine serum (FBS)), and a PPARgamma ligand (ciglitazone), on PPARgamma and cyclin D1 expression and proliferation of ASM cells. In addition, we examined the effects of ciglitazone on ASM cell proliferation. METHODS: We assessed PPARgamma and cyclin D1 mRNA and protein levels using quantitative PCR and immunoblotting. Cell proliferation was assessed using bromodeoxyuridine uptake. RESULTS: In the presence of 5% FBS, PPARgamma and cyclin D1 expression decreased over time in non-asthmatic cells but increased in asthmatic cells (compared with sub-confluent cells). FBS-induced proliferation of asthmatic cells increased at all time points, but occurred only at day 7 with non-asthmatic cells (compared with unstimulated time-matched control). Ciglitazone increased PPARgamma expression in both groups, but did not alter cell proliferation, while fluticasone increased PPARgamma protein only in asthmatic cells. CONCLUSIONS: Although in the presence of a mitogenic stimulus, PPARgamma was differentially expressed in asthma- and non-asthma-derived ASM; its expression was not related to the increased proliferation observed in asthmatic ASM.


Assuntos
Asma/metabolismo , Asma/patologia , Proliferação de Células , Ciclina D1/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , PPAR gama/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Androstadienos/farmacologia , Brônquios/patologia , Broncodilatadores/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Fluticasona , Humanos , Masculino , Pessoa de Meia-Idade , Mitógenos/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , RNA Mensageiro/metabolismo , Tiazolidinedionas/farmacologia , Adulto Jovem
17.
Am J Respir Cell Mol Biol ; 43(2): 227-33, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19783788

RESUMO

Rhinovirus (RV) infections are the major cause of asthma exacerbations in children and adults. Under normal circumstances, asthmatic airway obstruction improves spontaneously or characteristically briskly in response to inhaled beta(2)-adrenergic receptor (beta(2)AR) agonists. During virus-associated exacerbations, an impaired response to beta(2)AR agonists is observed; the reason for this is not known. The objective of this study was to determine the effect of RV infection on airway smooth muscle beta(2)AR function. The human cell line Beas-2B and primary human bronchial epithelial cells (HBECs) were infected with RV (multiplicity of infection = 1). After 1 or 5 days for primary and Beas-2B cells, respectively, cell culture supernatants were harvested, UV-irradiated to inactivate RV, and applied to human airway smooth muscle cells for 3 days to assess modifications of beta(2)AR function. RV conditioned medium from Beas-2B and HBECs decreased beta(2)AR agonist-induced cAMP by 50 and 65%, respectively (n = 5; P < 0.05). When cAMP was induced independently of the beta(2)AR using forskolin, no impairment was found. Using flow cytometry, we demonstrated that this decrease was likely the result of beta(2)AR desensitization because membrane but not total cell receptor beta(2)AR was decreased. Pretreatment of HBECs and Beas-2B cells but not human airway smooth muscle cells with the corticosteroids dexamethasone or fluticasone abolished virus-mediated beta(2)AR loss of function. This study shows that epithelial infection with RV induces a decrease of beta(2)AR function on airway smooth muscle cells, potentially explaining the clinical observation of loss of beta(2)AR agonist function during RV-induced asthma exacerbations.


Assuntos
Asma/complicações , Asma/virologia , Receptores Adrenérgicos beta 2/metabolismo , Rhinovirus/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Brônquios/patologia , Meios de Cultivo Condicionados/metabolismo , Células Epiteliais/citologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , Infecções por Picornaviridae/complicações , Infecções por Picornaviridae/virologia
18.
Am J Respir Crit Care Med ; 181(2): 106-15, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19875687

RESUMO

RATIONALE: Angiogenesis is a prominent feature of remodeling in asthma. Many proangiogenic factors are up-regulated in asthma, but little is known about levels of endogenous antiangiogenic agents. Collagen IV is decreased in the airway basement membrane in asthma. It has six alpha chains, of which the noncollagenous domain-1 domains have endogenous antiangiogenic properties. OBJECTIVES: To study the expression of the noncollagenous domain-1 of the alpha3 chain of collagen IV, tumstatin, in the airways of subjects with and without asthma and to examine the potential for tumstatin to regulate angiogenesis and inflammation. METHODS: We used immunohistochemistry and dot blots to examine the expression of tumstatin in bronchial biopsies, bronchoalveolar lavage fluid, and serum. We then used an in vitro angiogenesis assay and a murine model of allergic airways disease to explore tumstatin's biological function. MEASUREMENTS AND MAIN RESULTS: The level of tumstatin is decreased 18-fold in the airways of patients with asthma but not in subjects without asthma, including those with chronic obstructive pulmonary disease, cystic fibrosis, and bronchiectasis. In vitro, recombinant tumstatin inhibited primary pulmonary endothelial cell tube formation. In a mouse model of chronic allergic airways disease, tumstatin suppressed angiogenesis, airway hyperresponsiveness, inflammatory cell infiltration, and mucus secretion and decreased levels of vascular endothelial growth factor and IL-13. CONCLUSIONS: The observation that tumstatin is decreased in asthmatic airways and inhibits airway hyperresponsiveness and angiogenesis demonstrates the potential use of antiangiogenic agents such as tumstatin as a therapeutic intervention in diseases that are characterized by aberrant angiogenesis and tissue remodeling, such as asthma.


Assuntos
Asma/fisiopatologia , Autoantígenos/fisiologia , Brônquios/irrigação sanguínea , Hiper-Reatividade Brônquica/fisiopatologia , Bronquite/fisiopatologia , Colágeno Tipo IV/fisiologia , Neovascularização Patológica/fisiopatologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Remodelação das Vias Aéreas/fisiologia , Animais , Asma/patologia , Biópsia , Brônquios/patologia , Hiper-Reatividade Brônquica/patologia , Bronquite/patologia , Broncoscopia , Divisão Celular/fisiologia , Colágeno Tipo IV/metabolismo , Modelos Animais de Doenças , Células Endoteliais/patologia , Células Endoteliais/fisiologia , Eosinofilia/patologia , Eosinofilia/fisiopatologia , Feminino , Humanos , Interleucina-13/metabolismo , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Camundongos , Microscopia de Fluorescência , Pessoa de Meia-Idade , Neovascularização Patológica/patologia , Hipersensibilidade Respiratória/patologia , Hipersensibilidade Respiratória/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
19.
Am J Physiol Lung Cell Mol Physiol ; 293(4): L982-90, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17616643

RESUMO

It has been proposed that a hypoxia-induced inhibition of the Na(+)-Ca(2+) exchanger (NCX) contributes to hypoxic pulmonary vasoconstriction (HPV). By recording isometric tension development in rat intrapulmonary arteries (IPA), we examined the effect on HPV of maneuvers that reduce the ability of NCX to regulate intracellular Ca(2+) concentration ([Ca(2+)](i)). In some experiments, fura pentakis(acetoxymethyl) ester-3 (fura PE-3) was also used to monitor [Ca(2+)](i). HPV was elicited in IPA that were pretreated with 10 microM diltiazem and slightly preconstricted with PGF(2alpha), which enhances the hypoxic response. Substitution of Na(+) with Li(+) increased HPV and the associated rise in [Ca(2+)](i). Pretreatment with ouabain (100 microM) to diminish the Na(+) gradient or with the reverse-mode NCX inhibitor KB-R7943 (3 or 10 microM) had no significant effect on HPV. Combined treatment with ouabain and low-[Na(+)] (24 mM) solution enhanced HPV strongly. The role of NCX in Ca(2+) extrusion was examined by assessing the decrease in [Ca(2+)](i) in Ca(2+)-free physiological saline solution either containing or lacking Na(+) following a high K(+)-induced loading of cellular [Ca(2+)]. Although the large initial rapid fall in [Ca(2+)] was Na(+) independent, final recovery of [Ca(2+)] to its basal level was delayed in the absence of Na(+). Therefore, HPV persisted or was increased under conditions in which forward-mode NCX was already attenuated or prevented, demonstrating that inhibition of NCX by hypoxia is unlikely to initiate HPV. Instead, NCX appears to act to inhibit HPV as would be expected if it is functioning to extrude Ca(2+).


Assuntos
Hipóxia/fisiopatologia , Artéria Pulmonar/fisiopatologia , Trocador de Sódio e Cálcio/metabolismo , Vasoconstrição , Animais , Cálcio/metabolismo , Membranas Intracelulares/metabolismo , Contração Isométrica , Lítio/farmacologia , Masculino , Concentração Osmolar , Potássio/farmacologia , Artéria Pulmonar/metabolismo , Ratos , Ratos Wistar , Sódio/farmacologia , Vasoconstrição/efeitos dos fármacos
20.
Am J Respir Cell Mol Biol ; 36(6): 721-7, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17272821

RESUMO

Airway hyperresponsiveness (AHR) is associated with airway wall structural remodeling and alterations in airway smooth muscle (ASM) function. Previously, in bronchioles from Brown Norway rats challenged by repeated ovalbumin (OVA) inhalation, we have reported increased force generation and depletion of smooth muscle contractile proteins. Here, we investigated if cytoskeletal changes in smooth muscle could account for this paradox. Sensitized rats (n = 5/group) were repeatedly challenged with OVA or saline, and the lungs were removed 24 h after the last challenge. Levels of globular (G) and filamentous (F) actin in bronchioles were determined by DNase I inhibition and contraction assessed in intact small bronchioles using a myograph. DNase I inhibition assays showed that G-actin monomers were more abundant ( approximately 1F:2G) in extracts from resting small bronchioles from OVA- or saline-challenged animals. However, while contractile protein levels in bronchioles were reduced by OVA (P < 0.05), the proportion of F:G actin was 1.8-fold greater compared with saline challenge (P < 0.05). Consistent with induction of F-actin after OVA challenge, increases in maximum tension development to carbachol or KCl in small bronchioles from OVA-challenged animals were abrogated (P < 0.01) by actin cytoskeleton disruption with 0.5 microM latrunculin A. Cytoskeletal stabilization of F-actin with 0.1 microM jasplakinolide potentiated maximum contractions to carbachol or KCl (P < 0.05) in bronchioles from OVA- but not saline-treated rats. We conclude that alterations in the composition and/or arrangement of the contractile apparatus after OVA exposure confer enhanced contractile responses, possibly as a result of increased F-actin content. Such a mechanism may have relevance for AHR found in allergic asthma.


Assuntos
Alérgenos/imunologia , Brônquios/citologia , Brônquios/imunologia , Citoesqueleto/metabolismo , Músculo Liso/metabolismo , Actinas/metabolismo , Animais , Antineoplásicos/farmacologia , Asma/imunologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Brônquios/efeitos dos fármacos , Carbacol/farmacologia , Células Cultivadas , Citoesqueleto/efeitos dos fármacos , Depsipeptídeos/farmacologia , Exposição por Inalação , Masculino , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Liso/citologia , Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Ovalbumina/administração & dosagem , Ovalbumina/imunologia , Ratos , Tiazolidinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA