Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Expert Opin Drug Deliv ; 19(1): 75-86, 2022 01.
Article in English | MEDLINE | ID: mdl-35043744

ABSTRACT

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.


Subject(s)
Lymphangioleiomyomatosis , Nanoparticles , Humans , Lipids/therapeutic use , Liposomes , Lymphangioleiomyomatosis/drug therapy , Sirolimus/therapeutic use
2.
Nanomedicine (Lond) ; 15(20): 1947-1963, 2020 08.
Article in English | MEDLINE | ID: mdl-32812483

ABSTRACT

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.


Subject(s)
Lymphatic Vessels , Nanoparticles , Administration, Oral , Drug Carriers , Lipids , Lung , Particle Size , Sirolimus
3.
Drug Dev Ind Pharm ; 46(3): 427-442, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32070151

ABSTRACT

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.


Subject(s)
Cell-Penetrating Peptides/chemistry , DNA/administration & dosage , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , A549 Cells , Cell Line , Clathrin/metabolism , Emulsions , Endocytosis , Epithelial Cells , Gene Transfer Techniques , Humans , Lung/cytology , Lung/metabolism , Plasmids
4.
Eur J Pharm Sci ; 142: 105098, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31698038

ABSTRACT

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.


Subject(s)
Lipids/administration & dosage , Lymphangioleiomyomatosis/drug therapy , Nanoparticles/administration & dosage , Sirolimus/administration & dosage , Administration, Inhalation , Aerosols/administration & dosage , Animals , Bronchi/drug effects , Bronchi/metabolism , Cell Line , Humans , Lung/drug effects , Lymphangioleiomyomatosis/metabolism , Mice , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Tuberous Sclerosis/metabolism
5.
Pharm Res ; 36(1): 14, 2018 Nov 15.
Article in English | MEDLINE | ID: mdl-30443858

ABSTRACT

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.


Subject(s)
DNA/administration & dosage , Administration, Inhalation , Aerosols , Alveolar Epithelial Cells/metabolism , Biological Transport , Bronchi/metabolism , Cell Line , Cell Survival , Cell-Penetrating Peptides/chemistry , Drug Liberation , Gene Transfer Techniques , Humans , Lung/metabolism , Nebulizers and Vaporizers , Nucleic Acid Conformation , Particle Size , Plasmids , Surface Properties
6.
Pharmacol Ther ; 158: 114-24, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26713679

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/pathology , Animals , Humans , Lung/drug effects , Lung/pathology , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/metabolism , Mutation/genetics , Quality of Life , Sirolimus/pharmacology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors
7.
J Cell Mol Med ; 19(11): 2633-46, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282580

ABSTRACT

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.


Subject(s)
Doxycycline/therapeutic use , Focal Adhesion Kinase 2/drug effects , Lymphangioleiomyomatosis/etiology , Sirolimus/therapeutic use , Tuberous Sclerosis/drug therapy , rho-Associated Kinases/drug effects , Animals , Doxycycline/administration & dosage , Lymphangioleiomyomatosis/drug therapy , Mice , Rats
8.
Eur J Pharm Biopharm ; 93: 311-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25936858

ABSTRACT

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.


Subject(s)
Antifibrinolytic Agents/administration & dosage , Hemoptysis/drug therapy , Tranexamic Acid/administration & dosage , Administration, Inhalation , Aerosols , Antifibrinolytic Agents/chemistry , Cell Line , Chemistry, Pharmaceutical , Crystallography, X-Ray , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Equipment Design , Humans , Inflammation Mediators/metabolism , Microscopy, Electron, Scanning , Microscopy, Video , Nebulizers and Vaporizers , Particle Size , Powder Diffraction , Powders , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Technology, Pharmaceutical/methods , Thermogravimetry , Time Factors , Time-Lapse Imaging , Tranexamic Acid/chemistry , Wound Healing/drug effects
9.
Pulm Pharmacol Ther ; 29(2): 181-98, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24929072

ABSTRACT

Asthma and chronic obstructive pulmonary disease (COPD) are highly prevalent respiratory diseases characterized by airway inflammation, airway obstruction and airway hyperresponsiveness. Whilst current therapies, such as ß-agonists and glucocorticoids, may be effective at reducing symptoms, they do not reduce disease progression. Thus, there is a need to identify new therapeutic targets. In this review, we summarize the potential of novel targets or tools, including anti-inflammatories, phosphodiesterase inhibitors, kinase inhibitors, transient receptor potential channels, vitamin D and protease inhibitors, for the treatment of asthma and COPD.


Subject(s)
Asthma/drug therapy , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Anti-Asthmatic Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Humans , Phosphodiesterase Inhibitors/therapeutic use , Protease Inhibitors/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Transient Receptor Potential Channels/therapeutic use , Vitamin D/therapeutic use
11.
PLoS One ; 9(1): e85655, 2014.
Article in English | MEDLINE | ID: mdl-24454912

ABSTRACT

BACKGROUND: Tumstatin is a segment of the collagen-IV protein that is markedly reduced in the airways of asthmatics. Tumstatin can play an important role in the development of airway remodelling associated with asthma due to its anti-angiogenic properties. This study assessed the anti-angiogenic properties of smaller peptides derived from tumstatin, which contain the interface tumstatin uses to interact with the αVß3 integrin. METHODS: Primary human lung endothelial cells were exposed to the LF-15, T3 and T7 tumstatin-derived peptides and assessed for cell viability and tube formation in vitro. The impact of the anti-angiogenic properties on airways hyperresponsiveness (AHR) was then examined using a murine model of chronic OVA-induced allergic airways disease. RESULTS: The LF-15 and T7 peptides significantly reduced endothelial cell viability and attenuated tube formation in vitro. Mice exposed to OVA+ LF-15 or OVA+T7 also had reduced total lung vascularity and AHR was attenuated compared to mice exposed to OVA alone. T3 peptides reduced cell viability but had no effect on any other parameters. CONCLUSION: The LF-15 and T7 peptides may be appropriate candidates for use as novel pharmacotherapies due to their small size and anti-angiogenic properties observed in vitro and in vivo.


Subject(s)
Airway Remodeling/drug effects , Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Collagen Type IV/pharmacology , Peptide Fragments/pharmacology , Animals , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/physiology , Humans , Lung/blood supply , Lung/pathology , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Mitochondria/metabolism , Neovascularization, Physiologic , Primary Cell Culture
12.
Pulm Pharmacol Ther ; 26(1): 132-44, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22981423

ABSTRACT

Asthma is characterized in part by variable airflow obstruction and non-specific hyperresponsiveness to a variety of bronchoconstrictors, both of which are mediated by the airway smooth muscle (ASM). The ASM is also involved in the airway inflammation and airway wall remodeling observed in asthma. For all these reasons, the ASM provides an important target for the treatment of asthma. Several classes of drugs were developed decades ago which targeted the ASM - including ß-agonists, anti-cholinergics, anti-histamines and anti-leukotrienes - but no substantially new class of drug has appeared recently. In this review, we summarize the on-going work of several laboratories aimed at producing novel targets and/or tools for the treatment of asthma. These range from receptors and ion channels on the ASM plasmalemma, to intracellular effectors (particularly those related to cyclic nucleotide signaling, calcium-homeostasis and phosphorylation cascades), to anti-IgE therapy and outright destruction of the ASM itself.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Asthma/drug therapy , Muscle, Smooth/drug effects , Airway Obstruction/drug therapy , Airway Obstruction/pathology , Airway Remodeling/drug effects , Animals , Asthma/physiopathology , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/physiopathology , Drug Design , Humans , Inflammation/drug therapy , Inflammation/pathology , Molecular Targeted Therapy , Muscle, Smooth/metabolism
13.
Pulm Pharmacol Ther ; 26(1): 64-74, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22634303

ABSTRACT

Airway smooth muscle (ASM) is the main regulator of bronchomotor tone. Extensive studies show that in addition to their physical property, human airway smooth muscle (ASM) cells can participate in inflammatory processes modulating the initiation, perpetuation, amplification, and perhaps resolution of airway inflammation. Upon stimulation or interaction with immune cells, ASM cells produce and secrete a variety of inflammatory cytokines and chemokines, cell adhesion molecules, and extracellular matrix (ECM) proteins. These released mediators can, in turn, contribute to the inflammatory state, airway hyperresponsiveness, and airway remodeling present in asthma. As our knowledge of ASM myocyte biology improves, novel bioactive factors are emerging as potentially important regulators of inflammation. This review provides an overview of our understanding of some of these molecules, identifies rising questions, and proposes future studies to better define their role in ASM cell modulation of inflammation and immunity in the lung and respiratory diseases.


Subject(s)
Inflammation/pathology , Myocytes, Smooth Muscle/metabolism , Respiratory Tract Diseases/physiopathology , Airway Remodeling/immunology , Animals , Asthma/immunology , Asthma/physiopathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Humans , Inflammation/immunology , Inflammation Mediators/metabolism , Lung Diseases/immunology , Lung Diseases/physiopathology , Muscle, Smooth/cytology , Muscle, Smooth/immunology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/immunology , Respiratory Tract Diseases/immunology
14.
Pulm Pharmacol Ther ; 26(1): 50-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22874922

ABSTRACT

The airway smooth muscle (ASM) plays an important role in the pathophysiology of asthma and chronic obstructive pulmonary disease (COPD). ASM cells express a wide range of receptors involved in contraction, growth, matrix protein production and the secretion of cytokines and chemokines. Transforming growth factor beta (TGF-ß) is one of the major players in determining the structural and functional abnormalities of the ASM in asthma and COPD. It is increasingly evident that TGF-ß functions as a master switch, controlling a network of intracellular and autocrine signaling loops that effect ASM phenotype and function. In this review, the various elements that participate in non-canonical TGF-ß signaling, including MAPK, PI3K, WNT/ß-catenin, and Ca(2+), are discussed, focusing on their effect on ASM phenotype and function. In addition, new aspects of ASM biology and their possible association with non-canonical TGF-ß signaling will be discussed.


Subject(s)
Asthma/physiopathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Transforming Growth Factor beta/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , Humans , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Signal Transduction
15.
Am J Physiol Lung Cell Mol Physiol ; 303(8): L703-10, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22923640

ABSTRACT

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.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Integrin alpha1beta1/metabolism , Lung Neoplasms/metabolism , Lymphangioleiomyomatosis/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Leiomyoma , Lung Neoplasms/pathology , Lymphangioleiomyomatosis/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Mutant Strains , Multiprotein Complexes , Neoplasm Invasiveness/pathology , Proteins/metabolism , Rats , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , rho-Associated Kinases/metabolism
16.
J Allergy (Cairo) ; 2012: 403059, 2012.
Article in English | MEDLINE | ID: mdl-22500185

ABSTRACT

Background. Persistent asthma is characterized by airway remodeling. Whereas we have previously shown that neither ß(2)-agonists nor corticosteroids inhibit extracellular matrix (ECM) protein release from airway smooth muscle (ASM) cells, the effect of their combination is unknown and this forms the rationale for the present study. Methods. ASM cells from people with and without asthma were stimulated with TGFß1 (1 ng/ml) with or without budesonide (10(-8) M) and formoterol (10(-10) and 10(-8) M), and fibronectin expression and IL-6 release were measured by ELISA. Bronchial rings from nonasthmatic individuals were incubated with TGFß1 (1 ng/ml) with or without the drugs, and fibronectin expression was measured using immunohistochemistry. Results. Budesonide stimulated fibronectin deposition, in the presence or absence of TGFß1, and this was partially reversed by formoterol (10(-8) M) in both asthmatic and nonasthmatic cells. Budesonide and formoterol in combination failed to inhibit TGFß-induced fibronectin in either cell type. A similar pattern of expression of fibronectin was seen in bronchial rings. TGFß1-induced IL-6 release was inhibited by the combination of drugs. Conclusion. Current combination asthma therapies are unable to prevent or reverse remodeling events regulated by ASM cells.

17.
Am J Physiol Lung Cell Mol Physiol ; 302(3): L334-42, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22101762

ABSTRACT

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.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Asthma/pathology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Ethanolamines/pharmacology , Myocytes, Smooth Muscle/drug effects , RNA, Messenger/metabolism , Transcription, Genetic/drug effects , Adult , Aged , Aged, 80 and over , Budesonide/pharmacology , Case-Control Studies , Cell Adhesion Molecules/metabolism , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Female , Formoterol Fumarate , Glucocorticoids/pharmacology , Humans , Interleukin-6/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Microfilament Proteins/metabolism , Middle Aged , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/metabolism , Phosphoproteins/metabolism , Phosphorylation , Receptors, Adrenergic, beta-2/metabolism , Tachyphylaxis , Young Adult
18.
J Cell Physiol ; 227(8): 3044-52, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22015454

ABSTRACT

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.


Subject(s)
Asthma/metabolism , Gene Expression Regulation/drug effects , Interleukin-6/metabolism , Lung/metabolism , Muscle, Smooth/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adult , Aged , Aged, 80 and over , Asthma/pathology , Benzamides/pharmacology , Cells, Cultured , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Contractile Proteins/metabolism , Dioxoles/pharmacology , Female , Humans , Interleukin-6/genetics , Lung/cytology , Male , Middle Aged , Morpholines/pharmacology , Muscle, Smooth/cytology , Phosphatidylinositol 3-Kinases/genetics , Pulmonary Disease, Chronic Obstructive/pathology , Quinazolines/pharmacology , Tissue Donors , Transforming Growth Factor beta1/pharmacology
19.
J Allergy (Cairo) ; 2011: 457169, 2011.
Article in English | MEDLINE | ID: mdl-22121382

ABSTRACT

Rhinovirus-(RV-) induced asthma exacerbations account for high asthma-related health costs and morbidity in Australia. The cellular mechanism underlying this pathology is likely the result of RV-induced nuclear-factor-kappa-B-(NF-κB-) dependent inflammation. NF-κB may also be important in RV replication as inhibition of NF-κB inhibits replication of other viruses such as human immunodeficiency virus and cytomegalovirus. To establish the role of NF-κB inhibitors in RV-induced IL- 6 and IL-8 and RV replication, we used pharmacological inhibitors of NF-κB, and steroids and/or ß(2) agonists were used for comparison. Primary human lung fibroblasts were infected with RV-16 in the presence of NF-κB inhibitors: BAY-117085 and dimethyl fumarate; ß(2) agonist: salmeterol; and/or corticosteroids: dexamethasone; fluticasone. RV-induced IL-6 and IL-8 and RV replication were assessed using ELISAs and virus titration assays. RV replicated and increased IL-6 and IL-8 release. Salmeterol increased, while dexamethasone and fluticasone decreased RV-induced IL-6 and IL-8 (P<0.05). The NF-κB inhibitor BAY-117085 inhibited only RV-induced IL-6 (P<0.05) and dimethyl fumarate did not alter RV-induced IL-6 and IL-8. Dimethylfumarate increased RV replication whilst other drugs did not alter RV replication. These data suggest that inhibition of NF-κB alone is unlikely to be an effective treatment compared to current asthma therapeutics.

20.
PLoS One ; 6(5): e20000, 2011.
Article in English | MEDLINE | ID: mdl-21611147

ABSTRACT

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.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Asthma/pathology , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism , Lung/pathology , Muscle, Smooth/enzymology , Muscle, Smooth/pathology , Asthma/enzymology , Cell Proliferation/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4 , Humans , Isoproterenol/pharmacology , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Phenotype , Phosphodiesterase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...