Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Res Social Adm Pharm ; 20(6): 165-169, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38438294

ABSTRACT

The effective provision of professional pharmacy services is critical to support the delivery of primary health care. Structured frameworks and theoretical strategies are required to facilitate successful service implementation processes, outcomes and sustainability. This commentary discusses the considerations of what framework (adoption versus adaptation) would be suitable when implementing a new professional pharmacy service to a new environment. Utilizing Minor Ailments Services (MASs) as an exemplar as a professional pharmacy service case study, the research that underpinned these considerations enabled the development of a sequential, phased framework. There is the potential to utilize this framework for future evolving professional pharmacy services in the new setting.


Subject(s)
Pharmaceutical Services , Humans , Pharmaceutical Services/organization & administration , Primary Health Care/organization & administration , Pharmacists/organization & administration , Delivery of Health Care/organization & administration
2.
Front Cell Neurosci ; 17: 1156084, 2023.
Article in English | MEDLINE | ID: mdl-37124398

ABSTRACT

Introduction: Glaucoma is a progressive neurodegenerative disease associated with age. Accumulation of amyloid-beta (Aß) proteins in the ganglion cell layer (GCL) and subsequent retinal ganglion cell (RGC) loss is an established pathological hallmark of the disease. The mechanism through which Aß provokes RGC loss remains unclear. The receptor for the advanced glycation end product (RAGE), and its ligand Aß, have been shown to mediate neuronal loss via internalizing Aß within the neurons. In this study, we investigated whether the RAGE-Aß axis plays a role in RGC loss in experimental glaucoma. Methods: Retinal ischemia was induced by an acute elevation of intraocular pressure in RAGE-/- and wild-type (WT) control mice. In a subset of animals, oligomeric Aß was injected directly into the vitreous of both strains. RGC loss was assessed using histology and biochemical assays. Baseline and terminal positive scotopic threshold (pSTR) were also recorded. Results: Retinal ischemia resulted in 1.9-fold higher RGC loss in WT mice compared to RAGE-/- mice (36 ± 3% p < 0.0001 vs. 19 ± 2%, p = 0.004). Intravitreal injection of oligomeric Aß resulted in 2.3-fold greater RGC loss in WT mice compared to RAGE-/- mice, 7-days post-injection (55 ± 4% p = 0.008 vs. 24 ± 2%, p = 0.02). We also found a significant decline in the positive scotopic threshold response (pSTR) amplitude of WT mice compared to RAGE-/- (36 ± 3% vs. 16 ± 6%). Discussion: RAGE-/- mice are protected against RGC loss following retinal ischemia. Intravitreal injection of oligomeric Aß accelerated RGC loss in WT mice but not RAGE-/-. A co-localization of RAGE and Aß, suggests that RAGE-Aß binding may contribute to RGC loss.

3.
Thorax ; 78(7): 661-673, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36344253

ABSTRACT

BACKGROUND: Severe neutrophilic asthma is resistant to treatment with glucocorticoids. The immunomodulatory protein macrophage migration inhibitory factor (MIF) promotes neutrophil recruitment to the lung and antagonises responses to glucocorticoids. We hypothesised that MIF promotes glucocorticoid resistance of neutrophilic inflammation in severe asthma. METHODS: We examined whether sputum MIF protein correlated with clinical and molecular characteristics of severe neutrophilic asthma in the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort. We also investigated whether MIF regulates neutrophilic inflammation and glucocorticoid responsiveness in a murine model of severe asthma in vivo. RESULTS: MIF protein levels positively correlated with the number of exacerbations in the previous year, sputum neutrophils and oral corticosteroid use across all U-BIOPRED subjects. Further analysis of MIF protein expression according to U-BIOPRED-defined transcriptomic-associated clusters (TACs) revealed increased MIF protein and a corresponding decrease in annexin-A1 protein in TAC2, which is most closely associated with airway neutrophilia and NLRP3 inflammasome activation. In a murine model of severe asthma, treatment with the MIF antagonist ISO-1 significantly inhibited neutrophilic inflammation and increased glucocorticoid responsiveness. Coimmunoprecipitation studies using lung tissue lysates demonstrated that MIF directly interacts with and cleaves annexin-A1, potentially reducing its biological activity. CONCLUSION: Our data suggest that MIF promotes glucocorticoid-resistance of neutrophilic inflammation by reducing the biological activity of annexin-A1, a potent glucocorticoid-regulated protein that inhibits neutrophil accumulation at sites of inflammation. This represents a previously unrecognised role for MIF in the regulation of inflammation and points to MIF as a potential therapeutic target for the management of severe neutrophilic asthma.


Subject(s)
Asthma , Macrophage Migration-Inhibitory Factors , Humans , Animals , Mice , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/therapeutic use , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Disease Models, Animal , Asthma/drug therapy , Asthma/metabolism , Inflammation/metabolism , Neutrophils/metabolism , Annexins/metabolism , Annexins/therapeutic use
4.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L641-L652, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34405719

ABSTRACT

The receptor for advanced glycation end-products (RAGE) has been implicated in the pathophysiology of chronic obstructive pulmonary disease (COPD). However, it is still unknown whether RAGE directly contributes to alveolar epithelial damage and abnormal repair responses. We hypothesize that RAGE activation not only induces lung tissue damage but also hampers alveolar epithelial repair responses. The effects of the RAGE ligands LL-37 and HMGB1 were examined on airway inflammation and alveolar tissue damage in wild-type and RAGE-deficient mice and on lung damage and repair responses using murine precision cut lung slices (PCLS) and organoids. In addition, their effects were studied on the repair response of human alveolar epithelial A549 cells, using siRNA knockdown of RAGE and treatment with the RAGE inhibitor FPS-ZM1. We observed that intranasal installation of LL-37 and HMGB1 induces RAGE-dependent inflammation and severe alveolar tissue damage in mice within 6 h, with stronger effects in a mouse strain susceptible for emphysema compared with a nonsusceptible strain. In PCLS, RAGE inhibition reduced the recovery from elastase-induced alveolar tissue damage. In organoids, RAGE ligands reduced the organoid-forming efficiency and epithelial differentiation into pneumocyte-organoids. Finally, in A549 cells, we confirmed the role of RAGE in impaired repair responses upon exposure to LL-37. Together, our data indicate that activation of RAGE by its ligands LL-37 and HMGB1 induces acute lung tissue damage and that this impedes alveolar epithelial repair, illustrating the therapeutic potential of RAGE inhibitors for lung tissue repair in emphysema.


Subject(s)
Alveolar Epithelial Cells/pathology , Antimicrobial Cationic Peptides/metabolism , HMGB1 Protein/metabolism , Pulmonary Alveoli/injuries , Receptor for Advanced Glycation End Products/metabolism , A549 Cells , Animals , Benzamides/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Organoids/drug effects , Pancreatic Elastase/toxicity , Pulmonary Disease, Chronic Obstructive/pathology , Receptor for Advanced Glycation End Products/antagonists & inhibitors , Regeneration/physiology , Cathelicidins
5.
Curr Pharm Teach Learn ; 13(5): 460-470, 2021 05.
Article in English | MEDLINE | ID: mdl-33795097

ABSTRACT

INTRODUCTION: Minor ailments services (MASs) are pharmacy-based and support individuals to manage minor conditions. MASs are delivered by community pharmacists and non-pharmacist staff. Limited information exists regarding education, training, assessment requirements, and suitability of existing processes to support MAS delivery. The purpose of this study was to determine consensus amongst multiple stakeholder participants regarding these processes. METHODS: A modified Delphi process was utilized. Phase 1 consisted of stakeholder participants completing two rounds of an online questionnaire responding to Likert-items (Round 1 [R1] [n = 46]; Round 2 [R2] [n = 34]). Phase 2 consisted of three teleconference rounds discussing items that did not achieve consensus in the previous two rounds. Consensus was defined as ≥80% panel agreement. RESULTS: Forty MASs stakeholders participated in the study. MASs stakeholder participants included community pharmacists (n = 7), pharmacy student graduate pharmacist (n = 4), non-pharmacist staff (n = 13), faculty staff/academics (n = 5), general practitioners (n = 5), and individuals affiliated with pharmacy professional organizations (n = 6). Consensus was achieved on 22 of 46 statements in R1, 8 of 34 statements in R2, and 21 of 27 statements in Phase 2. CONCLUSIONS: It may be useful for MAS education and training to consider the clinical and non-clinical elements of service delivery. Training should be available to all community pharmacy staff. The results of this study may be useful to policymakers and professional organizations to enhance existing curricula or inform training guidelines for MAS delivery.


Subject(s)
Community Pharmacy Services , Pharmacies , Consensus , Delphi Technique , Humans , Pharmacists
6.
Int J Clin Pharm ; 43(3): 654-665, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33125624

ABSTRACT

Background Minor ailments services are structured pharmacy-based primary health care services that manage minor conditions. Limited training, education and assessment exists to promote the delivery of minor ailments services by pharmacy staff and it is unclear if the existing training and education processes meet professional requirements. Objective To explore the views and experiences of health professional stakeholders such as community pharmacists, intern pharmacists, medicines counter assistants and general medical practitioners with regards to minor ailments services education, training and assessment practices and preferences. Setting This study explored the views and experiences of health professional stakeholders in Australia. Method Semi-structured interviews were conducted, audio recorded, transcribed verbatim and then coded thematically using QSR Nvivo12. Main outcome measure Stakeholders' views and experiences regarding minor ailments services education, training and assessment practices and preferences. Results Twenty-eight interviews were conducted (community pharmacists n = 12; medicines counter assistants n = 4; intern pharmacists n = 9; general medical practitioners n = 3). Thematic analysis generated three themes: (1) pharmacy staff who require minor ailment service training; (2) acceptability and willingness to complete additional training; (3) learning preferences and approaches. Stakeholders reported considerations for the diverse roles in service delivery and fit for purpose tailored training. Conclusion Detailed practice guidelines may facilitate clarity of an individual staff member's role. Education and training in both clinical and non-clinical aspects of the service may be beneficial and may improve minor ailments service uptake and outcomes.


Subject(s)
Community Pharmacy Services , General Practitioners , Pharmacies , Attitude of Health Personnel , Humans , Pharmacists , Professional Role
7.
Allergy ; 76(4): 1123-1135, 2021 04.
Article in English | MEDLINE | ID: mdl-32799375

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4) is implicated in COPD. Although these receptors share common ligands and signalling pathways, it is not known whether they act in concert to drive pathological processes in COPD. We examined the impact of RAGE and/or TLR4 gene deficiency in a mouse model of COPD and also determined whether expression of these receptors correlates with airway neutrophilia and airway hyperresponsiveness (AHR) in COPD patients. METHODS: We measured airway inflammation and AHR in wild-type, RAGE-/- , TLR4-/- and TLR4-/- RAGE-/- mice following acute exposure to cigarette smoke (CS). We also examined the impact of smoking status on AGER (encodes RAGE) and TLR4 bronchial gene expression in patients with and without COPD. Finally, we determined whether expression of these receptors correlates with airway neutrophilia and AHR in COPD patients. RESULTS: RAGE-/- mice were protected against CS-induced neutrophilia and AHR. In contrast, TLR4-/- mice were not protected against CS-induced neutrophilia and had more severe CS-induced AHR. TLR4-/- RAGE-/- mice were not protected against CS-induced neutrophilia but were partially protected against CS-induced mediator release and AHR. Current smoking was associated with significantly lower AGER and TLR4 expression irrespective of COPD status, possibly reflecting negative feedback regulation. However, consistent with preclinical findings, AGER expression correlated with higher sputum neutrophil counts and more severe AHR in COPD patients. TLR4 expression did not correlate with neutrophilic inflammation or AHR. CONCLUSIONS: Inhibition of RAGE but not TLR4 signalling may protect against airway neutrophilia and AHR in COPD.


Subject(s)
Pulmonary Disease, Chronic Obstructive , Respiratory Hypersensitivity , Animals , Antigens, Neoplasm , Humans , Mice , Mitogen-Activated Protein Kinases , Pulmonary Disease, Chronic Obstructive/genetics , Receptor for Advanced Glycation End Products/genetics , Smoking , Toll-Like Receptor 4/genetics
8.
Curr Pharm Teach Learn ; 12(10): 1269-1287, 2020 10.
Article in English | MEDLINE | ID: mdl-32739065

ABSTRACT

BACKGROUND: Minor ailment services (MASs) are structured, protocol driven pharmacy services established locally or nationally. Community pharmacy staff may benefit from education and training to deliver MASs. Our objective was to examine the evidence regarding training, education, and assessment requirements associated with the delivery of MASs by community pharmacists and other community pharmacy staff. METHODS: Two independent literature search strategies were conducted to examine the grey literature and scientific literature. Inclusion criteria consisted of English written literature related to the training of pharmacists, medicine counter assistants (MCAs), pharmacy technicians, and pharmacy students in the context of MASs. RESULTS: Sixty-six grey literature records (n = 57) and scientific articles (n = 9) met inclusion criteria. Most trainings targeted community pharmacists and focused on clinical care aspects that did not include guidance on service parameters and MAS delivery. Training lacked uniformity and varied in terms of time commitment, cost, curricula, and assessment processes. Limited training was identified for community pharmacy staff, particularly MCAs. IMPLICATIONS: MAS training is primarily provided for community pharmacists, with scant MAS training for community pharmacy support staff. Furthermore, existing training for any stakeholder group did not include guidance pertaining to service delivery. A structured training approach for the entire community pharmacy team is recommended to promote MAS outcomes and deliver a robust, high quality service. Detailed protocols and guidelines may be needed to ensure skilled MAS providers can deliver quality patient care.


Subject(s)
Community Pharmacy Services , Pharmacies , Curriculum , Humans , Pharmacists , Pharmacy Technicians
9.
PLoS Pathog ; 16(7): e1008651, 2020 07.
Article in English | MEDLINE | ID: mdl-32658914

ABSTRACT

Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling.


Subject(s)
Airway Remodeling/immunology , HMGB1 Protein/immunology , Inflammation/immunology , Lymphocytes/immunology , Muscle, Smooth/immunology , Animals , Mice , Muscle, Smooth/pathology , Receptor for Advanced Glycation End Products/immunology
10.
Methods Mol Biol ; 2080: 203-212, 2020.
Article in English | MEDLINE | ID: mdl-31745883

ABSTRACT

Experimental mouse models of asthma are widely used to investigate the underlying mechanisms of this complex and heterogeneous disease. Using mouse models of ovalbumin-induced asthma, previous investigators have established a crucial role for MIF in the development of type 2-mediated eosinophilic asthma. Surprisingly, however, the role of MIF in other phenotypes of asthma has received little attention. MIF is an important mediator of neutrophilic inflammation, and also acts to antagonize the actions of corticosteroids. Thus, MIF may play a role in the development of severe forms of asthma in which airway neutrophilia and corticosteroid insensitivity are major features. In this chapter, we provide an experimental protocol that may be used to investigate the role of MIF in a mouse model of severe corticosteroid-resistant neutrophilic asthma.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Asthma/etiology , Asthma/metabolism , Drug Resistance/genetics , Intramolecular Oxidoreductases/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neutrophils/immunology , Neutrophils/metabolism , Animals , Antigens, Dermatophagoides/immunology , Asthma/diagnosis , Asthma/drug therapy , Dexamethasone/pharmacology , Disease Models, Animal , Drug Resistance/drug effects , Intramolecular Oxidoreductases/metabolism , Isoxazoles/pharmacology , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Neutrophils/drug effects , Severity of Illness Index
12.
J Proteome Res ; 17(1): 33-45, 2018 01 05.
Article in English | MEDLINE | ID: mdl-28976774

ABSTRACT

The release of damage-associated molecular patterns (DAMPs) by airway epithelial cells is believed to play a crucial role in the initiation and development of chronic airway conditions such as asthma and chronic obstructive pulmonary disease (COPD). Intriguingly, the classic DAMP high-mobility group box-1 (HMGB1) is detected in the culture supernatant of airway epithelial cells under basal conditions, indicating a role for HMGB1 in the regulation of epithelial cellular and immune homeostasis. To gain contextual insight into the potential role of HMGB1 in airway epithelial cell homeostasis, we used the orthogonal and complementary methods of high-resolution clear native electrophoresis, immunoprecipitation, and pull-downs coupled to liquid chromatography-tandem mass spectrometry (LC-MS/MS) to profile HMGB1 and its binding partners in the culture supernatant of unstimulated airway epithelial cells. We found that HMGB1 presents exclusively as a protein complex under basal conditions. Moreover, protein network analysis performed on 185 binding proteins revealed 14 that directly associate with HMGB1: amyloid precursor protein, F-actin-capping protein subunit alpha-1 (CAPZA1), glyceraldehyde-3 phosphate dehydrogenase (GAPDH), ubiquitin, several members of the heat shock protein family (HSPA8, HSP90B1, HSP90AA1), XRCC5 and XRCC6, high mobility group A1 (HMGA1), histone 3 (H3F3B), the FACT (facilitates chromatin transcription) complex constituents SUPT1H and SSRP1, and heterogeneous ribonucleoprotein K (HNRNPK). These studies provide a new understanding of the extracellular functions of HMGB1 in cellular and immune homeostasis at the airway mucosal surface and could have implications for therapeutic targeting.


Subject(s)
Epithelial Cells/physiology , HMGB1 Protein/analysis , Homeostasis , Proteomics/methods , Respiratory Mucosa/cytology , HMGB1 Protein/metabolism , HMGB1 Protein/physiology , Humans , Protein Binding
13.
Drug Dev Ind Pharm ; 43(9): 1510-1518, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28425305

ABSTRACT

OBJECTIVE: The aim of this work was to develop an amorphous solid dispersions/solutions (ASD) of a poorly soluble drug, budesonide (BUD) with a novel polymer Soluplus® (BASF, Germany) using a freeze-drying technique, in order to improve dissolution and absorption through the nasal route. SIGNIFICANCE: The small volume of fluid present in the nasal cavity limits the absorption of a poorly soluble drug. Budesonide is a corticosteroid, practically insoluble and normally administered as a suspension-based nasal spray. METHODS: The formulation was prepared through freeze-drying of polymer-drug solution. The formulation was assessed for its physicochemical (specific surface area, calorimetric analysis and X-ray powder diffraction), release properties and aerodynamic properties as well as transport in vitro using RPMI 2650 nasal cells, in order to elucidate the efficacy of the Soluplus-BUD formulation. RESULTS: The freeze-dried Soluplus-BUD formulation (LYO) showed a porous structure with a specific surface area of 1.4334 ± 0.0178 m2/g. The calorimetric analysis confirmed an interaction between BUD and Soluplus and X-ray powder diffraction the amorphous status of the drug. The freeze-dried formulation (LYO) showed faster release compared to both water-based suspension and dry powder commercial products. Furthermore, a LYO formulation, bulked with calcium carbonate (LYO-Ca), showed suitable aerodynamic characteristics for nasal drug delivery. The permeation across RPMI 2650 nasal cell model was higher compared to a commercial water-based BUD suspension. CONCLUSIONS: Soluplus has been shown to be a promising polymer for the formulation of BUD amorphous solid suspension/solution. This opens up opportunities to develop new formulations of poorly soluble drug for nasal delivery.


Subject(s)
Aerosols/administration & dosage , Budesonide/administration & dosage , Drug Carriers/administration & dosage , Polyethylene Glycols/administration & dosage , Polyvinyls/administration & dosage , Aerosols/chemistry , Budesonide/chemistry , Chemistry, Pharmaceutical , Desiccation , Drug Carriers/chemistry , Drug Delivery Systems , Freeze Drying , Polyethylene Glycols/chemistry , Polyvinyls/chemistry , Porosity , Powders/administration & dosage , X-Ray Diffraction
14.
Mol Immunol ; 86: 10-15, 2017 06.
Article in English | MEDLINE | ID: mdl-28249679

ABSTRACT

Autophagy is a ubiquitous cellular mechanism for the targeted lysosomal degradation of various cytosolic constituents, from proteins to organelles. As an essential homeostatic mechanism, autophagy is upregulated in response to numerous environmental and pharmacological stimuli, including starvation, where it facilitates the recycling of essential amino acids. In addition, autophagy plays specific roles within the immune system; it serves as a source of peptides for antigen presentation, a mechanism for the engulfment and degradation of intracellular pathogens and as a key regulator of inflammatory cytokines. In particular, autophagy has been shown to play a number of roles in regulating inflammasome activation, from the removal of inflammasome-activating endogenous signals, to the sequestration and degradation of inflammasome components. Autophagy also plays a role in determining the fate of IL-1ß, which is concentrated in autophagosomes. This review discusses a growing body of literature that suggests autophagy is a critical regulator of inflammasome activation and the subsequent release of IL-1 family cytokines.


Subject(s)
Autophagy/immunology , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1/metabolism , Animals , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Humans , Interleukin-18/metabolism , Membrane Proteins/metabolism , Mice , Mitochondria/physiology , Mitochondrial Proteins/metabolism , NLR Proteins/metabolism
15.
Elife ; 62017 01 18.
Article in English | MEDLINE | ID: mdl-28099113

ABSTRACT

Asthma is a chronic inflammatory disease. Although many patients with asthma develop type-2 dominated eosinophilic inflammation, a number of individuals develop paucigranulocytic asthma, which occurs in the absence of eosinophilia or neutrophilia. The aetiology of paucigranulocytic asthma is unknown. However, both respiratory syncytial virus (RSV) infection and mutations in the receptor for advanced glycation endproducts (RAGE) are risk factors for asthma development. Here, we show that RAGE deficiency impairs anti-viral immunity during an early-life infection with pneumonia virus of mice (PVM; a murine analogue of RSV). The elevated viral load was associated with the release of high mobility group box-1 (HMGB1) which triggered airway smooth muscle remodelling in early-life. Re-infection with PVM in later-life induced many of the cardinal features of asthma in the absence of eosinophilic or neutrophilic inflammation. Anti-HMGB1 mitigated both early-life viral disease and asthma-like features, highlighting HMGB1 as a possible novel therapeutic target.


Subject(s)
Agranulocytosis/complications , Agranulocytosis/genetics , Asthma/genetics , Asthma/pathology , Genetic Predisposition to Disease , HMGB1 Protein/metabolism , Receptor for Advanced Glycation End Products/deficiency , Animals , Mice , Murine pneumonia virus/immunology , Viral Load
17.
Br J Pharmacol ; 174(1): 3-14, 2017 01.
Article in English | MEDLINE | ID: mdl-27759879

ABSTRACT

The SPARC (secreted protein acidic and rich in cysteine) protein is matricellular molecule regulating interactions between cells and their surrounding extracellular matrix (ECM). This protein thus governs fundamental cellular functions such as cell adhesion, proliferation and differentiation. SPARC also regulates the expression and activity of numerous growth factors and matrix metalloproteinases essential for ECM degradation and turnover. Studies in SPARC-null mice have revealed a critical role for SPARC in tissue development, injury and repair and in the regulation of the immune response. In the lung, SPARC drives pathological responses in non-small cell lung cancer and idiopathic pulmonary fibrosis by promoting microvascular remodelling and excessive deposition of ECM proteins. Remarkably, although chronic airway conditions such as asthma and chronic obstructive pulmonary disease (COPD) involve significant remodelling in both the airway and vascular compartments, the role of SPARC in these conditions has thus far been overlooked. In this review, we discuss the role of SPARC in lung cancer and pulmonary fibrosis, as well as potential mechanisms by which it may contribute to the disease process in asthma and COPD.


Subject(s)
Lung Neoplasms/metabolism , Osteonectin/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Fibrosis/metabolism , Animals , Humans
18.
Int J Nanomedicine ; 11: 6575-6590, 2016.
Article in English | MEDLINE | ID: mdl-27994459

ABSTRACT

PURPOSE: Along with their cholesterol-lowering effect, statins have shown a wide range of pleiotropic effects potentially beneficial to neurodegenerative diseases. However, such effects are extremely elusive via the conventional oral administration. The purpose of the present study was to prepare and characterize the physicochemical properties and the in vivo biodistribution of simvastatin-loaded lecithin/chitosan nanoparticles (SVT-LCNs) suitable for nasal administration in view of an improved delivery of the statins to the brain. MATERIALS AND METHODS: Chitosan, lecithin, and different oil excipients were used to prepare nanocapsules loaded with simvastatin. Particle size distribution, surface charge, structure, simvastatin loading and release, and interaction with mucus of nanoparticles were determined. The nanoparticle nasal toxicity was evaluated in vitro using RPMI 2651 nasal cell lines. Finally, in vivo biodistribution was assessed by gamma scintigraphy via Tc99m labeling of the particles. RESULTS: Among the different types of nanoparticles produced, the SVT-LCN_MaiLab showed the most ideal physicochemical characteristics, with small diameter (200 nm), positive surface charge (+48 mV) and high encapsulation efficiency (EE; 98%). Size distribution was further confirmed by nanoparticle tracking analysis and electron microscopy. The particles showed a relatively fast release of simvastatin in vitro (35.6%±4.2% in 6 hours) in simulated nasal fluid. Blank nanoparticles did not show cytotoxicity, evidencing that the formulation is safe for nasal administration, while cytotoxicity of simvastatin-loaded nanoparticles (IC50) was found to be three times lower than the drug solution (9.92 vs 3.50 µM). In rats, a significantly higher radioactivity was evidenced in the brain after nasal delivery of simvastatin-loaded nanoparticles in comparison to the administration of a similar dose of simvastatin suspension. CONCLUSION: The SVT-LCNs developed presented some of the most desirable characteristics for mucosal delivery, that is, small particle size, positive surface charge, long-term stability, high EE, and mucoadhesion. In addition, they displayed two exciting features: First was their biodegradability by enzymes present in the mucus layer, such as lysozyme. This indicates a new Trojan-horse strategy which may enhance drug release in the proximity of the nasal mucosa. Second was their ability to enhance the nose-to-brain transport as evidenced by preliminary gamma scintigraphy studies.


Subject(s)
Brain/drug effects , Drug Delivery Systems , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Nanoparticles/administration & dosage , Nasal Mucosa/drug effects , Simvastatin/pharmacology , Administration, Intranasal , Animals , Brain/metabolism , Chitosan/chemistry , Drug Liberation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Microscopy, Electron, Scanning Transmission , Nanoparticles/chemistry , Nanoparticles/ultrastructure , Nasal Mucosa/metabolism , Particle Size , Rats , Rats, Wistar , Simvastatin/administration & dosage , Tissue Distribution
19.
J Allergy Clin Immunol ; 136(4): 1065-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25930193

ABSTRACT

BACKGROUND: A variant in the IL-6 receptor (IL-6R) gene increases asthma risk and is predicted to decrease IL-6 classic signaling and increase IL-6 trans-signaling. This suggests that inhibition of IL-6 trans-signaling, but not classic signaling, might suppress allergic airway inflammation. OBJECTIVES: We sought to determine whether IL-6 signaling contributes to (1) acute experimental asthma induced by clinically relevant allergens and (2) variation in asthma clinical phenotypes in asthmatic patients. METHODS: Mice were sensitized to house dust mite (HDM) or cockroach at day 0, treated with IL-6R inhibitors at day 13, and challenged with the same allergen at days 14 to 17. End points were measured 3 hours after the final challenge. IL-6 and soluble IL-6 receptor (sIL-6R) expression in induced sputum of asthmatic patients was correlated with asthma clinical phenotypes. RESULTS: Both HDM and cockroach induced a type 2/type 17 cytokine profile and mixed granulocytic inflammation in the airways. Both allergens increased IL-6 expression in the airways, but only cockroach induced sIL-6R expression. Therefore HDM challenge promoted IL-6 classic signaling but not trans-signaling; in this model treatment with anti-IL-6R did not suppress airway inflammation. In contrast, cockroach-induced inflammation involved activation of IL-6 trans-signaling and production of IL-17A by γδ T cells. Anti-IL-6R, selective blockade of sIL-6R, or γδ T-cell deficiency significantly attenuated cockroach-induced inflammation. Asthmatic patients with high airway IL-6 and sIL-6R levels were enriched for the neutrophilic and mixed granulocytic subtypes. CONCLUSION: Experimental asthma associated with both high IL-6 and high sIL-6R levels in the airways is attenuated by treatment with IL-6R inhibitors.


Subject(s)
Asthma/immunology , Interleukin-6/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Interleukin-6/immunology , Signal Transduction/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Allergens/immunology , Allergens/toxicity , Animals , Asthma/chemically induced , Asthma/pathology , Cockroaches/immunology , Mice , Pyroglyphidae/immunology , Signal Transduction/drug effects , Th17 Cells/pathology , Th2 Cells/pathology
20.
J Allergy Clin Immunol ; 134(2): 440-50, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24506934

ABSTRACT

BACKGROUND: The receptor for advanced glycation end products (RAGE) shares common ligands and signaling pathways with TLR4, a key mediator of house dust mite (Dermatophagoides pteronyssinus) (HDM) sensitization. We hypothesized that RAGE and its ligand high-mobility group box-1 (HMGB1) cooperate with TLR4 to mediate HDM sensitization. OBJECTIVES: To determine the requirement for HMGB1 and RAGE, and their relationship with TLR4, in airway sensitization. METHODS: TLR4(-/-), RAGE(-/-), and RAGE-TLR4(-/-) mice were intranasally exposed to HDM or cockroach (Blatella germanica) extracts, and features of allergic inflammation were measured during the sensitization or challenge phase. Anti-HMGB1 antibody and the IL-1 receptor antagonist Anakinra were used to inhibit HMGB1 and the IL-1 receptor, respectively. RESULTS: The magnitude of allergic airway inflammation in response to either HDM or cockroach sensitization and/or challenge was significantly reduced in the absence of RAGE but not further diminished in the absence of both RAGE and TLR4. HDM sensitization induced the release of HMGB1 from the airway epithelium in a biphasic manner, which corresponded to the sequential activation of TLR4 then RAGE. Release of HMGB1 in response to cockroach sensitization also was RAGE dependent. Significantly, HMGB1 release occurred downstream of TLR4-induced IL-1α, and upstream of IL-25 and IL-33 production. Adoptive transfer of HDM-pulsed RAGE(+/+)dendritic cells to RAGE(-/-) mice recapitulated the allergic responses after HDM challenge. Immunoneutralization of HMGB1 attenuated HDM-induced allergic airway inflammation. CONCLUSION: The HMGB1-RAGE axis mediates allergic airway sensitization and airway inflammation. Activation of this axis in response to different allergens acts to amplify the allergic inflammatory response, which exposes it as an attractive target for therapeutic intervention.


Subject(s)
Allergens/immunology , HMGB1 Protein/immunology , Receptors, Immunologic/immunology , Respiratory Hypersensitivity/immunology , Administration, Intranasal , Adoptive Transfer , Allergens/administration & dosage , Animals , Antibodies, Neutralizing/pharmacology , Blattellidae/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/transplantation , Dermatophagoides pteronyssinus/immunology , Gene Expression Regulation , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-33 , Interleukins/genetics , Interleukins/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , Mice, Knockout , Receptor for Advanced Glycation End Products , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Signal Transduction , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...