Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Lyn M. Moir is active.

Publication


Featured researches published by Lyn M. Moir.


Thorax | 2007

Comparison of gel contraction mediated by airway smooth muscle cells from patients with and without asthma

Hisako Matsumoto; Lyn M. Moir; Brian Oliver; Janette K. Burgess; Michael Roth; Judith L. Black; Brent E. McParland

Backgrounds: Exaggerated bronchial constriction is the most significant and life threatening response of patients with asthma to inhaled stimuli. However, few studies have investigated the contractility of airway smooth muscle (ASM) from these patients. The purpose of this study was to establish a method to measure contraction of ASM cells by embedding them into a collagen gel, and to compare the contraction between subjects with and without asthma. Methods: Gel contraction to histamine was examined in floating gels containing cultured ASM cells from subjects with and without asthma following overnight incubation while unattached (method 1) or attached (method 2) to casting plates. Smooth muscle myosin light chain kinase protein levels were also examined. Results: Collagen gels containing ASM cells reduced in size when stimulated with histamine in a concentration-dependent manner and reached a maximum at a mean (SE) of 15.7 (1.2) min. This gel contraction was decreased by inhibitors for phospholipase C (U73122), myosin light chain kinase (ML-7) and Rho kinase (Y27632). When comparing the two patient groups, the maximal decreased area of gels containing ASM cells from patients with asthma was 19 (2)% (n = 8) using method 1 and 22 (3)% (n = 6) using method 2, both of which were greater than that of cells from patients without asthma: 13 (2)% (n = 9, p = 0.05) and 10 (4)% (n = 5, p = 0.024), respectively. Smooth muscle myosin light chain kinase levels were not different between the two groups. Conclusion: The increased contraction of asthmatic ASM cells may be responsible for exaggerated bronchial constriction in asthma.


PLOS ONE | 2011

β2-Agonist induced cAMP is decreased in asthmatic airway smooth muscle due to increased PDE4D

Thomas Trian; Janette K. Burgess; Kyoko Niimi; Lyn M. Moir; Qi Mu Ge; Patrick Berger; Stephen B. Liggett; Judith L. Black; Brian Oliver

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 β-agonist induced ASM relaxation in asthmatics, but the mechanism is not known. Objective To characterize the potential defect in β-agonist induced cAMP in ASM derived from asthmatic in comparison to non-asthmatic subjects and to investigate its mechanism. Methods We examined β2-adrenergic (β2AR) receptor expression and basal β-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 β-agonist induced cAMP in ASM from asthmatics results from enhanced degradation due to increased PDE4D expression. Clinical manifestations of this dysregulation would be suboptimal β-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.


The Journal of Allergy and Clinical Immunology | 2008

Transforming growth factor β1 increases fibronectin deposition through integrin receptor α5β1 on human airway smooth muscle

Lyn M. Moir; Janette K. Burgess; Judith L. Black

BACKGROUND Integrin receptors signal to and from the extracellular matrix. Altered expression of the integrin receptors, such as the fibronectin receptor alpha(5)beta(1), might be implicated in extracellular matrix accumulation in airway remodeling in asthma. OBJECTIVE We examined the effect of TGF-beta stimulation on integrin alpha(5)beta(1) expression and the role of alpha(5)beta(1) in fibronectin deposition and proliferation. METHODS Integrin subunit alpha(5) and beta(1) expression in airway smooth muscle (ASM) from subjects with and without asthma was examined by means of PCR and flow cytometry. The effect of blocking alpha(5)beta(1) receptor on ASM proliferation to FBS was assessed by using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay. Cells were stimulated with TGF-beta in the presence or absence of extracellular signal-regulated kinase, phosphoinositide-3 kinase, or p38 inhibitors and antibodies to the alpha(5) and beta(1) subunits. The effect of blocking alpha(5)beta(1) receptor on fibronectin deposition was assessed by means of immunocytochemistry. RESULTS Proliferation of ASM cells from asthmatic and nonasthmatic subjects was inhibited by blocking the fibronectin receptor subunit alpha(5)beta(1). TGF-beta-induced alpha(5)beta(1) was extracellular signal-regulated kinase dependent but not phosphoinositide-3 kinase or p38 dependent. Blockade of the alpha(5)beta(1) receptor inhibited TGF-beta-induced fibronectin matrix deposition. CONCLUSION Through its increased expression by the profibrotic stimulus TGF-beta, integrin alpha(5)beta(1) might be important in regulating fibronectin deposition.


Pulmonary Pharmacology & Therapeutics | 2009

Tissue and matrix influences on airway smooth muscle function

Janette K. Burgess; Claudia Ceresa; Simon R. Johnson; Varsha Kanabar; Lyn M. Moir; Trang T.B. Nguyen; Brian Oliver; Michael Schuliga; Jane E. Ward

Asthma is characterized by structural changes in the airways - airway remodelling. These changes include an increase in the bulk of the airway smooth muscle (ASM) and alterations in the profile of extracellular matrix (ECM) proteins in the airway wall. The mechanisms leading to airway remodelling are not well understood. ASM cells have the potential to play a key role in these processes through the production and release of ECM proteins. The ASM cells and ECM proteins are each able to influence the behaviour and characteristics of the other. The modified ECM profile in the asthmatic airway may contribute to the altered behaviour of the ASM cells, such responses to ECM proteins are modulated through the cell surface expression of integrin receptors. ASM cells from asthmatic individuals express different levels of some integrin subunits compared to nonasthmatic ASM cells, which have the potential to further influence their responses to the ECM proteins in the airways. ECM homeostasis requires the presence and activation of matrix metalloproteinases and their tissue inhibitors, which in turn modulate the interaction of the ASM cells and the ECM proteins. Furthermore, the complex interactions of the ASM cells and the ECM in the asthmatic airways and the role played by external stimuli, such as viral infections, to modulate airway remodelling are currently unknown. This review summarises our current understanding of the influence of the ECM on ASM function.


Journal of Cellular Physiology | 2010

TGFβ1 induces IL-6 and inhibits IL-8 release in human bronchial epithelial cells: the role of Smad2/3

Qi Ge; Lyn M. Moir; Judith L. Black; Brian Oliver; Janette K. Burgess

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) β1, all of which are elevated in asthmatic airways. This study examines the signaling pathways leading to TGFβ1 induced IL‐6 and IL‐8 in primary HBE cells from asthmatic and non‐asthmatic volunteers. HBE cells were stimulated with TGFβ1 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. TGFβ1 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 TGFβ1 induced IL‐6 in both cell groups. TGFβ1 induced Smad2 phosphorylation, and blockade of Smad2/3 prevented both the TGFβ1 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 TGFβ1 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. J. Cell. Physiol. 225: 846–854, 2010.


American Journal of Respiratory and Critical Care Medicine | 2010

Reduction of tumstatin in asthmatic airways contributes to angiogenesis, inflammation, and hyperresponsiveness

Janette K. Burgess; Sarah Boustany; Lyn M. Moir; Markus Weckmann; Justine Y. Lau; Karryn T. Grafton; Melissa Baraket; Philip M. Hansbro; Nicole G. Hansbro; Paul S. Foster; Judith L. Black; Brian Oliver

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 tumstatins 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.


PLOS ONE | 2010

Fibulin-1 Is Increased in Asthma – A Novel Mediator of Airway Remodeling?

Justine Y. Lau; Brian Oliver; Melissa Baraket; Emma L. Beckett; Nicole G. Hansbro; Lyn M. Moir; S.D. Wilton; Carolyn Williams; Paul S. Foster; Philip M. Hansbro; Judith L. Black; Janette K. Burgess

Background The extracellular matrix is a dynamic and complex network of macromolecules responsible for maintaining and influencing cellular functions of the airway. The role of fibronectin, an extracellular matrix protein, is well documented in asthma. However, the expression and function of fibulin-1, a secreted glycoprotein which interacts with fibronectin, has not been reported. Fibulin-1 is widely expressed in basement membranes in many organs including the lung. There are four isoforms in humans (A–D) of which fibulin-1C and 1D predominate. The objective of this study was to study the expression of fibulin-1 in volunteers with and without asthma, and to examine its function in vitro. Methodology/Principal Findings We used immunohistochemistry and dot-blots to examine fibulin-1 levels in bronchial biopsies, bronchoalveolar lavage fluid and serum. Real-time PCR for fibulin-1C and 1D, and ELISA and western blotting for fibulin-1 were used to study the levels in airway smooth muscle cells. The function of fibulin-1C was determined by assessing its role, using an antisense oligonucleotide, in cell proliferation, migration and wound healing. A murine model of airway hyperresponsiveness (AHR) was used to explore the biological significance of fibulin-1. Levels of fibulin-1 were significantly increased in the serum and bronchoalveolar lavage fluid of 21 asthmatics compared with 11 healthy volunteers. In addition fibulin-1 was increased in asthma derived airway smooth muscle cells and fibulin-1C contributed to the enhanced proliferation and wound repair in these cells. These features were reversed when fibulin-1C was suppressed using an antisense oligomer. In a mouse model of AHR, treatment with an AO inhibited the development of AHR to methacholine. Conclusions Our data collectively suggest fibulin-1C may be worthy of further investigation as a target for airway remodeling in asthma.


Pulmonary Pharmacology & Therapeutics | 2013

Novel non-canonical TGF-β signaling networks: emerging roles in airway smooth muscle phenotype and function.

Behzad Yeganeh; Subhendu Mukherjee; Lyn M. Moir; Kuldeep Kumawat; Hessam H. Kashani; Rushita A. Bagchi; Hoeke A. Baarsma; Reinoud Gosens; Saeid Ghavami

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-β) 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-β 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-β signaling, including MAPK, PI3K, WNT/β-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-β signaling will be discussed.


Allergy | 2011

Human lung mast cells modulate the functions of airway smooth muscle cells in asthma.

Hatem Alkhouri; F. Hollins; Lyn M. Moir; Christopher E. Brightling; Carol L. Armour; J. M. Hughes

To cite this article: Alkhouri H, Hollins F, Moir LM, Brightling CE, Armour CL, Hughes JM. Human lung mast cells modulate the functions of airway smooth muscle cells in asthma. Allergy 2011; 66: 1231–1241.


Pulmonary Pharmacology & Therapeutics | 2013

Pro-inflammatory and immunomodulatory functions of airway smooth muscle: Emerging concepts

Yuxiu C. Xia; Naresh Singh Redhu; Lyn M. Moir; Cynthia Koziol-White; Alaina J. Ammit; Laila Al-Alwan; Blanca Camoretti-Mercado; Rachel L. Clifford

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.

Collaboration


Dive into the Lyn M. Moir's collaboration.

Top Co-Authors

Avatar

Janette K. Burgess

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Judith L. Black

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Qi Ge

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Vera P. Krymskaya

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Ho Yin Ng

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Markus Weckmann

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Carol L. Armour

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Kyoko Niimi

Woolcock Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge