Network


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

Hotspot


Dive into the research topics where Lynne A. Murray is active.

Publication


Featured researches published by Lynne A. Murray.


Journal of Clinical Investigation | 2004

Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis

Roderick J. Phillips; Marie D. Burdick; Kurt Hong; Marin Lutz; Lynne A. Murray; Ying Ying Xue; John A. Belperio; Michael P. Keane; Robert M. Strieter

Previous reports have identified a circulating pool of CD45(+) collagen I(+) CXCR4(+) (CD45(+)Col I(+)CXCR4(+)) cells, termed fibrocytes, that traffic to areas of fibrosis. No studies have demonstrated that these cells actually contribute to fibrosis, however. Pulmonary fibrosis was originally thought to be mediated solely by resident lung fibroblasts. Here we show that a population of human CD45(+)Col I(+)CXCR4(+) circulating fibrocytes migrates in response to CXCL12 and traffics to the lungs in a murine model of bleomycin-induced pulmonary fibrosis. Next, we demonstrated that murine CD45(+)Col I(+)CXCR4(+) fibrocytes also traffic to the lungs in response to a bleomycin challenge. Maximal intrapulmonary recruitment of CD45(+)Col I(+)CXCR4(+) fibrocytes directly correlated with increased collagen deposition in the lungs. Treatment of bleomycin-exposed animals with specific neutralizing anti-CXCL12 Abs inhibited intrapulmonary recruitment of CD45(+)Col I(+)CXCR4(+) circulating fibrocytes and attenuated lung fibrosis. Thus, our results demonstrate, we believe for the first time, that circulating fibrocytes contribute to the pathogenesis of pulmonary fibrosis.


Journal of Immunology | 2004

The Role of the Th2 CC Chemokine Ligand CCL17 in Pulmonary Fibrosis

John A. Belperio; Maria Dy; Lynne A. Murray; Marie D. Burdick; Ying Y. Xue; Robert M. Strieter; Michael P. Keane

Increasing evidence suggests that the development of pulmonary fibrosis is a Th2-mediated process. We hypothesized that the CC chemokines that are associated with a Th2 profile (CCL17 and CCL22) have an important role in the development of pulmonary fibrosis. We measured CCL17 and CCL22 during the pathogenesis of bleomycin-induced pulmonary fibrosis. We found that both CCL17 and CCL22 were significantly elevated through day 20 as compared with control mice. Peak expression of CCL22 preceded the peak levels of CCL17, as measured by real-time quantitative PCR. CCR4 is the receptor for CCL17 and CCL22 therefore, to further characterize the role of CCL17 and CCL22, we measured CCR4 mRNA in lung tissue of bleomycin-treated mice by real-time quantitative PCR. CCR4 was significantly elevated in bleomycin-treated mice as compared with control mice. Immunolocalization demonstrated that CCR4 was expressed predominantly on macrophages. Neutralization of CCL17, but not CCL22, led to a reduction in pulmonary fibrosis. Immunolocalization of bleomycin-treated lung tissue and human idiopathic pulmonary fibrosis tissue specimens showed that epithelial cells expressed CCL17. These findings demonstrate a central role for Th2 chemokines and the macrophage in the pathogenesis of pulmonary fibrosis and are further support for the role of a Th2 phenotype in the pathogenesis of pulmonary fibrosis.


Journal of Immunology | 2004

CXCR2 Is Critical to Hyperoxia-Induced Lung Injury

Richard D. Sue; John A. Belperio; Marie D. Burdick; Lynne A. Murray; Ying Ying Xue; Maria Dy; Jeffery J. Kwon; Michael P. Keane; Robert M. Strieter

Hyperoxia-induced lung injury is characterized by infiltration of activated neutrophils in conjunction with endothelial and epithelial cell injury, followed by fibrogenesis. Specific mechanisms recruiting neutrophils to the lung during hyperoxia-induced lung injury have not been fully elucidated. Because CXCL1 and CXCL2/3, acting through CXCR2, are potent neutrophil chemoattractants, we investigated their role in mediating hyperoxia-induced lung injury. Under variable concentrations of oxygen, murine survival during hyperoxia-induced lung injury was dose dependent. Eighty percent oxygen was associated with 50% mortality at 6 days, while greater oxygen concentrations were more lethal. Using 80% oxygen, we found that lungs harvested at day 6 demonstrated markedly increased neutrophil sequestration and lung injury. Expression of CXCR2 ligands paralleled neutrophil recruitment to the lung and CXCR2 mRNA expression. Inhibition of CXC chemokine ligands/CXCR2 interaction using CXCR2−/− mice exposed to hyperoxia significantly reduced neutrophil sequestration and lung injury, and led to a significant survival advantage as compared with CXCR2+/+ mice. These findings demonstrate that CXC chemokine ligand/CXCR2 biological axis is critical during the pathogenesis of hyperoxia-induced lung injury.


PLOS ONE | 2011

A micro RNA processing defect in rapidly progressing idiopathic pulmonary fibrosis

Sameer R. Oak; Lynne A. Murray; Athula Herath; Matthew A. Sleeman; Ian K. Anderson; Amrita Joshi; Ana Lucia Coelho; Kevin R. Flaherty; Galen B. Toews; Darryl A. Knight; Fernando J. Martinez; Cory M. Hogaboam

Background Idiopathic pulmonary fibrosis exhibits differential progression from the time of diagnosis but the molecular basis for varying progression rates is poorly understood. The aim of the present study was to ascertain whether differential miRNA expression might provide one explanation for rapidly versus slowly progressing forms of IPF. Methodology and Principal Findings miRNA and mRNA were isolated from surgical lung biopsies from IPF patients with a clinically documented rapid or slow course of disease over the first year after diagnosis. A quantitative PCR miRNA array containing 88 of the most abundant miRNA in the human genome was used to profile lung biopsies from 9 patients with rapidly progressing IPF, 6 patients with slowly progressing IPF, and 10 normal lung biopsies. Using this approach, 11 miRNA were significantly increased and 36 were significantly decreased in rapid biopsies compared with normal biopsies. Slowly progressive biopsies exhibited 4 significantly increased miRNA and 36 significantly decreased miRNA compared with normal lung. Among the miRNA present in IPF with validated mRNA targets were those with regulatory effects on epithelial-mesenchymal transition (EMT). Five miRNA (miR-302c, miR-423-5p, miR-210, miR-376c, and miR-185) were significantly increased in rapid compared with slow IPF lung biopsies. Additional analyses of rapid biopsies and fibroblasts grown from the same biopsies revealed that the expression of AGO1 and AGO2 (essential components of the miRNA processing RISC complex) were lower compared with either slow or normal lung biopsies and fibroblasts. Conclusion These findings suggest that the development and/or clinical progression of IPF might be the consequence of aberrant miRNA processing.


American Journal of Respiratory and Critical Care Medicine | 2008

Deleterious Role of TLR3 during Hyperoxia-induced Acute Lung Injury

Lynne A. Murray; Darryl A. Knight; Laura McAlonan; Rochelle L. Argentieri; Amrita Joshi; Furquan Shaheen; Mark Cunningham; Lena Alexopolou; Richard A. Flavell; Robert T. Sarisky; Cory M. Hogaboam

RATIONALE Acute respiratory distress syndrome (ARDS) manifests clinically as a consequence of septic and/or traumatic injury in the lung. Oxygen therapy remains a major therapeutic intervention in ARDS, but this can contribute further to lung damage. Patients with ARDS are highly susceptible to viral infection and it may be due to altered Toll-like receptor (TLR) expression. OBJECTIVES To evaluate the role of TLR3 in ARDS. METHODS TLR3 expression and signaling was determined in airway epithelial cells after in vitro hyperoxia challenge. Using a murine model of hyperoxia-induced lung injury, the role of TLR3 was determined using either TLR3-gene deficient mice or a specific neutralizing antibody directed to TLR3. MEASUREMENTS AND MAIN RESULTS Increased TLR3 expression was observed in airway epithelial cells from patients with ARDS. Further, hyperoxic conditions alone were a major stimulus for increased TLR3 expression and activation in cultured human epithelial cells. Interestingly, TLR3(-/-) mice exhibited less acute lung injury, activation of apoptotic cascades, and extracellular matrix deposition after 5 days of 80% oxygen compared with wild-type (TLR3(+/+)) mice under the same conditions. Administration of a monoclonal anti-TLR3 antibody to TLR3(+/+) mice exposed to hyperoxic conditions likewise protected these mice from lung injury and inflammation. CONCLUSIONS The potential for redundancy in function as well as cross-talk between distinct TLRs may indeed contribute to whether the inflammatory cascade can be effectively disrupted once signaling has been initiated. Together, these data show that TLR3 has a major role in the development of ARDS-like pathology in the absence of a viral pathogen.


American Journal of Respiratory Cell and Molecular Biology | 2014

Targeting Interleukin-13 with Tralokinumab Attenuates Lung Fibrosis and Epithelial Damage in a Humanized SCID Idiopathic Pulmonary Fibrosis Model

Lynne A. Murray; Huilan Zhang; Sameer R. Oak; Ana Lucia Coelho; Athula Herath; Kevin R. Flaherty; Joyce S. Lee; Matthew J. Bell; Darryl A. Knight; Fernando J. Martinez; Matthew A. Sleeman; Erica L. Herzog; Cory M. Hogaboam

The aberrant fibrotic and repair responses in the lung are major hallmarks of idiopathic pulmonary fibrosis (IPF). Numerous antifibrotic strategies have been used in the clinic with limited success, raising the possibility that an effective therapeutic strategy in this disease must inhibit fibrosis and promote appropriate lung repair mechanisms. IL-13 represents an attractive target in IPF, but its disease association and mechanism of action remains unknown. In the present study, an overexpression of IL-13 and IL-13 pathway markers was associated with IPF, particularly a rapidly progressive form of this disease. Targeting IL-13 in a humanized experimental model of pulmonary fibrosis using tralokinumab (CAT354) was found to therapeutically block aberrant lung remodeling in this model. However, targeting IL-13 was also found to promote lung repair and to restore epithelial integrity. Thus, targeting IL-13 inhibits fibrotic processes and enhances repair processes in the lung.


Fibrogenesis & Tissue Repair | 2013

Matrix regulation of idiopathic pulmonary fibrosis: the role of enzymes

Deborah Lee Clarke; Alan Carruthers; Tomas Mustelin; Lynne A. Murray

Repairing damaged tissues is an essential homeostatic mechanism that enables clearance of dead or damaged cells after injury, and the maintenance of tissue integrity. However, exaggeration of this process in the lung can lead to the development of fibrotic scar tissue. This is characterized by excessive accumulation of extracellular matrix (ECM) components such as fibronectin, proteoglycans, hyaluronic acid, and interstitial collagens. After tissue injury, or a breakdown of tissue integrity, a cascade of events unfolds to maintain normal tissue homeostasis. Inflammatory mediators are released from injured epithelium, leading to both platelet activation and inflammatory cell migration. Inflammatory cells are capable of releasing multiple pro-inflammatory and fibrogenic mediators such as transforming growth factor (TGF)β and interleukin (IL)-13, which can trigger myofibroblast proliferation and recruitment. The myofibroblast population is also expanded as a result of epithelial cells undergoing epithelial-to-mesenchymal transition and of the activation of resident fibroblasts, leading to ECM deposition and tissue remodeling. In the healthy lung, wound healing then proceeds to restore the normal architecture of the lung; however, fibrosis can develop when the wound is severe, the tissue injury persists, or the repair process becomes dysregulated. Understanding the processes regulating aberrant wound healing and the matrix in the chronic fibrotic lung disease idiopathic pulmonary fibrosis (IPF), is key to identifying new treatments for this chronic debilitating disease. This review focuses primarily on the emerging role of enzymes in the lungs of patients with IPF. Elevated expression of a number of enzymes that can directly modulate the ECM has been reported, and recent data indicates that modulating the activity of these enzymes can have a downstream effect on fibrotic tissue remodeling.


American Journal of Respiratory Cell and Molecular Biology | 2010

Human Lung Parenchyma but Not Proximal Bronchi Produces Fibroblasts with Enhanced TGF-β Signaling and α-SMA Expression

Dmitri V. Pechkovsky; Tillie L. Hackett; Steven S. An; Furquan Shaheen; Lynne A. Murray; Darryl A. Knight

Given the contribution various fibroblast subsets make to wound healing and tissue remodeling, the concept of lung fibroblast heterogeneity is of great interest. However, the mechanisms contributing to this heterogeneity are unknown. To this aim, we compared molecular and biophysical characteristics of fibroblasts concurrently isolated from normal human proximal bronchi (B-FBR) and distal lung parenchyma (P-FBR). Using quantitative RT-PCR, spontaneous expression of more than 30 genes related to repair and remodeling was analyzed. All P-FBR lines demonstrated significantly increased basal α-smooth muscle actin (α-SMA) mRNA and protein expression levels when compared with donor-matched B-FBR. These differences were not associated with sex, age, or disease history of lung tissue donors. In contrast to B-FBR, P-FBR displayed enhanced transforming growth factor (TGF)-β/Smad signaling at baseline, and inhibition of either ALK-5 or neutralization of endogenously produced and activated TGF-β substantially decreased basal α-SMA protein in P-FBR. Both B-FBR and P-FBR up-regulated α-SMA after stimulation with TGF-β1, and basal expression levels of TGF-β1, TGF-βRI, and TGF-βRII were not significantly different between fibroblast pairs. Blockade of metalloproteinase-dependent activation of endogenous TGF-β did not significantly modify α-SMA expression in P-FBR. However, resistance to mechanical tension of these cells was significantly higher in comparison with B-FBR, and added TGF-β1 significantly increased stiffness of both cell monolayers. Our data suggest that in contrast with human normal bronchial tissue explants, lung parenchyma produces mesenchymal cells with a myofibroblastic phenotype by intrinsic mechanisms of TGF-β activation in feed-forward manner. These results also offer a new insight into mechanisms of human fibroblast heterogeneity and their function in the airway and lung tissue repair and remodeling.


Science Translational Medicine | 2014

Selective Targeting of TGF-β Activation to Treat Fibroinflammatory Airway Disease

Shunsuke Minagawa; Jianlong Lou; Robert Seed; Anthony Cormier; Shenping Wu; Yifan Cheng; Lynne A. Murray; Ping Tsui; Jane Connor; Ronald Herbst; Cedric Govaerts; Tyren Barker; Stephanie Cambier; Haruhiko Yanagisawa; Amanda Goodsell; Mitsuo Hashimoto; Oliver J. Brand; Ran Cheng; Royce Ma; Kate J. McKnelly; W. Wen; Arthur Hill; David M. Jablons; Paul J. Wolters; Hideya Kitamura; Jun Araya; Andrea J. Barczak; David J. Erle; Louis F. Reichardt; James D. Marks

Therapeutic targeting of an extended-closed conformation of the integrin αvβ8 inhibits TGF-β activation and ameliorates symptoms of experimental airway disease in mice. Breathing Freely Narrowing of the airways through accumulation of scar tissue and inflammation results from chronic injury in common diseases such as chronic obstructive pulmonary disease (COPD) and severe chronic asthma. Such airway narrowing causes the obstruction responsible for the breathlessness that these patients experience, and there are no available treatments that ameliorate fibroinflammatory airway narrowing. In a new study, Minagawa et al. engineered a monoclonal antibody that locks in a specific inactive conformation of a protein named integrin αvβ8. This protein is a crucial receptor required for activation of transforming growth factor–β, a central mediator of pathological inflammation and fibrosis. This antibody, when administered to mice engineered to express only human and not mouse αvβ8, reduced airway inflammation and fibrosis in response to a variety of injurious agents including cigarette smoke and allergens that are involved in the pathogenesis of COPD. Airway remodeling, caused by inflammation and fibrosis, is a major component of chronic obstructive pulmonary disease (COPD) and currently has no effective treatment. Transforming growth factor–β (TGF-β) has been widely implicated in the pathogenesis of airway remodeling in COPD. TGF-β is expressed in a latent form that requires activation. The integrin αvβ8 (encoded by the itgb8 gene) is a receptor for latent TGF-β and is essential for its activation. Expression of integrin αvβ8 is increased in airway fibroblasts in COPD and thus is an attractive therapeutic target for the treatment of airway remodeling in COPD. We demonstrate that an engineered optimized antibody to human αvβ8 (B5) inhibited TGF-β activation in transgenic mice expressing only human and not mouse ITGB8. The B5 engineered antibody blocked fibroinflammatory responses induced by tobacco smoke, cytokines, and allergens by inhibiting TGF-β activation. To clarify the mechanism of action of B5, we used hydrodynamic, mutational, and electron microscopic methods to demonstrate that αvβ8 predominantly adopts a constitutively active, extended-closed headpiece conformation. Epitope mapping and functional characterization of B5 revealed an allosteric mechanism of action due to locking-in of a low-affinity αvβ8 conformation. Collectively, these data demonstrate a new model for integrin function and present a strategy to selectively target the TGF-β pathway to treat fibroinflammatory airway diseases.


Current Drug Targets - Inflammation & Allergy | 2005

The role of CXCR2/CXCR2 ligands in acute lung injury.

Robert M. Strieter; Michael P. Keane; Marie D. Burdick; Ammar Sakkour; Lynne A. Murray; John A. Belperio

The mortality of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) remain high despite advances in our knowledge and intensive care. This supports the contention that we need to further our understanding of the mediators that are involved in the pathogenesis of ARDS. The pathogenesis of ARDS proceeds as a continuum from exudation and inflammation to a fibroproliferative phase of diffuse alveolar damage. While a number of mediators are involved in the pathogenesis of ARDS, members of the CXC chemokine family have been determined to play a critical and pleiotropic role in promoting both recruitment of inflammatory cells, as well as in mediating aberrant vascular remodeling during both phases of ARDS. The importance of the biology of CXC chemokines in ALI/ARDS will be discussed in this review.

Collaboration


Dive into the Lynne A. Murray's collaboration.

Top Co-Authors

Avatar

Cory M. Hogaboam

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana Lucia Coelho

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge