Network


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

Hotspot


Dive into the research topics where Gregory P. Cosgrove is active.

Publication


Featured researches published by Gregory P. Cosgrove.


Nature Genetics | 2013

Genome-wide association study identifies multiple susceptibility loci for pulmonary fibrosis

Tasha E. Fingerlin; Elissa Murphy; Weiming Zhang; Anna L. Peljto; Kevin K. Brown; Mark P. Steele; James E. Loyd; Gregory P. Cosgrove; David A. Lynch; Steve D. Groshong; Harold R. Collard; Paul J. Wolters; Williamson Ziegler Bradford; Karl Kossen; Scott D. Seiwert; Roland M. du Bois; Christine Kim Garcia; Megan S. Devine; Gunnar Gudmundsson; Helgi J. Ísaksson; Naftali Kaminski; Yingze Zhang; Kevin F. Gibson; Lisa H. Lancaster; Joy D. Cogan; Wendi R. Mason; Toby M. Maher; Philip L. Molyneaux; Athol U. Wells; Miriam F. Moffatt

We performed a genome-wide association study of non-Hispanic, white individuals with fibrotic idiopathic interstitial pneumonias (IIPs; n = 1,616) and controls (n = 4,683), with follow-up replication analyses in 876 cases and 1,890 controls. We confirmed association with TERT at 5p15, MUC5B at 11p15 and the 3q26 region near TERC, and we identified seven newly associated loci (Pmeta = 2.4 × 10−8 to 1.1 × 10−19), including FAM13A (4q22), DSP (6p24), OBFC1 (10q24), ATP11A (13q34), DPP9 (19p13) and chromosomal regions 7q22 and 15q14-15. Our results suggest that genes involved in host defense, cell-cell adhesion and DNA repair contribute to risk of fibrotic IIPs.


Nature Medicine | 2010

Rtp801, a suppressor of mTOR signaling, is an essential mediator of cigarette smoke-induced pulmonary injury and emphysema

Toshinori Yoshida; Igor Mett; Anil K. Bhunia; Joel Bowman; Mario J. Perez; Li Zhang; Aneta Gandjeva; Lijie Zhen; Ugonma Chukwueke; Tianzhi Mao; Amy Richter; Emile N. Brown; Hagit Ashush; Natalie Notkin; Anna Gelfand; Rajesh K. Thimmulappa; Tirumalai Rangasamy; Thomas E. Sussan; Gregory P. Cosgrove; Majd Mouded; Steven D. Shapiro; Irina Petrache; Shyam Biswal; Elena Feinstein; Rubin M. Tuder

Rtp801 (also known as Redd1, and encoded by Ddit4), a stress-related protein triggered by adverse environmental conditions, inhibits mammalian target of rapamycin (mTOR) by stabilizing the TSC1-TSC2 inhibitory complex and enhances oxidative stress–dependent cell death. We postulated that Rtp801 acts as a potential amplifying switch in the development of cigarette smoke–induced lung injury, leading to emphysema. Rtp801 mRNA and protein were overexpressed in human emphysematous lungs and in lungs of mice exposed to cigarette smoke. The regulation of Rtp801 expression by cigarette smoke may rely on oxidative stress–dependent activation of the CCAAT response element in its promoter. We also found that Rtp801 was necessary and sufficient for nuclear factor-κB (NF-κB) activation in cultured cells and, when forcefully expressed in mouse lungs, it promoted NF-κB activation, alveolar inflammation, oxidative stress and apoptosis of alveolar septal cells. In contrast, Rtp801 knockout mice were markedly protected against acute cigarette smoke–induced lung injury, partly via increased mTOR signaling, and, when exposed chronically to cigarette smoke, against emphysema. Our data support the notion that Rtp801 may represent a major molecular sensor and mediator of cigarette smoke–induced lung injury.


The Journal of Rheumatology | 2013

Mycophenolate Mofetil Improves Lung Function in Connective Tissue Disease-associated Interstitial Lung Disease

Aryeh Fischer; Brown Kk; Du Bois Rm; Stephen K. Frankel; Gregory P. Cosgrove; Evans R. Fernandez-Perez; Tristan J. Huie; Krishnamoorthy M; Richard T. Meehan; Joshua J. Solomon; Jeffrey J. Swigris

Objective. Small series suggest mycophenolate mofetil (MMF) is well tolerated and may be an effective therapy for connective tissue disease-associated interstitial lung disease (CTD-ILD). We examined the tolerability and longitudinal changes in pulmonary physiology in a large and diverse cohort of patients with CTD-ILD treated with MMF. Methods. We identified consecutive patients evaluated at our center between January 2008 and January 2011 and prescribed MMF for CTD-ILD. We assessed safety and tolerability of MMF and used longitudinal data analyses to examine changes in pulmonary physiology over time, before and after initiation of MMF. Results. We identified 125 subjects treated with MMF for a median 897 days. MMF was discontinued in 13 subjects. MMF was associated with significant improvements in estimated percentage of predicted forced vital capacity (FVC%) from MMF initiation to 52, 104, and 156 weeks (4.9% ± 1.9%, p = 0.01; 6.1% ± 1.8%, p = 0.0008; and 7.3% ± 2.6%, p = 0.004, respectively); and in estimated percentage predicted diffusing capacity (DLCO%) from MMF initiation to 52 and 104 weeks (6.3% ± 2.8%, p = 0.02; 7.1% ± 2.8%, p = 0.01). In the subgroup without usual interstitial pneumonia (UIP)-pattern injury, MMF significantly improved FVC% and DLCO%, and in the subgroup with UIP-pattern injury, MMF was associated with stability in FVC% and DLCO%. Conclusion. In a large diverse cohort of CTD-ILD, MMF was well tolerated and had a low rate of discontinuation. Treatment with MMF was associated with either stable or improved pulmonary physiology over a median 2.5 years of followup. MMF appears to be a promising therapy for the spectrum of CTD-ILD.


American Journal of Respiratory and Critical Care Medicine | 2014

Future Directions in Idiopathic Pulmonary Fibrosis Research. An NHLBI Workshop Report

Timothy S. Blackwell; Andrew M. Tager; Zea Borok; Bethany B. Moore; David A. Schwartz; Kevin J. Anstrom; Ziv Bar-Joseph; Peter B. Bitterman; Michael R. Blackburn; William Bradford; Kevin K. Brown; Harold A. Chapman; Harold R. Collard; Gregory P. Cosgrove; Robin R. Deterding; Ramona Doyle; Kevin R. Flaherty; Christine Kim Garcia; James S. Hagood; Craig A. Henke; Erica L. Herzog; Cory M. Hogaboam; Jeffrey C. Horowitz; Talmadge E. King; James E. Loyd; William Lawson; Clay B. Marsh; Paul W. Noble; Imre Noth; Dean Sheppard

The median survival of patients with idiopathic pulmonary fibrosis (IPF) continues to be approximately 3 years from the time of diagnosis, underscoring the lack of effective medical therapies for this disease. In the United States alone, approximately 40,000 patients die of this disease annually. In November 2012, the NHLBI held a workshop aimed at coordinating research efforts and accelerating the development of IPF therapies. Basic, translational, and clinical researchers gathered with representatives from the NHLBI, patient advocacy groups, pharmaceutical companies, and the U.S. Food and Drug Administration to review the current state of IPF research and identify priority areas, opportunities for collaborations, and directions for future research. The workshop was organized into groups that were tasked with assessing and making recommendations to promote progress in one of the following six critical areas of research: (1) biology of alveolar epithelial injury and aberrant repair; (2) role of extracellular matrix; (3) preclinical modeling; (4) role of inflammation and immunity; (5) genetic, epigenetic, and environmental determinants; (6) translation of discoveries into diagnostics and therapeutics. The workshop recommendations provide a basis for directing future research and strategic planning by scientific, professional, and patient communities and the NHLBI.


Journal of Immunology | 2007

Pulmonary stromal-derived factor-1 expression and effect on neutrophil recruitment during acute lung injury

Joseph M. Petty; Viranuj Sueblinvong; Christopher C. Lenox; Christine C. Jones; Gregory P. Cosgrove; Carlyne D. Cool; Pradeep R. Rai; Kevin K. Brown; Daniel J. Weiss; Matthew E. Poynter; Benjamin T. Suratt

The severe and protracted inflammation that characterizes acute lung injury (ALI) is driven by the ongoing recruitment of neutrophils to the lung. Although much of the cytokine signaling responsible for the initial phase of ALI has been elaborated, relatively little is known about the mechanisms governing the recruitment of neutrophils from the bone marrow to the lung in the later period of this disease. Given its previously described chemoattractant effects on marrow neutrophils, we investigated whether stromal-derived factor-1 (SDF-1) (CXCL12) might participate in this later phase of recruitment. Using immunohistochemistry to examine both banked human lung specimens from patients with ALI and lungs from mice with LPS-induced pneumonitis, we found that pulmonary SDF-1 expression increases during ALI. We further determined that both lung SDF-1 protein expression and mRNA expression rise in a delayed but sustained pattern in this mouse model and that the major source of the increase in expression appears to be the lung epithelium. Lastly, we found that expression of the SDF-1 receptor CXCR4 rises in a similar temporal pattern on neutrophils in both the blood and airspace of LPS-injured mice and that Ab-mediated SDF-1 blockade significantly attenuates late but not early pulmonary neutrophilia in this model. These results implicate SDF-1 in neutrophil recruitment to the lung in the later period of acute lung injury and suggest a novel role for this cytokine in coordinating the transition from the inflammatory response to the initiation of tissue repair.


PLOS ONE | 2013

The Idiopathic Pulmonary Fibrosis Honeycomb Cyst Contains A Mucocilary Pseudostratified Epithelium

Max A. Seibold; Russell W. Smith; Cydney Urbanek; Steve D. Groshong; Gregory P. Cosgrove; Kevin K. Brown; Marvin I. Schwarz; David A. Schwartz; Susan D. Reynolds

Background We previously identified a MUC5B gene promoter-variant that is a risk allele for sporadic and familial Idiopathic Pulmonary Fibrosis/Usual Interstitial Pneumonia (IPF/UIP). This allele was strongly associated with increased MUC5B gene expression in lung tissue from unaffected subjects. Despite the strong association of this airway epithelial marker with disease, little is known of mucin expressing structures or of airway involvement in IPF/UIP. Methods Immunofluorescence was used to subtype mucus cells according to MUC5B and MUC5AC expression and to identify ciliated, basal, and alveolar type II (ATII) cells in tissue sections from control and IPF/UIP subjects. Staining patterns were quantified for distal airways (Control and IPF/UIP) and in honeycomb cysts (HC). Results MUC5B-expressing cells (EC) were detected in the majority of control distal airways. MUC5AC-EC were identified in half of these airways and only in airways that contained MUC5B-EC. The frequency of MUC5B+ and MUC5AC+ distal airways was increased in IPF/UIP subjects. MUC5B-EC were the dominant mucus cell type in the HC epithelium. The distal airway epithelium from control and IPF/UIP subjects and HC was populated by basal and ciliated cells. Most honeycombing regions were distinct from ATII hyperplasic regions. ATII cells were undetectable in the overwhelming majority of HC. Conclusions The distal airway contains a pseudostratified mucocilary epithelium that is defined by basal epithelial cells and mucus cells that express MUC5B predominantly. These data suggest that the HC is derived from the distal airway.


Thorax | 2013

Expression of cilium-associated genes defines novel molecular subtypes of idiopathic pulmonary fibrosis

Yang; Christopher D. Coldren; Sonia M. Leach; Max A. Seibold; Elissa Murphy; Jia Lin; Rosen R; Neidermyer Aj; David F. McKean; Steve D. Groshong; Carlyne D. Cool; Gregory P. Cosgrove; David A. Lynch; Kevin K. Brown; Marvin I. Schwarz; Tasha E. Fingerlin; David A. Schwartz

Background Idiopathic pulmonary fibrosis (IPF) is an untreatable lung disease with a median survival of only 3–5 years that is diagnosed using a combination of clinical, radiographic and pathologic criteria. Histologically, IPF is characterised by usual interstitial pneumonia (UIP), a fibrosing interstitial pneumonia with a pattern of heterogeneous, subpleural regions of fibrotic and remodelled lung. We hypothesised that gene expression profiles of lung tissue may identify molecular subtypes of disease that could classify subtypes of IPF/UIP that have clinical implications. Methods and findings We collected transcriptional profiles on lung tissue from 119 patients with IPF/UIP and 50 non-diseased controls. Differential expression of individual transcripts was identified using an analysis of covariance (ANCOVA) model incorporating the clinical diagnosis of each patient as well as age, gender and smoking status. Validation was performed in an independent cohort of 111 IPF/UIP and 39 non-diseased controls. Our analysis identified two subtypes of IPF/UIP based on a strong molecular signature associated with expression of genes previously associated with fibrosis (matrix metalloproteinases, osteopontin, keratins), cilium genes and genes with unknown function. We demonstrate that elevated expression of cilium genes is associated with more extensive microscopic honeycombing and higher expression of both the airway mucin gene MUC5B and the metalloproteinase MMP7, a gene recently implicated in attenuating ciliated cell differentiation during wound repair. Conclusions Expression of cilium genes appears to identify two unique molecular phenotypes of IPF/UIP. The different molecular profiles may be relevant to therapeutic responsiveness in patients with IPF/UIP.


Chest | 2009

Heart Rate Recovery After 6-Min Walk Test Predicts Survival in Patients With Idiopathic Pulmonary Fibrosis

Jeffrey J. Swigris; Jeff Swick; Frederick S. Wamboldt; David Sprunger; Roland M. du Bois; Aryeh Fischer; Gregory P. Cosgrove; Stephen K. Frankel; Evans R. Fernandez-Perez; Dolly Kervitsky; Kevin K. Brown

BACKGROUND In patients with idiopathic pulmonary fibrosis (IPF), our objectives were to identify predictors of abnormal heart rate recovery (HRR) at 1 min after completion of a 6-min walk test (6MWT) [HRR1] and 2 min after completion of a 6MWT (HRR2), and to determine whether abnormal HRR predicts mortality. METHODS From 2003 to 2008, we identified IPF patients who had been evaluated at our center (n = 76) with a pulmonary physiologic examination and the 6MWT. We used logistic regression to identify predictors of abnormal HRR, the product-limit method to compare survival in the sample stratified on HRR, and Cox proportional hazards analysis to estimate the prognostic capability of abnormal HRR. RESULTS Cutoff values were 13 beats for abnormal HRR1 and 22 beats for HRR2. In a multivariable model, predictors of abnormal HRR1 were diffusing capacity of the lung for carbon monoxide (odds ratio [OR], 0.4 per 10% predicted; 95% confidence interval [CI], 0.2 to 0.7; p = 0.003), change in heart rate from baseline to maximum (OR, 0.9; 95% CI, 0.8 to 0.97; p = 0.01), and having a right ventricular systolic pressure > 35 mm Hg as determined by transthoracic echocardiogram (OR, 12.7; 95% CI, 2.0 to 79.7; p = 0.01). Subjects with an abnormal HRR had significantly worse survival than subjects with a normal HRR (for HRR1, p = 0.0007 [log-rank test]; for HRR2, p = 0.03 [log-rank test]); these results held for the subgroup of 30 subjects without resting pulmonary hypertension (HRR1, p = 0.04 [log-rank test]). Among several candidate variables, abnormal HRR1 appeared to be the most potent predictor of mortality (hazard ratio, 5.2; 95% CI, 1.8 to 15.2; p = 0.004). CONCLUSION Abnormal HRR after 6MWT predicts mortality in IPF patients. Research is needed to confirm these findings prospectively and to examine the mechanisms of HRR in IPF patients.


Respirology | 2010

A detailed evaluation of acute respiratory decline in patients with fibrotic lung disease: aetiology and outcomes.

Tristan J. Huie; Amy L. Olson; Gregory P. Cosgrove; William J. Janssen; Abigail R. Lara; David A. Lynch; Steve D. Groshong; Marc Moss; Marvin I. Schwarz; Kevin K. Brown; Stephen K. Frankel

Background and objective:  A comprehensive diagnostic evaluation is recommended for all patients with fibrotic lung disease and acute respiratory decompensation. However, the effect on clinical outcomes of this evaluation remains unknown.


Journal of Immunology | 2011

Increased Cell Surface Fas Expression Is Necessary and Sufficient To Sensitize Lung Fibroblasts to Fas Ligation-Induced Apoptosis: Implications for Fibroblast Accumulation in Idiopathic Pulmonary Fibrosis

Murry W. Wynes; Benjamin L. Edelman; Amanda G. Kostyk; Michael G. Edwards; Christopher D. Coldren; Steve D. Groshong; Gregory P. Cosgrove; Elizabeth F. Redente; Alison Bamberg; Kevin K. Brown; Nichole Reisdorph; Rebecca C. Keith; Stephen K. Frankel; David W. H. Riches

Idiopathic pulmonary fibrosis (IPF) is associated with the accumulation of collagen-secreting fibroblasts and myofibroblasts in the lung parenchyma. Many mechanisms contribute to their accumulation, including resistance to apoptosis. In previous work, we showed that exposure to the proinflammatory cytokines TNF-α and IFN-γ reverses the resistance of lung fibroblasts to apoptosis. In this study, we investigate the underlying mechanisms. Based on an interrogation of the transcriptomes of unstimulated and TNF-α– and IFN-γ–stimulated primary lung fibroblasts and the lung fibroblast cell line MRC5, we show that among Fas-signaling pathway molecules, Fas expression was increased ∼6-fold in an NF-κB– and p38mapk-dependent fashion. Prevention of the increase in Fas expression using Fas small interfering RNAs blocked the ability of TNF-α and IFN-γ to sensitize fibroblasts to Fas ligation-induced apoptosis, whereas enforced adenovirus-mediated Fas overexpression was sufficient to overcome basal resistance to Fas-induced apoptosis. Examination of lung tissues from IPF patients revealed low to absent staining of Fas in fibroblastic cells of fibroblast foci. Collectively, these findings suggest that increased expression of Fas is necessary and sufficient to overcome the resistance of lung fibroblasts to Fas-induced apoptosis. Our findings also suggest that approaches aimed at increasing Fas expression by lung fibroblasts and myofibroblasts may be therapeutically relevant in IPF.

Collaboration


Dive into the Gregory P. Cosgrove's collaboration.

Top Co-Authors

Avatar

Kevin K. Brown

University of Washington

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steve D. Groshong

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Marvin I. Schwarz

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

David A. Schwartz

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Jeffrey J. Swigris

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Aryeh Fischer

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Carlyne D. Cool

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

David A. Lynch

University of California

View shared research outputs
Top Co-Authors

Avatar

Ivana V. Yang

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge