Network


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

Hotspot


Dive into the research topics where Mark P. Steele is active.

Publication


Featured researches published by Mark P. Steele.


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.


JAMA | 2013

Association Between the MUC5B Promoter Polymorphism and Survival in Patients With Idiopathic Pulmonary Fibrosis

Anna L. Peljto; Yingze Zhang; Tasha E. Fingerlin; Shwu-Fan Ma; Joe G. N. Garcia; Thomas J. Richards; Lori J. Silveira; Kathleen O. Lindell; Mark P. Steele; James E. Loyd; Kevin F. Gibson; Max A. Seibold; Kevin K. Brown; Janet Talbert; Cheryl Markin; Karl Kossen; Scott D. Seiwert; Elissa Murphy; Imre Noth; Marvin I. Schwarz; Naftali Kaminski; David A. Schwartz

IMPORTANCEnCurrent prediction models of mortality in idiopathic pulmonary fibrosis (IPF), which are based on clinical and physiological parameters, have modest value in predicting which patients will progress. In addition to the potential for improving prognostic models, identifying genetic and molecular features that are associated with IPF mortality may provide insight into the underlying mechanisms of disease and inform clinical trials.nnnOBJECTIVEnTo determine whether the MUC5B promoter polymorphism (rs35705950), previously reported to be associated with the development of pulmonary fibrosis, is associated with survival in IPF.nnnDESIGN, SETTING, AND PARTICIPANTSnRetrospective study of survival in 2 independent cohorts of patients with IPF: the INSPIRE cohort, consisting of patients enrolled in the interferon-γ1b trial (n = 438; December 15, 2003-May 2, 2009; 81 centers in 7 European countries, the United States, and Canada), and the Chicago cohort, consisting of IPF participants recruited from the Interstitial Lung Disease Clinic at the University of Chicago (n = 148; 2007-2010). The INSPIRE cohort was used to model the association of the MUC5B genotype with survival, accounting for the effect of matrix metalloproteinase 7 (MMP-7) blood concentration and other demographic and clinical covariates. The Chicago cohort was used for replication of findings.nnnMAIN OUTCOMES AND MEASURESnThe primary end point was all-cause mortality.nnnRESULTSnThe numbers of patients in the GG, GT, and TT genotype groups were 148 (34%), 259 (59%), and 31 (7%), respectively, in the INSPIRE cohort and 41 (28%), 98 (66%), and 9 (6%), respectively, in the Chicago cohort. The median follow-up period was 1.6 years for INSPIRE and 2.1 years for Chicago. During follow-up, there were 73 deaths (36 GG, 35 GT, and 2 TT) among INSPIRE patients and 64 deaths (26 GG, 36 GT, and 2 TT) among Chicago patients. The unadjusted 2-year cumulative incidence of death was lower among patients carrying 1 or more copies of the IPF risk allele (T) in both the INSPIRE cohort (0.25 [95% CI, 0.17-0.32] for GG, 0.17 [95% CI, 0.11-0.23] for GT, and 0.03 [95% CI, 0.00-0.09] for TT) and the Chicago cohort (0.50 [95% CI, 0.31-0.63] for GG, 0.22 [95% CI, 0.13-0.31] for GT, and 0.11 [95% CI, 0.00-0.28] for TT). In the INSPIRE cohort, the TT and GT genotypes (risk for IPF) were associated with improved survival compared with GG (hazard ratios, 0.23 [95% CI, 0.10-0.52] and 0.48 [95% CI, 0.31-0.72], respectively; P < .001). This finding was replicated in the Chicago cohort (hazard ratios, 0.15 [95% CI, 0.05-0.49] and 0.39 [95% CI, 0.21-0.70], respectively; P < .002). The observed association of MUC5B with survival was independent of age, sex, forced vital capacity, diffusing capacity of carbon monoxide, MMP-7, and treatment status. The addition of the MUC5B genotype to the survival models significantly improved the predictive accuracy of the model in both the INSPIRE cohort (C = 0.71 [95% CI, 0.64-0.75] vs C = 0.68 [95% CI, 0.61-0.73]; P < .001) and the Chicago cohort (C = 0.73 [95% CI, 0.62-0.78] vs C = 0.69 [95% CI, 0.59-0.75]; P = .01).nnnCONCLUSIONS AND RELEVANCEnAmong patients with IPF, a common risk polymorphism in MUC5B was significantly associated with improved survival. Further research is necessary to refine the risk estimates and to determine the clinical implications of these findings.


American Journal of Respiratory and Critical Care Medicine | 2015

RARE VARIANTS IN RTEL1 ARE ASSOCIATED WITH FAMILIAL INTERSTITIAL PNEUMONIA

Joy D. Cogan; Jonathan A. Kropski; Min Zhao; Daphne B. Mitchell; Lynette Rives; Cheryl Markin; Errine T. Garnett; Keri H. Montgomery; Wendi R. Mason; David F. McKean; Julia Powers; Elissa Murphy; Lana M. Olson; Leena Choi; Dong-Sheng Cheng; Elizabeth Blue; Lisa R. Young; Lisa H. Lancaster; Mark P. Steele; Kevin K. Brown; Marvin I. Schwarz; Tasha E. Fingerlin; David A. Schwartz; William Lawson; James E. Loyd; Zhongming Zhao; John A. Phillips; Timothy S. Blackwell

RATIONALEnUp to 20% of cases of idiopathic interstitial pneumonia cluster in families, comprising the syndrome of familial interstitial pneumonia (FIP); however, the genetic basis of FIP remains uncertain in most families.nnnOBJECTIVESnTo determine if new disease-causing rare genetic variants could be identified using whole-exome sequencing of affected members from FIP families, providing additional insights into disease pathogenesis.nnnMETHODSnAffected subjects from 25 kindreds were selected from an ongoing FIP registry for whole-exome sequencing from genomic DNA. Candidate rare variants were confirmed by Sanger sequencing, and cosegregation analysis was performed in families, followed by additional sequencing of affected individuals from another 163 kindreds.nnnMEASUREMENTS AND MAIN RESULTSnWe identified a potentially damaging rare variant in the gene encoding for regulator of telomere elongation helicase 1 (RTEL1) that segregated with disease and was associated with very short telomeres in peripheral blood mononuclear cells in 1 of 25 families in our original whole-exome sequencing cohort. Evaluation of affected individuals in 163 additional kindreds revealed another eight families (4.7%) with heterozygous rare variants in RTEL1 that segregated with clinical FIP. Probands and unaffected carriers of these rare variants had short telomeres (<10% for age) in peripheral blood mononuclear cells and increased T-circle formation, suggesting impaired RTEL1 function.nnnCONCLUSIONSnRare loss-of-function variants in RTEL1 represent a newly defined genetic predisposition for FIP, supporting the importance of telomere-related pathways in pulmonary fibrosis.


PLOS ONE | 2013

Bronchoscopic Cryobiopsy for the Diagnosis of Diffuse Parenchymal Lung Disease

Jonathan A. Kropski; Jason M. Pritchett; Wendi R. Mason; Lakshmi Sivarajan; Linda A. Gleaves; Joyce E. Johnson; Lisa H. Lancaster; William Lawson; Timothy S. Blackwell; Mark P. Steele; James E. Loyd; Otis B. Rickman

Background Although in some cases clinical and radiographic features may be sufficient to establish a diagnosis of diffuse parenchymal lung disease (DPLD), surgical lung biopsy is frequently required. Recently a new technique for bronchoscopic lung biopsy has been developed using flexible cryo-probes. In this study we describe our clinical experience using bronchoscopic cryobiopsy for diagnosis of diffuse lung disease. Methods A retrospective study of subjects who had undergone bronchoscopic cryobiopsy for evaluation of DPLD at an academic tertiary care center from January 1, 2012 through January 15, 2013 was performed. The procedure was performed using a flexible bronchoscope to acquire biopsies of lung parenchyma. H&E stained biopsies were reviewed by an expert lung pathologist. Results Twenty-five eligible subjects were identified. With a mean area of 64.2 mm2, cryobiopsies were larger than that typically encountered with traditional transbronchial forceps biopsy. In 19 of the 25 subjects, a specific diagnosis was obtained. In one additional subject, biopsies demonstrating normal parenchyma were felt sufficient to exclude diffuse lung disease as a cause of dyspnea. The overall diagnostic yield of bronchoscopic cryobiopsy was 80% (20/25). The most frequent diagnosis was usual interstitial pneumonia (UIP) (nu200a=u200a7). Three of the 25 subjects ultimately required surgical lung biopsy. There were no significant complications. Conclusion In patients with suspected diffuse parenchymal lung disease, bronchoscopic cryobiopsy is a promising and minimally invasive approach to obtain lung tissue with high diagnostic yield.


Annual Review of Medicine | 2013

Molecular Mechanisms in Progressive Idiopathic Pulmonary Fibrosis

Mark P. Steele; David A. Schwartz

There is clear evidence that environmental exposures and genetic predisposition contribute to the pathogenesis of idiopathic pulmonary fibrosis (IPF). Cigarette smoking increases the risk of developing IPF several-fold, as do other exposures such as metal-fume and wood-dust exposure. Occupations that increase the risk of IPF are agricultural work, hairdressing, and stone polishing, supporting the role of environmental exposure in disease pathogenesis. Genetic predisposition to IPF is evident from its familial aggregation and the fact that pulmonary fibrosis develops in several rare genetic disorders. Mutations in surfactant proteins lead to pulmonary fibrosis and are associated with endoplasmic reticulum stress in alveolar type II epithelial cells. Mutations in telomerase have been found in several families with IPF, and shortened telomeres are found in sporadic cases of IPF. A common variant in mucin 5B predisposes to both familial and sporadic IPF and is present in the majority of cases, indicating sporadic IPF occurs in those with genetic predisposition.


American Journal of Respiratory and Critical Care Medicine | 2015

Extensive Phenotyping of Individuals at Risk for Familial Interstitial Pneumonia Reveals Clues to the Pathogenesis of Interstitial Lung Disease

Jonathan A. Kropski; Jason M. Pritchett; Donald F. Zoz; Peter F. Crossno; Cheryl Markin; Errine T. Garnett; Amber L. Degryse; Daphne B. Mitchell; Vasiliy V. Polosukhin; Otis B. Rickman; Leena Choi; Dong Sheng Cheng; Melinda E. McConaha; Brittany R. Jones; Linda A. Gleaves; Frank B. McMahon; John A. Worrell; Joseph F. Solus; Lorraine B. Ware; Jae-Woo Lee; Pierre P. Massion; Rinat Zaynagetdinov; Eric S. White; Jonathan D. Kurtis; Joyce E. Johnson; Steve D. Groshong; Lisa H. Lancaster; Lisa R. Young; Mark P. Steele; John A. Phillips

RATIONALEnAsymptomatic relatives of patients with familial interstitial pneumonia (FIP), the inherited form of idiopathic interstitial pneumonia, carry increased risk for developing interstitial lung disease.nnnOBJECTIVESnStudying these at-risk individuals provides a unique opportunity to investigate early stages of FIP pathogenesis and develop predictive models of disease onset.nnnMETHODSnSeventy-five asymptomatic first-degree relatives of FIP patients (mean age, 50.8 yr) underwent blood sampling and high-resolution chest computed tomography (HRCT) scanning in an ongoing cohort study; 72 consented to bronchoscopy with bronchoalveolar lavage (BAL) and transbronchial biopsies. Twenty-seven healthy individuals were used as control subjects.nnnMEASUREMENTS AND MAIN RESULTSnEleven of 75 at-risk subjects (14%) had evidence of interstitial changes by HRCT, whereas 35.2% had abnormalities on transbronchial biopsies. No differences were noted in inflammatory cells in BAL between at-risk individuals and control subjects. At-risk subjects had increased herpesvirus DNA in cell-free BAL and evidence of herpesvirus antigen expression in alveolar epithelial cells (AECs), which correlated with expression of endoplasmic reticulum stress markers in AECs. Peripheral blood mononuclear cell and AEC telomere length were shorter in at-risk individuals than healthy control subjects. The minor allele frequency of the Muc5B rs35705950 promoter polymorphism was increased in at-risk subjects. Levels of several plasma biomarkers differed between at-risk subjects and control subjects, and correlated with abnormal HRCT scans.nnnCONCLUSIONSnEvidence of lung parenchymal remodeling and epithelial dysfunction was identified in asymptomatic individuals at risk for FIP. Together, these findings offer new insights into the early pathogenesis of idiopathic interstitial pneumonia and provide an ongoing opportunity to characterize presymptomatic abnormalities that predict progression to clinical disease.


PLOS ONE | 2012

The peripheral blood transcriptome identifies the presence and extent of disease in idiopathic pulmonary fibrosis.

Ivana V. Yang; Leah Luna; Jennifer Cotter; Janet Talbert; Sonia M. Leach; Raven Kidd; Julia Turner; Nathan Kummer; Dolly Kervitsky; Kevin K. Brown; Kathy Boon; Marvin I. Schwarz; David A. Schwartz; Mark P. Steele

Rationale Peripheral blood biomarkers are needed to identify and determine the extent of idiopathic pulmonary fibrosis (IPF). Current physiologic and radiographic prognostic indicators diagnose IPF too late in the course of disease. We hypothesize that peripheral blood biomarkers will identify disease in its early stages, and facilitate monitoring for disease progression. Methods Gene expression profiles of peripheral blood RNA from 130 IPF patients were collected on Agilent microarrays. Significance analysis of microarrays (SAM) with a false discovery rate (FDR) of 1% was utilized to identify genes that were differentially-expressed in samples categorized based on percent predicted DLCO and FVC. Main Measurements and Results At 1% FDR, 1428 genes were differentially-expressed in mild IPF (DLCO >65%) compared to controls and 2790 transcripts were differentially- expressed in severe IPF (DLCO >35%) compared to controls. When categorized by percent predicted DLCO, SAM demonstrated 13 differentially-expressed transcripts between mild and severe IPF (< 5% FDR). These include CAMP, CEACAM6, CTSG, DEFA3 and A4, OLFM4, HLTF, PACSIN1, GABBR1, IGHM, and 3 unknown genes. Principal component analysis (PCA) was performed to determine outliers based on severity of disease, and demonstrated 1 mild case to be clinically misclassified as a severe case of IPF. No differentially-expressed transcripts were identified between mild and severe IPF when categorized by percent predicted FVC. Conclusions These results demonstrate that the peripheral blood transcriptome has the potential to distinguish normal individuals from patients with IPF, as well as extent of disease when samples were classified by percent predicted DLCO, but not FVC.


Chest | 2012

The Pulmonary Fibrosis-Associated MUC5B Promoter Polymorphism Does Not Influence the Development of Interstitial Pneumonia in Systemic Sclerosis

Anna L. Peljto; Mark P. Steele; Tasha E. Fingerlin; Monique Hinchcliff; Elissa Murphy; Sofia Podlusky; Mary Carns; Marvin I. Schwarz; John Varga; David A. Schwartz

BACKGROUNDnMore than 80% of patients with systemic sclerosis (SSc) develop lung involvement, most commonly interstitial pneumonia (IP). We recently identified a common variant in the promoter region of MUC5B (rs35705950) that has a significant effect on the risk of developing both familial and sporadic forms of IP. We hypothesized that this MUC5B promoter polymorphism is also associated with IP in subjects with SSc.nnnMETHODSnWe examined the minor allele frequency of the MUC5B polymorphism among 231 subjects with SSc, 109 with IP, and 122 without IP. IP diagnosis was confirmed by HRCT imaging and defined as the presence of reticular infiltrates and/or honeycomb cysts. FVC and diffusing capacity of the lung for carbon monoxide (Dlco) were also assessed.nnnRESULTSnWe found no association between IP and the MUC5B polymorphism among subjects with SSc (OR = 1.1, P = .80). The frequencies of the MUC5B polymorphism among subjects with SSc with IP (10.6%) and without IP (9.4%) were similar to the frequency observed in a population of unaffected control subjects (9.0%). In secondary analyses, we found the MUC5B polymorphism was not significantly associated with either FVC (P = .42) or Dlco (P = .06). No association with SSc-associated IP was found even when we used a more conservative definition of IP (FVC ≤ 70% and evidence of reticulations or honeycombing vs SSc FVC > 70% and no evidence of reticulation or honeycombing).nnnCONCLUSIONSnAlthough SSc-associated IP is clinically, radiologically, and histologically similar to other forms of IP, it appears to have distinct genetic risk factors. This study highlights the genetic and phenotypic heterogeneity of IP in general.


The Lancet Respiratory Medicine | 2015

Classification of usual interstitial pneumonia in patients with interstitial lung disease: assessment of a machine learning approach using high-dimensional transcriptional data

Su Yeon Kim; James Diggans; Dan Pankratz; Jing Huang; Moraima Pagan; Nicole Sindy; Ed Y. Tom; Jessica R. Anderson; Yoonha Choi; David A. Lynch; Mark P. Steele; Kevin R. Flaherty; Kevin K. Brown; Humam Farah; Michael J. Bukstein; Annie Pardo; Moisés Selman; Paul J. Wolters; Steven D. Nathan; Thomas V. Colby; Jeffrey L. Myers; Anna Luise A Katzenstein; Ganesh Raghu; Giulia C. Kennedy

BACKGROUNDnIdiopathic pulmonary fibrosis is a progressive fibrotic lung disease that distorts pulmonary architecture, leading to hypoxia, respiratory failure, and death. Diagnosis is difficult because other interstitial lung diseases have similar radiological and histopathological characteristics. A usual interstitial pneumonia pattern is a hallmark of idiopathic pulmonary fibrosis and is essential for its diagnosis. We aimed to develop a molecular test that distinguishes usual interstitial pneumonia from other interstitial lung diseases in surgical lung biopsy samples. The eventual goal of this research is to develop a method to diagnose idiopathic pulmonary fibrosis without the patient having to undergo surgery.nnnMETHODSnWe collected surgical lung biopsy samples from patients with various interstitial lung diseases at 11 hospitals in North America. Pathology diagnoses were confirmed by an expert panel. We measured RNA expression levels for 33u2008297 transcripts on microarrays in all samples. A classifier algorithm was trained on one set of samples and tested in a second set. We subjected a subset of samples to next-generation RNA sequencing (RNAseq) generating expression levels on 55u2008097 transcripts, and assessed a classifier trained on RNAseq data by cross-validation.nnnFINDINGSnWe took 125 surgical lung biopsies from 86 patients. 58 samples were identified by the expert panel as usual interstitial pneumonia, 23 as non-specific interstitial pneumonia, 16 as hypersensitivity pneumonitis, four as sarcoidosis, four as respiratory bronchiolitis, two as organising pneumonia, and 18 as subtypes other than usual interstitial pneumonia. The microarray classifier was trained on 77 samples and was assessed in a test set of 48 samples, for which it had a specificity of 92% (95% CI 81-100) and a sensitivity of 82% (64-95). Based on a subset of 36 samples, the RNAseq classifier had a specificity of 95% (84-100) and a sensitivity of 59% (35-82).nnnINTERPRETATIONnOur results show that the development of a genomic signature that predicts usual interstitial pneumonia is feasible. These findings are an important first step towards the development of a molecular test that could be applied to bronchoscopy samples, thus avoiding surgery in the diagnosis of idiopathic pulmonary fibrosis.nnnFUNDINGnVeracyte.


BMC Genetics | 2016

Genome-wide imputation study identifies novel HLA locus for pulmonary fibrosis and potential role for auto-immunity in fibrotic idiopathic interstitial pneumonia.

Tasha E. Fingerlin; Weiming Zhang; Ivana V. Yang; Hannah C. Ainsworth; Pamela Russell; Rachel Z. Blumhagen; Marvin I. Schwarz; 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; Toby M. Maher; Philip L. Molyneaux; Athol U. Wells

BackgroundFibrotic idiopathic interstitial pneumonias (fIIP) are a group of fatal lung diseases with largely unknown etiology and without definitive treatment other than lung transplant to prolong life. There is strong evidence for the importance of both rare and common genetic risk alleles in familial and sporadic disease. We have previously used genome-wide single nucleotide polymorphism data to identify 10 risk loci for fIIP. Here we extend that work to imputed genome-wide genotypes and conduct new RNA sequencing studies of lung tissue to identify and characterize new fIIP risk loci.ResultsWe performed genome-wide genotype imputation association analyses in 1616 non-Hispanic white (NHW) cases and 4683 NHW controls followed by validation and replication (878 cases, 2017 controls) genotyping and targeted gene expression in lung tissue. Following meta-analysis of the discovery and replication populations, we identified a novel fIIP locus in the HLA region of chromosome 6 (rs7887 Pmetau2009=u20093.7u2009×u200910−09). Imputation of classic HLA alleles identified two in high linkage disequilibrium that are associated with fIIP (DRB1*15:01 Pu2009=u20091.3u2009×u200910−7 and DQB1*06:02 Pu2009=u20096.1u2009×u200910−8). Targeted RNA-sequencing of the HLA locus identified 21 genes differentially expressed between fibrotic and control lung tissue (Qu2009<u20090.001), many of which are involved in immune and inflammatory response regulation. In addition, the putative risk alleles, DRB1*15:01 and DQB1*06:02, are associated with expression of the DQB1 gene among fIIP cases (Qu2009<u20091u2009×u200910−16).ConclusionsWe have identified a genome-wide significant association between the HLA region and fIIP. Two HLA alleles are associated with fIIP and affect expression of HLA genes in lung tissue, indicating that the potential genetic risk due to HLA alleles may involve gene regulation in addition to altered protein structure. These studies reveal the importance of the HLA region for risk of fIIP and a basis for the potential etiologic role of auto-immunity in fIIP.

Collaboration


Dive into the Mark P. Steele's collaboration.

Top Co-Authors

Avatar

Kevin K. Brown

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

Marvin I. Schwarz

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Tasha E. Fingerlin

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Elissa Murphy

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Ivana V. Yang

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Steve D. Groshong

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Anna L. Peljto

University of Colorado Denver

View shared research outputs
Researchain Logo
Decentralizing Knowledge