1
|
Yang L, Xu Z, Wang Z, Ding F, Wu Z, Shi X, Wang J, Ma Y, Jin J. Increased pro-SFTPB in HDL promotes the pro-inflammatory transition of HDL and represents a sign of poor prognosis in ARDS patients. J Transl Med 2025; 23:75. [PMID: 39819672 PMCID: PMC11740663 DOI: 10.1186/s12967-025-06100-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is causatively associated with excessive alveolar inflammation involving deregulated pro-inflammatory macrophage polarization. High-density lipoprotein (HDL) showed critical anti-inflammatory roles by modulating macrophage function, and its adverse transition to pro-inflammation has an important role in the pathogenesis of ARDS. However, the relationship between HDL protein constituents and functional remodeling is unknown in ARDS. METHODS Proteomic techniques were applied to examine the protein profile changes in HDL from septic-ARDS patients versus HDL from healthy controls across two distinct cohorts: a discovery cohort (8 patients and 8 healthy controls) and a validation cohort (22 patients and 10 healthy controls). The changed components significantly associated with prognosis were identified. Luminex assessed the levels of 38 plasma cytokines and chemokines. The in vitro constructed pro-SFTPB enriched HDL was applied to confirm the effect on M1 polarization of THP1-derived macrophage. RESULTS 18 proteins were validated from 102 changed HDL proteins identified in the discovery cohort, including HDL particle components, such as apolipoproteins, pro-inflammatory substances known as serum amyloid As (SAAs), and anti-oxidative proteins like paraoxonases (PONs). Among these proteins, only the increase of pro-SFTPB in HDL was significantly associated with poor prognosis of ARDS patients. Notably, HDL-pro-SFTPB level was correlated with plasma pro-inflammatory cytokines and chemokines levels. The correlation assay of pro-SFTPB with other HDL components showed that it was positively and negatively correlated with SAA2 and PON3, respectively. Furthermore, the in vitro studies confirmed that the pro-SFTPB enriched HDL significantly promoted M1 polarization of monocyte-derived macrophages. CONCLUSIONS The increase of HDL-pro-SFTPB promotes HDL pro-inflammatory transition during septic ARDS, leading to exacerbated progression of ARDS through enhancing M1 macrophage polarization. HDL-pro-SFTPB could be a useful prognostic biomarker for septic ARDS.
Collapse
Affiliation(s)
- Liu Yang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No.8 Xi Tou Tiao, Youanmen Wai, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
| | - Zhuo Xu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No.8 Xi Tou Tiao, Youanmen Wai, Beijing, China
| | - Zhenyan Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
| | - Fangping Ding
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No.8 Xi Tou Tiao, Youanmen Wai, Beijing, China
| | - Zhipeng Wu
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No.8 Xi Tou Tiao, Youanmen Wai, Beijing, China
| | - Xiaoqian Shi
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
| | - Jing Wang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No.8 Xi Tou Tiao, Youanmen Wai, Beijing, China.
| | - Jiawei Jin
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China.
- Medical Research Center, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, N0.5 Jingyuan Road, Beijing, China.
| |
Collapse
|
2
|
Doroodgar Jorshery S, Chandra J, Walia AS, Stumiolo A, Corey K, Zekavat SM, Zinzuwadia AN, Patel K, Short S, Mega JL, Plowman RS, Pagidipati N, Sullivan SS, Mahaffey KW, Shah SH, Hernandez AF, Christiani D, Aerts HJWL, Weiss J, Lu MT, Raghu VK. Leveraging Deep Learning of Chest Radiograph Images to Identify Individuals at High Risk for Chronic Obstructive Pulmonary Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.14.24317055. [PMID: 39606360 PMCID: PMC11601700 DOI: 10.1101/2024.11.14.24317055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background This study assessed whether deep learning applied to routine outpatient chest X-rays (CXRs) can identify individuals at high risk for incident chronic obstructive pulmonary disease (COPD). Methods Using cancer screening trial data, we previously developed a convolutional neural network (CXR-Lung-Risk) to predict lung-related mortality from a CXR image. In this study, we externally validated CXR-Lung-Risk to predict incident COPD from routine CXRs. We identified outpatients without lung cancer, COPD, or emphysema who had a CXR taken from 2013-2014 at a Mass General Brigham site in Boston, Massachusetts. The primary outcome was 6-year incident COPD. Discrimination was assessed using AUC compared to the TargetCOPD clinical risk score. All analyses were stratified by smoking status. A secondary analysis was conducted in the Project Baseline Health Study (PBHS) to test associations between CXR-Lung-Risk with pulmonary function and protein abundance. Findings The primary analysis consisted of 12,550 ever-smokers (mean age 62·4±6·8 years, 48.9% male, 12.4% rate of 6-year COPD) and 15,298 never-smokers (mean age 63·0±8·1 years, 42.8% male, 3.8% rate of 6-year COPD). CXR-Lung-Risk had additive predictive value beyond the TargetCOPD score for 6-year incident COPD in both ever-smokers (CXR-Lung-Risk + TargetCOPD AUC: 0·73 [95% CI: 0·72-0·74] vs. TargetCOPD alone AUC: 0·66 [0·65-0·68], p<0·01) and never-smokers (CXR-Lung-Risk + TargetCOPD AUC: 0·70 [0·67-0·72] vs. TargetCOPD AUC: 0·60 [0·57-0·62], p<0·01). In secondary analyses of 2,097 individuals in the PBHS, CXR-Lung-Risk was associated with worse pulmonary function and with abundance of SCGB3A2 (secretoglobin family 3A member 2) and LYZ (lysozyme), proteins involved in pulmonary physiology. Interpretation In external validation, a deep learning model applied to a routine CXR image identified individuals at high risk for incident COPD, beyond known risk factors. Funding The Project Baseline Health Study and this analysis were funded by Verily Life Sciences, San Francisco, California. ClinicalTrialsgov Identifier NCT03154346.
Collapse
|
3
|
Banfi C, Piarulli F, Ragazzi E, Ghilardi S, Greco A, Lapolla A, Sartore G. Immature Surfactant Protein Type B and Surfactant Protein Type D Correlate with Coronary Heart Disease in Patients with Type 2 Diabetes. Life (Basel) 2024; 14:886. [PMID: 39063639 PMCID: PMC11277833 DOI: 10.3390/life14070886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Different specific surfactant proteins (SPs) have been associated with various pathological conditions, not only of the respiratory system, but also more recently with cardiovascular diseases, such as heart failure. The aim of the present study was to evaluate the role of SP-A, SP-D, and the precursor protein of SP-B (proSP-B) in the pathogenesis of cardiovascular damage in patients affected by type 2 diabetes (T2D). METHODS The study considered 31 patients with T2D (DN group), 34 patients with both T2D and coronary heart disease (CHD) (DC group), and 30 patients without diabetes but with a diagnosis of CHD (NC group). SP-A, SP-D, and proSP-B concentrations were determined in plasma samples, and were statistically compared using parametric and multivariate methods. RESULTS Higher plasma concentrations of SP-D and proSP-B were found in patients affected by both T2D and CHD (DC group), and in patients with CHD without diabetes (NC group), in comparison to T2D patients (DN group). A significant correlation, both with linear regression (r = 0.3565, p = 0.001) and Principal Component Analysis (PCA), was found between the plasma levels of SP-D and proSP-B in the overall cohort of patients. No differences in SP-A were observed among the three groups of subjects. CONCLUSION The present study extends the knowledge on the role of plasma SPs' levels as possible indicators of the risk of CHD being linked to T2D disease progression.
Collapse
Affiliation(s)
- Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.G.); (A.G.)
| | - Francesco Piarulli
- Department of Medicine-DIMED, University of Padova, 35122 Padova, Italy; (F.P.); (A.L.); (G.S.)
| | - Eugenio Ragazzi
- Studium Patavinum, University of Padova, 35122 Padova, Italy;
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.G.); (A.G.)
| | - Arianna Greco
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (S.G.); (A.G.)
| | - Annunziata Lapolla
- Department of Medicine-DIMED, University of Padova, 35122 Padova, Italy; (F.P.); (A.L.); (G.S.)
| | - Giovanni Sartore
- Department of Medicine-DIMED, University of Padova, 35122 Padova, Italy; (F.P.); (A.L.); (G.S.)
| |
Collapse
|
4
|
Feng S, Fu D, Zhang Y, Zhang L, Ji Y, Li H, A L. Serum pro-surfactant protein B is correlated with clinical properties in osteosarcoma patients. Biochem Cell Biol 2023; 101:456-463. [PMID: 37192548 DOI: 10.1139/bcb-2022-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
It is critical to find efficient non-invasive prognostic factor for osteosarcoma. In this study, we demonstrated that serum protein of pro-surfactant protein B (pro-SFTPB) may be a potential diagnostic indicator in osteosarcoma. We found that serum pro-SFTPB was highly expressed in osteosarcoma patients and presented good diagnostic value to discern osteosarcoma patients from non-osteosarcoma control subjects. Serum pro-SFTPB was also significantly correlated with advanced clinical stage, distant metastasis, and shorter overall survival. In addition, serum pro-SFTPB was demonstrated to be an independent prognostic factor for osteosarcoma. Overall, our study demonstrated that serum pro-SFTPB may be a useful diagnostic factor for osteosarcoma.
Collapse
Affiliation(s)
- Shi Feng
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Di Fu
- Department of General Medicine, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Yong Zhang
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Le Zhang
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Yingnan Ji
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Hongqiu Li
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| | - Liang A
- Department of Second Orthopedics, Affiliated Central Hospital of Shenyang Medical College, Shenyang Tiexi District China
| |
Collapse
|
5
|
Luyapan J, Bossé Y, Li Z, Xiao X, Rosenberger A, Hung RJ, Lam S, Zienolddiny S, Liu G, Kiemeney LA, Chen C, McKay J, Johansson M, Johansson M, Tardon A, Fernandez-Tardon G, Brennan P, Field JK, Davies MP, Woll PJ, Cox A, Taylor F, Arnold SM, Lazarus P, Grankvist K, Landi MT, Christiani DC, MacKenzie TA, Amos CI. Candidate pathway analysis of surfactant proteins identifies CTSH and SFTA2 that influences lung cancer risk. Hum Mol Genet 2023; 32:2842-2855. [PMID: 37471639 PMCID: PMC10481107 DOI: 10.1093/hmg/ddad095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
Pulmonary surfactant is a lipoprotein synthesized and secreted by alveolar type II cells in lung. We evaluated the associations between 200,139 single nucleotide polymorphisms (SNPs) of 40 surfactant-related genes and lung cancer risk using genotyped data from two independent lung cancer genome-wide association studies. Discovery data included 18,082 cases and 13,780 controls of European ancestry. Replication data included 1,914 cases and 3,065 controls of European descent. Using multivariate logistic regression, we found novel SNPs in surfactant-related genes CTSH [rs34577742 C > T, odds ratio (OR) = 0.90, 95% confidence interval (CI) = 0.89-0.93, P = 7.64 × 10-9] and SFTA2 (rs3095153 G > A, OR = 1.16, 95% CI = 1.10-1.21, P = 1.27 × 10-9) associated with overall lung cancer in the discovery data and validated in an independent replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.80-0.96, P = 5.76 × 10-3) and SFTA2 (rs3095153 G > A, OR = 1.14, 95% CI = 1.01-1.28, P = 3.25 × 10-2). Among ever smokers, we found SNPs in CTSH (rs34577742 C > T, OR = 0.89, 95% CI = 0.85-0.92, P = 1.94 × 10-7) and SFTA2 (rs3095152 G > A, OR = 1.20, 95% CI = 1.14-1.27, P = 4.25 × 10-11) associated with overall lung cancer in the discovery data and validated in the replication data-CTSH (rs34577742 C > T, OR = 0.88, 95% CI = 0.79-0.97, P = 1.64 × 10-2) and SFTA2 (rs3095152 G > A, OR = 1.15, 95% CI = 1.01-1.30, P = 3.81 × 10-2). Subsequent transcriptome-wide association study using expression weights from a lung expression quantitative trait loci study revealed genes most strongly associated with lung cancer are CTSH (PTWAS = 2.44 × 10-4) and SFTA2 (PTWAS = 2.32 × 10-6).
Collapse
Affiliation(s)
- Jennifer Luyapan
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
- Department of Molecular Medicine, Laval University, Quebec City, G1V 0A6, Canada
| | - Zhonglin Li
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, G1V 0A6, Canada
| | - Xiangjun Xiao
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Albert Rosenberger
- Institut für Genetische Epidemiologie, Georg-August-Universität Göttingen, Gottingen, Niedersachsen, Germany
| | - Rayjean J Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbuaum Research Institute, Sinai Health System, Toronto, ON, M5G 1X5, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, V5Z 4E6, Canada
| | - Shanbeh Zienolddiny
- Department of Toxicology, National Institute of Occupational Health, Oslo 0033, Norway
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, Princess Margaret Research Institute, Epidemiology Division,Toronto, ON, M5G 1L7, Canada
| | - Lambertus A Kiemeney
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, 6525 GA, the Netherlands
| | - Chu Chen
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James McKay
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mattias Johansson
- International Agency for Research on Cancer (IARC/WHO), Genomic Epidemiology Branch Lyon 69008, France
| | - Mikael Johansson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, 901 87, Sweden
| | - Adonina Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Guillermo Fernandez-Tardon
- Health Research Institute of the Principality of Asturias, University of Oviedo and CIBERSP, Oviedo, Asturias, 33071, Spain
| | - Paul Brennan
- Institute of Medical Informatics, Biometry and Epidemiology, Chair of Epidemiology, Ludwig Maximillians University, Munich, Bavaria, 80539, Germany
| | - John K Field
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Michael P Davies
- Molecular and Clinical Cancer Medicine, Roy Castle Lung Cancer Research Programme, The University of Liverpool Institute of Translational Medicine, Liverpool, L69 7ZX, UK
| | - Penella J Woll
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield, S10 2AH, UK
| | - Angela Cox
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Fiona Taylor
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, S10 2AH, UK
| | - Susanne M Arnold
- Division of Medical Oncology, Cancer Center, University of Kentucky, Lexington, KY 40508, USA
| | - Philip Lazarus
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA, 99163, USA
| | - Kjell Grankvist
- Department of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, 901 87, Sweden
| | - Maria T Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - David C Christiani
- Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Todd A MacKenzie
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
| | - Christopher I Amos
- Quantitative Biomedical Science Program, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA
- Department of Medicine, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
6
|
Garavaglia ML, Bodega F, Porta C, Milzani A, Sironi C, Dalle-Donne I. Molecular Impact of Conventional and Electronic Cigarettes on Pulmonary Surfactant. Int J Mol Sci 2023; 24:11702. [PMID: 37511463 PMCID: PMC10380520 DOI: 10.3390/ijms241411702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
The alveolar epithelium is covered by a non-cellular layer consisting of an aqueous hypophase topped by pulmonary surfactant, a lipo-protein mixture with surface-active properties. Exposure to cigarette smoke (CS) affects lung physiology and is linked to the development of several diseases. The macroscopic effects of CS are determined by several types of cell and molecular dysfunction, which, among other consequences, lead to surfactant alterations. The purpose of this review is to summarize the published studies aimed at uncovering the effects of CS on both the lipid and protein constituents of surfactant, discussing the molecular mechanisms involved in surfactant homeostasis that are altered by CS. Although surfactant homeostasis has been the topic of several studies and some molecular pathways can be deduced from an analysis of the literature, it remains evident that many aspects of the mechanisms of action of CS on surfactant homeostasis deserve further investigation.
Collapse
Affiliation(s)
| | - Francesca Bodega
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Cristina Porta
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Aldo Milzani
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| | - Chiara Sironi
- Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Università degli Studi di Milano, 20133 Milan, Italy
| | - Isabella Dalle-Donne
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
7
|
Surfactant Protein B Plasma Levels: Reliability as a Biomarker in COPD Patients. Biomedicines 2023; 11:biomedicines11010124. [PMID: 36672632 PMCID: PMC9855771 DOI: 10.3390/biomedicines11010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/24/2022] [Accepted: 12/27/2022] [Indexed: 01/06/2023] Open
Abstract
Background: The diagnosis of COPD is based on both clinical signs and functional tests. Although there are different functional tests used to assess COPD, no reliable biomarkers able to provide information on pathogenesis and severity are available. The aim of the present study is to explore the relationship between surfactant protein B (Sp-B) serum levels and clinical, radiological, and functional pulmonary parameters in COPD patients. Methods: Forty COPD patients and twenty smokers without airflow limitations or respiratory symptoms were enrolled. Each patient was given questionnaires (CAT and mMRC) and 6MWT, spirometry, DLCO, and computer tomography (CT) were performed. All participants underwent a venous blood sample drawing, and quantitative detection of their Sp-B plasma levels was performed by an enzyme-linked immunosorbent assay. The spirometry and Sp-B plasma levels were assessed after 12 months. Results: A statistically significant difference was found in the plasma Sp-B levels between COPD patients compared to the other group (4.72 + 3.2 ng/mL vs. 1.78 + 1.5 ng/mL; p < 0.001). The change in FEV1 after 12 months (Delta FEV1) showed a significantly negative correlation with respect to the change in Sp-B levels (Delta SpB) (r = −0.4; p < 0.05). This correlation indicates that increasing the plasma dosage of SpB is a foretoken of functional decline. Conclusions: SpB may be considered as a useful marker in COPD assessment and provides prognostic information on lung functional decline. Despite its usefulness, further studies are needed to define its reliability as a biomarker.
Collapse
|
8
|
Gautam LK, Harriott NC, Caceres AM, Ryan AL. Basic Science Perspective on Engineering and Modeling the Large Airways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:73-106. [PMID: 37195527 DOI: 10.1007/978-3-031-26625-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The airway epithelium provides a physical and biochemical barrier playing a key role in protecting the lung from infiltration of pathogens and irritants and is, therefore, crucial in maintaining tissue homeostasis and regulating innate immunity. Due to continual inspiration and expiration of air during breathing, the epithelium is exposed to a plethora of environmental insults. When severe or persistent, these insults lead to inflammation and infection. The effectiveness of the epithelium as a barrier is reliant upon its capacity for mucociliary clearance, immune surveillance, and regeneration upon injury. These functions are accomplished by the cells that comprise the airway epithelium and the niche in which they reside. Engineering of new physiological and pathological models of the proximal airways requires the generation of complex structures comprising the surface airway epithelium, submucosal gland epithelium, extracellular matrix, and niche cells, including smooth muscle cells, fibroblasts, and immune cells. This chapter focuses on the structure-function relationships in the airways and the challenges of developing complex engineered models of the human airway.
Collapse
Affiliation(s)
- Lalit K Gautam
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Noa C Harriott
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Adrian M Caceres
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
9
|
Uysal P. Novel Applications of Biomarkers in Chronic Obstructive Pulmonary Disease. Biomark Med 2022. [DOI: 10.2174/9789815040463122010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an important health
problem and an increasing cause of morbidity and mortality worldwide. Currently,
COPD is considered a multisystem disease. Although it primarily affects the lungs,
structural and functional changes occur in other organs due to systemic inflammation.
It is stated that in patients with COPD, airway and systemic inflammatory markers are
increased and that these markers are high are associated with a faster decline in lung
functions. In recent years, numerous articles have been published on the discovery and
evaluation of biomarkers in COPD. Many markers have also been studied to accurately
assess COPD exacerbations and provide effective treatment. However, based on the
evidence from published studies, a single molecule has not been adequately validated
for broad clinical use.
Collapse
Affiliation(s)
- Pelin Uysal
- Department of Chest Diseases, Faculty of Medicine, Mehmet Ali Aydınlar University, Atakent
Hospital, Istanbul, Turkey
| |
Collapse
|
10
|
Hernández Cordero AI, Yang CX, Yang J, Li X, Horvath S, Shaipanich T, MacIsaac J, Lin D, McEwen L, Kobor MS, Guillemi S, Harris M, Lam W, Lam S, Obeidat M, Novak RM, Hudson F, Klinker H, Dharan N, Montaner J, Man SP, Kunisaki K, Sin DD, Leung JM. The relationship between the epigenetic aging biomarker "grimage" and lung function in both the airway and blood of people living with HIV: An observational cohort study. EBioMedicine 2022; 83:104206. [PMID: 35944348 PMCID: PMC9379521 DOI: 10.1016/j.ebiom.2022.104206] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/14/2022] [Accepted: 07/24/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Age-related comorbidities such as chronic obstructive pulmonary disease (COPD) are common in people living with human immunodeficiency virus (PLWH). We investigated the relationship between COPD and the epigenetic age of the airway epithelium and peripheral blood of PLWH. METHODS Airway epithelial brushings from 34 PLWH enrolled in the St. Paul's Hospital HIV Bronchoscopy cohort and peripheral blood from 378 PLWH enrolled in The Strategic Timing of Antiretroviral Treatment (START) study were profiled for DNA methylation. The DNA methylation biomarker of age and healthspan, GrimAge, was calculated in both tissue compartments. We tested the association of GrimAge with COPD in the airway epithelium and airflow obstruction as defined by an FEV1/FVC<0.70, and FEV1 decline over 6 years in blood. FINDINGS The airway epithelium of PLWH with COPD was associated with greater GrimAge residuals compared to PLWH without COPD (Beta=3.18, 95%CI=1.06-5.31, P=0.005). In blood, FEV1/FVC<LLN was associated with greater GrimAge residuals (Beta=1.74, 95%CI=0.37-3.24, P=0.019). FEV1 decline was inversely correlated with GrimAge residuals in blood (r=-0.13, P=0.012). PLWH who had normal lung function but who subsequently developed an FEV1/FVC<0.70 over the course of 6 years had higher GrimAge residuals at baseline (Beta=2.33, 95%CI=0.23-4.44, P=0.031). INTERPRETATION GrimAge may reflect lung and systemic epigenetic changes that occur with advanced airflow obstruction and may help to identify PLWH with a higher risk of developing COPD. FUNDING Canadian Institutes of Health Research and the British Columbia Lung Association. The START substudy was funded by NIH grants: UM1-AI068641, UM1-AI120197, and RO1HL096453.
Collapse
Affiliation(s)
- Ana I Hernández Cordero
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Chen Xi Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia Yang
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Xuan Li
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Steve Horvath
- Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles (UCLA), Los Angeles, CA 90095, United States,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA 90095, United States
| | - Tawimas Shaipanich
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Julia MacIsaac
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - David Lin
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa McEwen
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael S. Kobor
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Silvia Guillemi
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, British Columbia, Canada,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, British Columbia, Canada,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wan Lam
- British Columbia Cancer Research Centre and the University of British Columbia, Vancouver, British Columbia
| | - Stephen Lam
- British Columbia Cancer Research Centre and the University of British Columbia, Vancouver, British Columbia
| | - Ma'en Obeidat
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard M. Novak
- Section of Infectious Diseases, University of Illinois at Chicago, Chicago, IL, United States
| | - Fleur Hudson
- MRC Clinical Trials Unit, University College London, London, UK
| | - Hartwig Klinker
- Medizinische Klinik und Poliklinik II, Universitatsklinikum Wurzburg, Wurzburg, Germany
| | - Nila Dharan
- The Kirby Institute, UNSW Sydney, Sydney, New South Wales, Australia
| | - Julio Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Providence Health Care, Vancouver, British Columbia, Canada,Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - S.F. Paul Man
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ken Kunisaki
- Minneapolis Veterans Affairs Health Care System, Section of Pulmonary, Critical Care and Sleep Medicine and the Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Don D. Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Janice M. Leung
- Centre for Heart Lung Innovation, St. Paul's Hospital and University of British Columbia, Vancouver, British Columbia, Canada,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada,Corresponding author at: Room 166-1081 Burrard St, Centre for Heart Lung Innovation, Vancouver, BC V6Z 1Y6.
| | | |
Collapse
|
11
|
Axelsson GT, Gudmundsson G, Pratte KA, Aspelund T, Putman RK, Sanders JL, Gudmundsson EF, Hatabu H, Gudmundsdottir V, Gudjonsson A, Hino T, Hida T, Hobbs BD, Cho MH, Silverman EK, Bowler RP, Launer LJ, Jennings LL, Hunninghake GM, Emilsson V, Gudnason V. The Proteomic Profile of Interstitial Lung Abnormalities. Am J Respir Crit Care Med 2022; 206:337-346. [PMID: 35438610 PMCID: PMC9890263 DOI: 10.1164/rccm.202110-2296oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Rationale: Knowledge on biomarkers of interstitial lung disease is incomplete. Interstitial lung abnormalities (ILAs) are radiologic changes that may present in its early stages. Objectives: To uncover blood proteins associated with ILAs using large-scale proteomics methods. Methods: Data from two prospective cohort studies, the AGES-Reykjavik (Age, Gene/Environment Susceptibility-Reykjavik) study (N = 5,259) for biomarker discovery and the COPDGene (Genetic Epidemiology of COPD) study (N = 4,899) for replication, were used. Blood proteins were measured using DNA aptamers, targeting more than 4,700 protein analytes. The association of proteins with ILAs and ILA progression was assessed with regression modeling, as were associations with genetic risk factors. Adaptive Least Absolute Shrinkage and Selection Operator models were applied to bootstrap data samples to discover sets of proteins predictive of ILAs and their progression. Measurements and Main Results: Of 287 associations, SFTPB (surfactant protein B) (odds ratio [OR], 3.71 [95% confidence interval (CI), 3.20-4.30]; P = 4.28 × 10-67), SCGB3A1 (Secretoglobin family 3A member 1) (OR, 2.43 [95% CI, 2.13-2.77]; P = 8.01 × 10-40), and WFDC2 (WAP four-disulfide core domain protein 2) (OR, 2.42 [95% CI, 2.11-2.78]; P = 4.01 × 10-36) were most significantly associated with ILA in AGES-Reykjavik and were replicated in COPDGene. In AGES-Reykjavik, concentrations of SFTPB were associated with the rs35705950 MUC5B (mucin 5B) promoter polymorphism, and SFTPB and WFDC2 had the strongest associations with ILA progression. Multivariate models of ILAs in AGES-Reykjavik, ILAs in COPDGene, and ILA progression in AGES-Reykjavik had validated areas under the receiver operating characteristic curve of 0.880, 0.826, and 0.824, respectively. Conclusions: Novel, replicated associations of ILA, its progression, and genetic risk factors with numerous blood proteins are demonstrated as well as machine-learning-based models with favorable predictive potential. Several proteins are revealed as potential markers of early fibrotic lung disease.
Collapse
Affiliation(s)
- Gisli Thor Axelsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | - Gunnar Gudmundsson
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Department of Respiratory Medicine, Landspitali University Hospital, Reykjavik, Iceland
| | | | - Thor Aspelund
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | | | | | | | - Hiroto Hatabu
- Department of Radiology, and,Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Valborg Gudmundsdottir
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| | | | - Takuya Hino
- Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tomoyuki Hida
- Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts;,Department of Clinical Radiology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Brian D. Hobbs
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Michael H. Cho
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Edwin K. Silverman
- Pulmonary and Critical Care Division,,Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Russell P. Bowler
- National Jewish Health, Denver, Colorado;,School of Medicine, University of Colorado, Aurora, Colorado
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, Maryland; and
| | - Lori L. Jennings
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Gary M. Hunninghake
- Pulmonary and Critical Care Division,,Center for Pulmonary Functional Imaging, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland;,Icelandic Heart Association, Kopavogur, Iceland
| |
Collapse
|
12
|
Zafari Z, Lee TY, Sadatsafavi M. External validation of lung function predictions in real-world cohorts of COPD. Respir Med 2022; 198:106859. [PMID: 35524999 DOI: 10.1016/j.rmed.2022.106859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/11/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND An individualized FEV1 prediction model has recently been developed based on the Lung Health Study (LHS). An ongoing evaluation of the utility of prediction models in representative settings outside the setup of clinical trials is needed. OBJECTIVE To evaluate the external validity of the LHS FEV1 prediction model in a representative sample of Canadian ever-smoker COPD patients. METHODS We used the sample of ever-smokers with spirometrically-confirmed COPD from the Canadian Cohort of Obstructive Lung Disease (CanCOLD), an ongoing prospective, longitudinal, multicenter population-based cohort study. We compared the mean predicted against the mean observed FEV1 at follow-up visits. We calculated the root mean square error (RMSE) and the actual coverage probability of the 95% individualized prediction intervals. RESULTS The final CanCOLD sample consisted of 360 ever-smoker COPD patients, contributing to 970 FEV1 observations over an average of 3 years of follow-up (standard deviation (SD): 0.6 years). The mean observed vs. predicted FEV1 for the first follow-up visit (1.67 years from baseline) was 2.28L vs. 2.28L, and for the second visit (3.13 years from baseline) was 2.19L vs. 2.18L. The RMSE of the predictions was 0.205L, and the actual coverage probability of the 95% prediction intervals was 93%. The model had a similar performance across multiple subgroups. CONCLUSIONS The model showed robust performance in predicting FEV1 in the entire sample and in several subgroups of CanCOLD. Our results demonstrate that LHS predictions are valid for at least three years in the general COPD population.
Collapse
Affiliation(s)
- Zafar Zafari
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, USA.
| | - Tsung-Ying Lee
- Department of Pharmaceutical Health Services Research, University of Maryland School of Pharmacy, USA
| | - Mohsen Sadatsafavi
- Respiratory Evaluation Sciences Program, Faculty of Pharmaceutical Sciences, The University of British Columbia, Canada
| |
Collapse
|
13
|
Shao B, Snell-Bergeon JK, Pyle LL, Thomas KE, de Boer IH, Kothari V, Segrest J, Davidson WS, Bornfeldt KE, Heinecke JW. Pulmonary surfactant protein B carried by HDL predicts incident CVD in patients with type 1 diabetes. J Lipid Res 2022; 63:100196. [PMID: 35300983 PMCID: PMC9010748 DOI: 10.1016/j.jlr.2022.100196] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 12/22/2022] Open
Abstract
Atherosclerotic CVD is the major cause of death in patients with type 1 diabetes mellitus (T1DM). Alterations in the HDL proteome have been shown to associate with prevalent CVD in T1DM. We therefore sought to determine which proteins carried by HDL might predict incident CVD in patients with T1DM. Using targeted MS/MS, we quantified 50 proteins in HDL from 181 T1DM subjects enrolled in the prospective Coronary Artery Calcification in Type 1 Diabetes study. We used Cox proportional regression analysis and a case-cohort design to test associations of HDL proteins with incident CVD (myocardial infarction, coronary artery bypass grafting, angioplasty, or death from coronary heart disease). We found that only one HDL protein-SFTPB (pulmonary surfactant protein B)-predicted incident CVD in all the models tested. In a fully adjusted model that controlled for lipids and other risk factors, the hazard ratio was 2.17 per SD increase of SFTPB (95% confidence interval, 1.12-4.21, P = 0.022). In addition, plasma fractionation demonstrated that SFTPB is nearly entirely bound to HDL. Although previous studies have shown that high plasma levels of SFTPB associate with prevalent atherosclerosis only in smokers, we found that SFTPB predicted incident CVD in T1DM independently of smoking status and a wide range of confounding factors, including HDL-C, LDL-C, and triglyceride levels. Because SFTPB is almost entirely bound to plasma HDL, our observations support the proposal that SFTPB carried by HDL is a marker-and perhaps mediator-of CVD risk in patients with T1DM.
Collapse
Affiliation(s)
- Baohai Shao
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | | | - Laura L Pyle
- Department of Pediatrics, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Katie E Thomas
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Ian H de Boer
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Vishal Kothari
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jere Segrest
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - William S Davidson
- Center for Lipid and Arteriosclerosis Science, Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| | | | - Jay W Heinecke
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
14
|
Zhang YH, Hoopmann MR, Castaldi PJ, Simonsen KA, Midha MK, Cho MH, Criner GJ, Bueno R, Liu J, Moritz RL, Silverman EK. Lung proteomic biomarkers associated with chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1119-L1130. [PMID: 34668408 PMCID: PMC8715017 DOI: 10.1152/ajplung.00198.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/27/2021] [Accepted: 10/15/2021] [Indexed: 11/22/2022] Open
Abstract
Identifying protein biomarkers for chronic obstructive pulmonary disease (COPD) has been challenging. Most previous studies have used individual proteins or preselected protein panels measured in blood samples. Mass spectrometry proteomic studies of lung tissue have been based on small sample sizes. We used mass spectrometry proteomic approaches to discover protein biomarkers from 150 lung tissue samples representing COPD cases and controls. Top COPD-associated proteins were identified based on multiple linear regression analysis with false discovery rate (FDR) < 0.05. Correlations between pairs of COPD-associated proteins were examined. Machine learning models were also evaluated to identify potential combinations of protein biomarkers related to COPD. We identified 4,407 proteins passing quality controls. Twenty-five proteins were significantly associated with COPD at FDR < 0.05, including interleukin 33, ferritin (light chain and heavy chain), and two proteins related to caveolae (CAV1 and CAVIN1). Multiple previously reported plasma protein biomarkers for COPD were not significantly associated with proteomic analysis of COPD in lung tissue, although RAGE was borderline significant. Eleven pairs of top significant proteins were highly correlated (r > 0.8), including several strongly correlated with RAGE (EHD2 and CAVIN1). Machine learning models using Random Forests with the top 5% of protein biomarkers demonstrated reasonable accuracy (0.707) and area under the curve (0.714) for COPD prediction. Mass spectrometry-based proteomic analysis of lung tissue is a promising approach for the identification of biomarkers for COPD.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Peter J Castaldi
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | | | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gerard J Criner
- Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jiangyuan Liu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Wang Y, Gao X, Li Y, Wang X, Li Y, Zhang S, Liu H, Guo H, Lu W, Sun D. Pulmonary surfactant-associated protein B regulates prostaglandin-endoperoxide synthase-2 and inflammation in chronic obstructive pulmonary disease. Exp Physiol 2021; 106:1303-1311. [PMID: 33729612 DOI: 10.1113/ep089244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
NEW FINDINGS What is the central question of this study? It is reported that polymorphism of the gene for pulmonary surfactant-associated protein B (SFTPB) is associated with chronic obstructive pulmonary disease (COPD): what are the function and mechanism of action of SFTPB in COPD? What is the main finding and its importance? Under stimulation of the risk factors of COPD, SFTPB expression is decreased, which may be involved in the formation of COPD. The progress of COPD induces an inflammatory response and reduces SFTPB expression. Levels of prostaglandin-endoperoxide synthase-2 (PTGS2) and inflammatory responses are changed by SFTPB, which indicates that SFTPB promotes the progression of COPD by PTGS2 and inflammation. ABSTRACT Pulmonary surfactant-associated protein B (SFTPB) is a critical protein for lung homeostasis, and polymorphism of its gene is associated with chronic obstructive pulmonary disease (COPD). However, few studies have so far confirmed the functional involvement of SFTPB in COPD. Serum SFTPB and inflammatory cytokine levels were measured in 54 patients with acute exacerbation of COPD and 29 healthy controls. A549 cells were induced using 10% cigarette smoke extract (CSE) and treated with dexamethasone to investigate the effect of inflammation on SFTPB expression, and the effect of SFTPB overexpression and silencing on inflammatory cytokines was measured using real-time PCR and enzyme-linked immunosorbent assay. SFTPB expression was assessed in mouse lung tissues using immunofluorescence. Serum levels of SFTPB were significantly lower in COPD patients than in controls (P = 0.009). Conversely, levels of interleukin (IL)-6 and prostaglandin-endoperoxide synthase-2 (PTGS2) were increased in COPD patients (IL-6: P = 0.006; PTGS2: P = 0.043). After CSE treatment, SFTPB mRNA and protein levels were significantly decreased compared to controls (mRNA: P = 0.002; protein: P = 0.011), while IL-6, IL-8 and PTGS2 were elevated. Dexamethasone treatment increased SFTPB levels. Following overexpression of SFTPB in A549 cells, mRNA and protein levels of IL-6, IL-8 and PTGS2 were significantly reduced, while gene silencing induced the opposite effect. SFTPB levels were significantly reduced in the lung tissue of a mouse model of COPD compared to controls. Reduced SFTPB levels may induce PTGS2 and inflammatory responses in COPD and SFTPB could be a key protein for evaluation of COPD progression.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Xiaoyu Gao
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Yuan Li
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Xiao Wang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Yuanyuan Li
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sainan Zhang
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Hongyan Liu
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Hui Guo
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dejun Sun
- Key Laboratory of National Health Commission for the Diagnosis & Treatment of COPD, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China.,Graduate School, Baotou Medical College, Baotou, Inner Mongolia, China
| |
Collapse
|
16
|
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, Jaitovich A. Large-Scale Multi-omic Analysis of COVID-19 Severity. Cell Syst 2021; 12:23-40.e7. [PMID: 33096026 PMCID: PMC7543711 DOI: 10.1016/j.cels.2020.10.003] [Citation(s) in RCA: 397] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/24/2020] [Accepted: 10/05/2020] [Indexed: 01/08/2023]
Abstract
We performed RNA-seq and high-resolution mass spectrometry on 128 blood samples from COVID-19-positive and COVID-19-negative patients with diverse disease severities and outcomes. Quantified transcripts, proteins, metabolites, and lipids were associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many of which were involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a machine learning approach for prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Katherine A Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Morgridge Institute for Research, Madison, WI 53562, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ian J Miller
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | | | - Trenton M Peters-Clarke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Jesse G Meyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Qiuwen Quan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Laura K Muehlbauer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Edna A Trujillo
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yuchen He
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Amit Chopra
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Hau C Chieng
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Anupama Tiwari
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Division of Sleep Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Marc A Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA
| | - Brett Paulson
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Dain R Brademan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yunyun Zhu
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lia R Serrano
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Vanessa Linke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lisa A Drake
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Alejandro P Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA; Department of Ophthalmology, Albany Medical College, Albany, NY 12208, USA
| | | | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Deane F Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA; Morgridge Institute for Research, Madison, WI 53562, USA; Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA; Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA.
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
17
|
Contoli M, Morandi L, Di Marco F, Carone M. A perspective for chronic obstructive pulmonary disease (COPD) management: six key clinical questions to improve disease treatment. Expert Opin Pharmacother 2020; 22:427-437. [PMID: 33021128 DOI: 10.1080/14656566.2020.1828352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION In 2011, the GOLD recommendations for the treatment of Chronic Obstructive Pulmonary Disease (COPD) introduced new clinical elements to classify the severity of the disease and to guide pharmacological choice. For the first time in the GOLD documents, treatment decision was no longer guided only by pulmonary function, but by a more complex combination of pulmonary function and clinical aspects. The recent versions of the GOLD recommendations introduce new aspects for the clinicians and pose new question for the management of the disease. In addition, inflammatory biomarkers and blood eosinophil levels, have been considered to guide treatment selection. AREA COVERED The evolution of disease management proposed by the GOLD document opens several areas of debate. A series of roundtable discussions among respiratory physicians took place in Italy to address key clinical questions. Particularly, the role of lung function and the use of biomarkers, the adherence to international guidelines and the possibility to personalize the pharmacological approach in COPD patients have been discussed, summarized and analyzed. EXPERT OPINION The authors believe that the development of a precision medicine approach tailoring the specific treatment for each patient is the goal of COPD management and may be achieved by considering the phenotypic classification of COPD patients.
Collapse
Affiliation(s)
- Marco Contoli
- Department of Morphology, Surgery and Experimental Medicine, Università Di Ferrara, Ferrara, Italy
| | - Luca Morandi
- Department of Morphology, Surgery and Experimental Medicine, Università Di Ferrara, Ferrara, Italy
| | - Fabiano Di Marco
- Department of Health Science, Università degli studi di Milano, Respiratory Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Mauro Carone
- Division of Pneumology, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
18
|
Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, He Y, Chopra A, Chieng HC, Tiwari A, Judson MA, Paulson B, Brademan DR, Zhu Y, Serrano LR, Linke V, Drake LA, Adam AP, Schwartz BS, Singer HA, Swanson S, Mosher DF, Stewart R, Coon JJ, Jaitovich A. Large-scale Multi-omic Analysis of COVID-19 Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.07.17.20156513. [PMID: 32743614 PMCID: PMC7388490 DOI: 10.1101/2020.07.17.20156513] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We performed RNA-Seq and high-resolution mass spectrometry on 128 blood samples from COVID-19 positive and negative patients with diverse disease severities. Over 17,000 transcripts, proteins, metabolites, and lipids were quantified and associated with clinical outcomes in a curated relational database, uniquely enabling systems analysis and cross-ome correlations to molecules and patient prognoses. We mapped 219 molecular features with high significance to COVID-19 status and severity, many involved in complement activation, dysregulated lipid transport, and neutrophil activation. We identified sets of covarying molecules, e.g., protein gelsolin and metabolite citrate or plasmalogens and apolipoproteins, offering pathophysiological insights and therapeutic suggestions. The observed dysregulation of platelet function, blood coagulation, acute phase response, and endotheliopathy further illuminated the unique COVID-19 phenotype. We present a web-based tool (covid-omics.app) enabling interactive exploration of our compendium and illustrate its utility through a comparative analysis with published data and a machine learning approach for prediction of COVID-19 severity.
Collapse
Affiliation(s)
- Katherine A. Overmyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Evgenia Shishkova
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ian J. Miller
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Joseph Balnis
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | | | - Trenton M. Peters-Clarke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Jesse G. Meyer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Qiuwen Quan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Laura K. Muehlbauer
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Edna A. Trujillo
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yuchen He
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Amit Chopra
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Hau C. Chieng
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Anupama Tiwari
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Division of Sleep Medicine, Albany Medical Center, Albany, NY, USA
| | - Marc A. Judson
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
| | - Brett Paulson
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Dain R. Brademan
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Yunyun Zhu
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lia R. Serrano
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Vanessa Linke
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Lisa A. Drake
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Alejandro P. Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
- Department of Ophthalmology, Albany Medical College, Albany, NY, USA
| | | | - Harold A. Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Scott Swanson
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Deane F. Mosher
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53562, USA
| | - Joshua J. Coon
- National Center for Quantitative Biology of Complex Systems, Madison, WI 53562, USA
- Morgridge Institute for Research, Madison, WI 53562, USA
- Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI 53562, USA
- Department of Chemistry, University of Wisconsin, Madison, WI 53562, USA
| | - Ariel Jaitovich
- Division of Pulmonary and Critical Care Medicine, Albany Medical Center, Albany, NY, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
19
|
Mourad BH. Prediction of lung cancer risk using circulating pro-surfactant protein B and high-sensitivity C-reactive protein among Egyptian workers in the rubber industry. Toxicol Ind Health 2020; 36:237-249. [PMID: 32419650 DOI: 10.1177/0748233720923504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE For several decades, there has been increasing evidence for excess incidence of lung cancer among workers in the rubber industry. The purpose of this study was to assess the risk of lung cancer occurrence among Egyptian workers involved in the rubber industry using two circulating protein biomarkers. METHODS This study was performed in a rubber manufacturing factory in Shubra El-Kheima region in Greater Cairo, Egypt. Environmental assessment for the suspended particulate matter of size 10 µm (PM10) concentrations was done. Levels of plasma pro-surfactant protein B (pro-SFTPB) and serum high-sensitivity C-reactive protein (HsCRP) were measured among the studied population (n = 155) who were divided into two groups. The first group included 75 workers exposed to rubber manufacturing process while the control group involved 80 administrative subjects. RESULTS The levels of PM10 neither exceeded the Egyptian nor the international permissible limits where the highest levels were observed in the mixing department. However, through medical history and clinical examination, it was observed that some general and respiratory manifestations were more prevalent among the exposed group when compared with their controls. Laboratory investigations revealed that the mean values of pro-SFTPB and HsCRP levels among exposed workers were significantly higher than those of the control group. These increased circulating proteins levels were strongly and positively correlated with each other and with the duration of employment of exposed workers. CONCLUSION The study results support the conclusion that prolonged occupational exposure to rubber manufacturing process is associated with an elevated risk of lung cancer.
Collapse
Affiliation(s)
- Basma Hussein Mourad
- Department of Occupational and Environmental Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
20
|
Sun Y, Zhou J. New insights into early intervention of chronic obstructive pulmonary disease with mild airflow limitation. Int J Chron Obstruct Pulmon Dis 2019; 14:1119-1125. [PMID: 31213792 PMCID: PMC6536809 DOI: 10.2147/copd.s205382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/18/2019] [Indexed: 02/01/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) has become one of the major public health problems worldwide due to its high morbidity and mortality. Up until now, COPD is still under-diagnosed and under-treated, especially for mild or moderate patients. It is widely accepted that the majority of patients with COPD are in the early stages, yet this subpopulation is underestimated. In recent years, growing evidence indicates that substantial physiological and clinical abnormalities exist in patients with mild COPD compared with healthy controls. Furthermore, recent studies suggest that pharmacologic intervention in early COPD has the potential to alter clinical outcomes. The main objective of this review is to summarize recent research regarding the heterogeneous pathophysiology, clinical features, and treatment of mild and moderate COPD. We also discuss promising markers of disease progression, which may contribute to the development of precision medicine in early COPD.
Collapse
Affiliation(s)
- Yilan Sun
- Department of Respiratory Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| | - Jianying Zhou
- Department of Respiratory Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People's Republic of China
| |
Collapse
|
21
|
Wang K, Huang Q, Zhao G, Yang J, Yang K, Huang Y. Gene polymorphisms of SFTPB rs7316, rs9752 and PAOX rs1046175 affect the diagnostic value of plasma Pro-SFTPB and DAS in Chinese Han non-small-cell lung cancer patients. J Cell Biochem 2019; 120:14804-14812. [PMID: 31016788 DOI: 10.1002/jcb.28741] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/14/2019] [Accepted: 03/22/2019] [Indexed: 12/20/2022]
Abstract
Plasma pro-surfactant protein B (pro-SFTPB) and N1,N12-diacetylspermine (DAS) can be used as markers for the diagnosis of non-small-cell lung carcinoma (NSCLC). Whether the genetic diversity affects the application value of Pro-SFTPB and DAS as a diagnostic marker for NSCLC is still unknown. This study aims to explore the relationship between SFTPB rs7316, rs9752 and PAOX rs1046175 gene polymorphisms and the diagnostic value of plasma Pro-SFTPB and DAS in patients with Chinese Han lung cancer. SFTPB rs7316, rs9752 and PAOX rs1046175 genotypes were analyzed by direct sequencing in 425 patients with NSCLC and 425 controls, and the levels of Pro-SFTPB and DAS in plasma were determined by enzyme-linked immunosorbent assay (ELISA). The area under the curve (AUC) of the SFTPB rs7316 locus TT genotype for the diagnosis of NSCLC was 0.758, and the AUC of the TC/CC genotype for the diagnosis of NSCLC was 0.872. The AUC of the SFTPB rs9752 locus GG genotype for the diagnosis of NSCLC was 0.935, and the AUC of the GC/CC genotype for the diagnosis of NSCLC was 0.648. The AUC of the PAOX rs1046175 locus GG for the diagnosis of NSCLC was 0.669, and the AUC of the GC/CC genotype for the diagnosis of NSCLC was 0.749. In conclusion, SFTPB rs7316, rs9752, and PAOX rs1046175 gene polymorphisms affect the diagnostic value of plasma Pro-SFTPB and DAS in patients with Chinese Han NSCLC.
Collapse
Affiliation(s)
- Kun Wang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Qiubo Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Guangqiang Zhao
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Jiapeng Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Kaiyun Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| |
Collapse
|
22
|
Shiels MS, Kirk GD, Drummond MB, Dhillon D, Hanash SM, Taguchi A, Engels EA. HIV Infection and Circulating Levels of Prosurfactant Protein B and Surfactant Protein D. J Infect Dis 2019; 217:413-417. [PMID: 29272472 DOI: 10.1093/infdis/jix510] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/22/2017] [Indexed: 01/17/2023] Open
Abstract
Prosurfactant protein B (pro-SFTPB) and surfactant protein D (SFTPD) are markers of lung inflammation and damage. We estimated geometric mean pro-SFTPB and SFTPD levels in 500 human immunodeficiency virus (HIV)-infected and 300 HIV-uninfected injection drug users, adjusting for smoking and other covariates. Pro-SFTPB levels were significantly higher among people with HIV (PWH) (adjusted geometric mean, 21.4 vs 18.1 ng/mL; P = .03), and were higher with lower CD4 counts (P trend = .001), higher HIV RNA (P trend = .05), and without highly active antiretroviral therapy (P = .03). These associations were not observed for SFTPD. Serum levels of pro-SFTPB are elevated among PWH and are associated with immunosuppression and uncontrolled viremia.
Collapse
Affiliation(s)
- Meredith S Shiels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Gregory D Kirk
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - M Bradley Drummond
- Division of Pulmonary and Critical Care Medicine, University of North Carolina, Chapel Hill
| | - Dilsher Dhillon
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston
| | - Samir M Hanash
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston
| | - Ayumu Taguchi
- Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
23
|
Lung function trajectories and chronic obstructive pulmonary disease: current understanding and knowledge gaps. Curr Opin Pulm Med 2019; 24:124-129. [PMID: 29206658 DOI: 10.1097/mcp.0000000000000456] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OR REVIEW Population-based studies have shown a significant heterogeneity in patients with chronic obstructive pulmonary disease (COPD), regarding both the attainment of maximal lung function and the subsequent decline over time. This review will highlight recent advances in the understanding of lung function trajectory in COPD, focusing on factors that influence peak adult lung function, markers of accelerated lung function decline and pharmacologic interventions in early phases of the disease. RECENT FINDINGS Recent data have shown that individuals with lower lung function early in life will go on to develop lower forced expiratory volume in 1 s (FEV1) in adulthood. Smoking can amplify the effect of specific childhood exposures on maximal adult lung function. Clinical symptoms such as chronic mucous hypersecretion and the biomarker club cell secretory protein have been associated with lung function decline over time. New computed tomography imaging markers also show promise as a way to detect early small airway disease, but need to be examined more longitudinally. In addition to these advances, a slower decline in FEV1 has been demonstrated in two randomized clinical trials studying tiotropium and inhaled fluticasone. SUMMARY A better understanding of lung function development and eventual decline in those at risk for progression to COPD will aide in a precision medicine approach, in which markers for those at risk of low maximal lung function and accelerated decline are identified. Targeted therapy can then be used early to modify disease activity and outcomes.
Collapse
|
24
|
He Y, Jiang Z, Tong F, Li M, Yin X, Hu S, Wang L. Experimental study of peripheral-blood pro-surfactant protein B for screening non-small cell lung cancer. Acta Cir Bras 2017; 32:568-575. [PMID: 28793041 DOI: 10.1590/s0102-865020170070000008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/14/2017] [Indexed: 12/23/2022] Open
Abstract
Purpose: To evaluate the possibility of using peripheral-blood presurfactant protein B (Pro-SFTPB) for screening non-small cell lung cancer (NSCLC). Methods: A total of 873 healthy volunteers and 165 lung cancer patients hospitalized in the Fifth People's Hospital of Dalian were tested Pro-SFTPB once every half year from January 2014 to September 2015. The healthy volunteers were also conducted spiral computed tomography (CT) examination once every year. The data were then com-pared and statistically analyzed. Results: The positive expression rate of Pro-SFTPB in NSCLC was significantly higher than that in healthy volunteers, and significantly higher in lung adenocarcinoma than in squamous cell carcinoma; additionally, the expression rate was increased with the in-crease of smoking index, and the intergroup differences showed statistical signifi-cance (p≤0.05). The positive rate of newly diagnosed lung cancer was 29.55%, higher than healthy volunteers (22.34%), but there was no significant difference (p>0.05). Conclusion: Pro-SFTPB is over expressed in non-small cell lung cancer, especially in lung adeno-carcinoma, but it can't be used as a clinical screening tool for lung cancer.
Collapse
Affiliation(s)
- Yong He
- Master, Department of Thoracic Surgery, 5th People's Hospital of Dalian, China. Conception of the study, interpretation of data, statistical analysis
| | - Zhenjie Jiang
- Master, Department of Thoracic Surgery, People's Hospital of Dalian, China. Acquisition of data, statistical analysis
| | - Fengzhi Tong
- Master, Clinical Laboratory, People's Hospital of Dalian, China. Acquisition and interpretation of data
| | - Mingwu Li
- Bachelor, Department of Thoracic Surgery, 5th People's Hospital of Dalian, China. Acquisition and interpretation of data
| | - Xingru Yin
- Bachelor, Department of Thoracic Surgery, 5th People's Hospital of Dalian, China. Acquisition of data, statistical analysis
| | - Shixin Hu
- Bachelor, Department of Thoracic Surgery, 5th People's Hospital of Dalian, China. Acquisition of data, statistical analysis
| | - Linlin Wang
- Bachelor, Matron, Department of Thoracic Surgery, 5th People's Hospital of Dalian, China. Acquisition of data, statistical analysis
| |
Collapse
|
25
|
Zemans RL, Jacobson S, Keene J, Kechris K, Miller BE, Tal-Singer R, Bowler RP. Multiple biomarkers predict disease severity, progression and mortality in COPD. Respir Res 2017; 18:117. [PMID: 28610627 PMCID: PMC5470282 DOI: 10.1186/s12931-017-0597-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/25/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by multiple subtypes and variable disease progression. Blood biomarkers have been variably associated with subtype, severity, and disease progression. Just as combined clinical variables are more highly predictive of outcomes than individual clinical variables, we hypothesized that multiple biomarkers may be more informative than individual biomarkers to predict subtypes, disease severity, disease progression, and mortality. METHODS Fibrinogen, C-Reactive Protein (CRP), surfactant protein D (SP-D), soluble Receptor for Advanced Glycation Endproducts (sRAGE), and Club Cell Secretory Protein (CC16) were measured in the plasma of 1465 subjects from the COPDGene cohort and 2746 subjects from the ECLIPSE cohort. Regression analysis was performed to determine whether these biomarkers, individually or in combination, were predictive of subtypes, disease severity, disease progression, or mortality, after adjustment for clinical covariates. RESULTS In COPDGene, the best combinations of biomarkers were: CC16, sRAGE, fibrinogen, CRP, and SP-D for airflow limitation (p < 10-4), SP-D, CRP, sRAGE and fibrinogen for emphysema (p < 10-3), CC16, fibrinogen, and sRAGE for decline in FEV1 (p < 0.05) and progression of emphysema (p < 10-3), and all five biomarkers together for mortality (p < 0.05). All associations except mortality were validated in ECLIPSE. The combination of SP-D, CRP, and fibrinogen was the best model for mortality in ECLIPSE (p < 0.05), and this combination was also significant in COPDGene. CONCLUSION This comprehensive analysis of two large cohorts revealed that combinations of biomarkers improve predictive value compared with clinical variables and individual biomarkers for relevant cross-sectional and longitudinal COPD outcomes.
Collapse
Affiliation(s)
- Rachel L Zemans
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., Denver, CO, 80206, USA. .,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, University of Colorado Anschutz Medical Campus, Research Building 2, 9th Floor, 12700 E. 19th Ave., Aurora, CO, USA.
| | - Sean Jacobson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., Denver, CO, 80206, USA
| | - Jason Keene
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, University of Colorado Anschutz Medical Campus, Research Building 2, 9th Floor, 12700 E. 19th Ave., Aurora, CO, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, University of Colorado Denver, Colorado School of Public Health, Mail Stop B119, 13001 E. 17th Place, Aurora, CO, 80045, USA
| | - Bruce E Miller
- GlaxoSmithKline R&D, 709 Swedeland Road #1539, King Of Prussia, PA, USA
| | - Ruth Tal-Singer
- GlaxoSmithKline R&D, 709 Swedeland Road #1539, King Of Prussia, PA, USA
| | - Russell P Bowler
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, 1400 Jackson St., Denver, CO, 80206, USA.,Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Denver, University of Colorado Anschutz Medical Campus, Research Building 2, 9th Floor, 12700 E. 19th Ave., Aurora, CO, USA
| |
Collapse
|
26
|
Zafari Z, Sin DD, Postma DS, Löfdahl CG, Vonk J, Bryan S, Lam S, Tammemagi CM, Khakban R, Man SFP, Tashkin D, Wise RA, Connett JE, McManus B, Ng R, Hollander Z, Sadatsafavi M. Individualized prediction of lung-function decline in chronic obstructive pulmonary disease. CMAJ 2016; 188:1004-1011. [PMID: 27486205 PMCID: PMC5047815 DOI: 10.1503/cmaj.151483] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2016] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND The rate of lung-function decline in chronic obstructive pulmonary disease (COPD) varies substantially among individuals. We sought to develop and validate an individualized prediction model for forced expiratory volume at 1 second (FEV1) in current smokers with mild-to-moderate COPD. METHODS Using data from a large long-term clinical trial (the Lung Health Study), we derived mixed-effects regression models to predict future FEV1 values over 11 years according to clinical traits. We modelled heterogeneity by allowing regression coefficients to vary across individuals. Two independent cohorts with COPD were used for validating the equations. RESULTS We used data from 5594 patients (mean age 48.4 yr, 63% men, mean baseline FEV1 2.75 L) to create the individualized prediction equations. There was significant between-individual variability in the rate of FEV1 decline, with the interval for the annual rate of decline that contained 95% of individuals being -124 to -15 mL/yr for smokers and -83 to 15 mL/yr for sustained quitters. Clinical variables in the final model explained 88% of variation around follow-up FEV1. The C statistic for predicting severity grades was 0.90. Prediction equations performed robustly in the 2 external data sets. INTERPRETATION A substantial part of individual variation in FEV1 decline can be explained by easily measured clinical variables. The model developed in this work can be used for prediction of future lung health in patients with mild-to-moderate COPD. TRIAL REGISTRATION Lung Health Study - ClinicalTrials.gov, no. NCT00000568; Pan-Canadian Early Detection of Lung Cancer Study - ClinicalTrials.gov, no. NCT00751660.
Collapse
Affiliation(s)
- Zafar Zafari
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Don D Sin
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont.
| | - Dirkje S Postma
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Claes-Göran Löfdahl
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Judith Vonk
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Stirling Bryan
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Stephen Lam
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - C Martin Tammemagi
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Rahman Khakban
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - S F Paul Man
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Donald Tashkin
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Robert A Wise
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - John E Connett
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Bruce McManus
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Raymond Ng
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Zsuszanna Hollander
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| | - Mohsen Sadatsafavi
- Centre for Clinical Epidemiology and Evaluation (Zafari, Bryan, Sadatsafavi), Vancouver Coastal Health Research Institute, University of British Columbia; Centre for Heart and Lung Innovation (Sin, Man, McManus, Hollander), St. Paul's Hospital; Institute for Heart and Lung Health (Sin, Man, McManus, Hollander, Sadatsafavi), University of British Columbia; Division of Respiratory Medicine (Sin, Lam, Man, Sadatsafavi), Department of Medicine, University of British Columbia, Vancouver, BC; University Medical Center Groningen (Postma, Vonk), Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen; University Medical Center Groningen (Postma), Department of Pulmonary Diseases, University of Groningen, Groningen, the Netherlands; Department of Respiratory Medicine and Allergology (Löfdahl), Lund University, Lund, Sweden; Department of Epidemiology (Vonk), University of Groningen, University Medical Center Groningen, the Netherlands; School of Population and Public Health (Bryan), University of British Columbia, Vancouver, BC; Collaboration for Outcomes Research and Evaluation, Faculty of Pharmaceutical Sciences (Khakban), University of British Columbia, Vancouver, BC; David Geffen School of Medicine at UCLA (Tashkin), Los Angeles, Calif.; Johns Hopkins University School of Medicine (Wise), Baltimore, Md.; University of Minnesota School of Public Health (Connett), Minneapolis, Minn.; PROOF Centre for Excellence (McManus, Ng, Hollander), Vancouver, BC; Department of Community Health Sciences (Tammemagi), Brock University, St. Catharines, Ont
| |
Collapse
|
27
|
Sin DD, Hollander Z, DeMarco ML, McManus BM, Ng RT. Biomarker Development for Chronic Obstructive Pulmonary Disease. From Discovery to Clinical Implementation. Am J Respir Crit Care Med 2016; 192:1162-70. [PMID: 26176936 DOI: 10.1164/rccm.201505-0871pp] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the major causes of morbidity and mortality in the world. Regrettably, there are no biomarkers to objectively diagnose COPD exacerbations, which are the major drivers of hospitalization and deaths from COPD. Moreover, there are no biomarkers to guide therapeutic choices or to risk stratify patients for imminent exacerbations and no objective biomarkers of disease activity or disease progression. Although there has been a tremendous investment in COPD biomarker discovery over the past 2 decades, clinical translation and implementation have not matched these efforts. In this article, we outline the challenges of biomarker development in COPD and provide an overview of a developmental pipeline that may be able to surmount these challenges and bring novel biomarker solutions to accelerate therapeutic discoveries and to improve the care and outcomes of the millions of individuals worldwide with COPD.
Collapse
Affiliation(s)
- Don D Sin
- 1 Centre for Heart Lung Innovation, James Hogg Research Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada.,2 Institute for Heart and Lung Health.,3 Division of Respiratory Medicine, Department of Medicine
| | - Zsuzsanna Hollander
- 1 Centre for Heart Lung Innovation, James Hogg Research Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada.,2 Institute for Heart and Lung Health.,4 PROOF Centre of Excellence, Vancouver, British Columbia, Canada
| | | | - Bruce M McManus
- 1 Centre for Heart Lung Innovation, James Hogg Research Centre, St. Paul's Hospital, Vancouver, British Columbia, Canada.,2 Institute for Heart and Lung Health.,5 Department of Pathology and Laboratory Medicine, and.,4 PROOF Centre of Excellence, Vancouver, British Columbia, Canada
| | - Raymond T Ng
- 6 Department of Computer Sciences, University of British Columbia, Vancouver, British Columbia, Canada; and.,4 PROOF Centre of Excellence, Vancouver, British Columbia, Canada
| |
Collapse
|
28
|
Wikoff WR, Hanash S, DeFelice B, Miyamoto S, Barnett M, Zhao Y, Goodman G, Feng Z, Gandara D, Fiehn O, Taguchi A. Diacetylspermine Is a Novel Prediagnostic Serum Biomarker for Non-Small-Cell Lung Cancer and Has Additive Performance With Pro-Surfactant Protein B. J Clin Oncol 2015; 33:3880-6. [PMID: 26282655 DOI: 10.1200/jco.2015.61.7779] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE We have investigated the potential of metabolomics to discover blood-based biomarkers relevant to lung cancer screening and early detection. An untargeted metabolomics approach was applied to identify biomarker candidates using prediagnostic sera from the Beta-Carotene and Retinol Efficacy Trial (CARET) study. PATIENTS AND METHODS A liquid chromatography/mass spectrometry hydrophilic interaction method designed to profile a wide range of metabolites was applied to prediagnostic serum samples from CARET participants (current or former heavy smokers), consisting of 100 patients who subsequently developed non-small-cell lung cancer (NSCLC) and 199 matched controls. A separate aliquot was used to quantify levels of pro-surfactant protein B (pro-SFTPB), a previously established protein biomarker for NSCLC. On the basis of the results from the discovery set, blinded validation of a metabolite, identified as N(1),N(12)-diacetylspermine (DAS), and pro-SFTPB was performed using an independent set of CARET prediagnostic sera from 108 patients with NSCLC and 216 matched controls. RESULTS Serum DAS was elevated by 1.9-fold, demonstrating significant specificity and sensitivity in the discovery set for samples collected up to 6 months before diagnosis of NSCLC. In addition, DAS significantly complemented performance of pro-SFTPB in both the discovery and validations sets, with a combined area under the curve in the validation set of 0.808 (P < .001 v pro-SFTPB). CONCLUSION DAS is a novel serum metabolite with significant performance in prediagnostic NSCLC and has additive performance with pro-SFTPB.
Collapse
Affiliation(s)
- William R Wikoff
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Samir Hanash
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Brian DeFelice
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Suzanne Miyamoto
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Matt Barnett
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Yang Zhao
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Gary Goodman
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Ziding Feng
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - David Gandara
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| | - Oliver Fiehn
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA.
| | - Ayumu Taguchi
- William R. Wikoff, Brian DeFelice, and Oliver Fiehn, National Institutes of Health West Coast Metabolomics Center, University of California, Davis, Davis; Suzanne Miyamoto and David Gandara, University of California, Davis, Davis Comprehensive Cancer Center, Sacramento, CA; Samir Hanash, Yang Zhao, Ziding Feng, and Ayumu Taguchi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Matt Barnett and Gary Goodman, Fred Hutchison Cancer Research Center, Seattle, WA
| |
Collapse
|
29
|
|