Serum amino acid concentrations and clinical outcomes in smokers: SPIROMICS metabolomics study

Wassim W Labaki, Tian Gu, Susan Murray, Jeffrey L Curtis, Larisa Yeomans, Russell P Bowler, R Graham Barr, Alejandro P Comellas, Nadia N Hansel, Christopher B Cooper, Igor Barjaktarevic, Richard E Kanner, Robert Paine 3rd, Merry-Lynn N McDonald, Jerry A Krishnan, Stephen P Peters, Prescott G Woodruff, Wanda K O'Neal, Wenqi Diao, Bei He, Fernando J Martinez, Theodore J Standiford, Kathleen A Stringer, MeiLan K Han, Wassim W Labaki, Tian Gu, Susan Murray, Jeffrey L Curtis, Larisa Yeomans, Russell P Bowler, R Graham Barr, Alejandro P Comellas, Nadia N Hansel, Christopher B Cooper, Igor Barjaktarevic, Richard E Kanner, Robert Paine 3rd, Merry-Lynn N McDonald, Jerry A Krishnan, Stephen P Peters, Prescott G Woodruff, Wanda K O'Neal, Wenqi Diao, Bei He, Fernando J Martinez, Theodore J Standiford, Kathleen A Stringer, MeiLan K Han

Abstract

Metabolomics is an emerging science that can inform pathogenic mechanisms behind clinical phenotypes in COPD. We aimed to understand disturbances in the serum metabolome associated with respiratory outcomes in ever-smokers from the SPIROMICS cohort. We measured 27 serum metabolites, mostly amino acids, by 1H-nuclear magnetic resonance spectroscopy in 157 white ever-smokers with and without COPD. We tested the association between log-transformed metabolite concentrations and one-year incidence of respiratory exacerbations after adjusting for age, sex, current smoking, body mass index, diabetes, inhaled or oral corticosteroid use, study site and clinical predictors of exacerbations, including FEV1% predicted and history of exacerbations. The mean age of participants was 53.7 years and 58% had COPD. Lower concentrations of serum amino acids were independently associated with 1-year incidence of respiratory exacerbations, including tryptophan (β = -4.1, 95% CI [-7.0; -1.1], p = 0.007) and the branched-chain amino acids (leucine: β = -6.0, 95% CI [-9.5; -2.4], p = 0.001; isoleucine: β = -5.2, 95% CI [-8.6; -1.8], p = 0.003; valine: β = -4.1, 95% CI [-6.9; -1.4], p = 0.003). Tryptophan concentration was inversely associated with the blood neutrophil-to-lymphocyte ratio (p = 0.03) and the BODE index (p = 0.03). Reduced serum amino acid concentrations in ever-smokers with and without COPD are associated with an increased incidence of respiratory exacerbations.

Conflict of interest statement

WWL, TG, SM, JLC, APC, LY, REK, RP, MLNM, SPP, WKO, WD, BH, TJS and KAS declare no competing interests. RPB serves on the advisory boards of GSK, BI and Mylan, and received research grants from GSK and BI. RGB reports grants from NIH during the conduct of the study; grants from Alpha1 Foundation, Foundation for the NIH, and COPD Foundation and personal fees from UpToDate outside the submitted work. N. N. H. serves on the advisory board of AstraZeneca, GlaxoSmithKline, and Mylan, and has received research grants fromAstraZeneca, Boehringer-Ingelheim Pharmaceuticals, Inc, Forest Research Institute, Inc, and GlaxoSmithKline. CBC reports grants from Equinox Health Clubs, personal fees from Equinox Health Clubs, grants from Amgen, personal fees from PulmonX, other from GlaxoSmithKline and part-time employment in scientific engagement for the GlaxoSmithKline Global Respiratory Franchise. IB reports personal fees from Astra Zeneca, grants from GE Healthcare, personal fees from Grifols, personal fees from CSL Behring, grants from Amgen, grants from PCORI. JAK reported serving on a data monitoring committee for Sanofi and receiving grants from the National Institutes of Health and the Patient-Centered Outcomes Research Institute. PGW reports personal fees from AstraZeneca, Theravance, Regeneron, Sanofi, Genentech, and Novartis. FJM reports personal fees and non-financial support from American College of Chest Physicians, AstraZeneca, Boehringer Ingelheim, Continuing Education, ConCert, Genentech, GlaxoSmithKline, Inova Fairfax Health System, Miller Communications, National Association for Continuing Education, Novartis, Pearl Pharmaceuticals, PeerView Communications, Prime Communications, Puerto Rican Respiratory Society, Chiesi, Roche, Sunovion, and Theravance; non-financial support from ProterrixBio; personal fees from Columbia University, Haymarket Communications, Integritas, Inthought Research, MD Magazine, Methodist Hospital Brooklyn, New York University, Unity, UpToDate, WebMD/MedScape, Western Connecticut Health Network, and American Thoracic Society. MKH reports personal fees from GSK, personal fees from BI, personal fees from AZ, other from Novartis, other from Sunovion.

Figures

Figure 1
Figure 1
Radar plot showing mean-centered and range-scaled log-transformed metabolite concentrations by exacerbation group. Asterisks (*) denote statistical significance at the 10% false discovery rate.
Figure 2
Figure 2
Volcano plot showing the adjusted association between log-transformed metabolite concentrations and incident respiratory exacerbations. Red dots represent metabolites that are statistically significant at the 10% false discovery rate. Black dots represent metabolites that are not statistically significant.
Figure 3
Figure 3
Volcano plots showing the association between log-transformed metabolite concentrations and FEV1% predicted (A), % low attenuation area (LAA) <−950 Hounsfield Units (HU) on chest computed tomography (CT) (B), and 6-minute walking distance (C). Black dots represent individual metabolites.

References

    1. Vogelmeier CF, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease 2017 Report. GOLD Executive Summary. Am J Respir Crit Care Med. 2017;195:557–582. doi: 10.1164/rccm.201701-0218PP.
    1. Han MK, et al. Frequency of exacerbations in patients with chronic obstructive pulmonary disease: an analysis of the SPIROMICS cohort. Lancet. Respir Med. 2017;5:619–626. doi: 10.1016/S2213-2600(17)30207-2.
    1. Hurst JR, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128–1138. doi: 10.1056/NEJMoa0909883.
    1. Coxson HO, et al. The presence and progression of emphysema in COPD as determined by CT scanning and biomarker expression: a prospective analysis from the ECLIPSE study. Lancet Respir Med. 2013;1:129–136. doi: 10.1016/S2213-2600(13)70006-7.
    1. Bowler RP, et al. New Strategies and Challenges in Lung Proteomics and Metabolomics. An Official American Thoracic Society Workshop Report. Ann Am Thorac Soc. 2017;14:1721–1743. doi: 10.1513/AnnalsATS.201710-770WS.
    1. Clish CB. Metabolomics: an emerging but powerful tool for precision medicine. Cold Spring Harb Mol Case Stud. 2015;1:a000588. doi: 10.1101/mcs.a000588.
    1. Chen Q, et al. Serum Metabolite Biomarkers Discriminate Healthy Smokers from COPD Smokers. PLoS One. 2015;10:e0143937. doi: 10.1371/journal.pone.0143937.
    1. de Laurentiis G, et al. Separating smoking-related diseases using NMR-based metabolomics of exhaled breath condensate. J Proteome Res. 2013;12:1502–1511. doi: 10.1021/pr301171p.
    1. Paige M, et al. Pilot analysis of the plasma metabolite profiles associated with emphysematous Chronic Obstructive Pulmonary Disease phenotype. Biochem Biophys Res Commun. 2011;413:588–593. doi: 10.1016/j.bbrc.2011.09.006.
    1. Wang L, et al. Metabonomic profiling of serum and urine by (1)H NMR-based spectroscopy discriminates patients with chronic obstructive pulmonary disease and healthy individuals. PLoS One. 2013;8:e65675. doi: 10.1371/journal.pone.0065675.
    1. De Benedetto F, et al. Supplementation with Qter((R)) and Creatine improves functional performance in COPD patients on long term oxygen therapy. Respir Med. 2018;142:86–93. doi: 10.1016/j.rmed.2018.08.002.
    1. Dransfield MT, et al. Acute exacerbations and lung function loss in smokers with and without chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2017;195:324–330. doi: 10.1164/rccm.201605-1014OC.
    1. Soler-Cataluna JJ, et al. Severe acute exacerbations and mortality in patients with chronic obstructive pulmonary disease. Thorax. 2005;60:925–931. doi: 10.1136/thx.2005.040527.
    1. Gulcev M, et al. Tryptophan catabolism in acute exacerbations of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2016;11:2435–2446. doi: 10.2147/COPD.S107844.
    1. Couper D, et al. Design of the Subpopulations and Intermediate Outcomes in COPD Study (SPIROMICS) Thorax. 2014;69:491–494. doi: 10.1136/thoraxjnl-2013-203897.
    1. Wu G. Amino acids: metabolism, functions, and nutrition. Amino Acids. 2009;37:1–17. doi: 10.1007/s00726-009-0269-0.
    1. Ubhi BK, et al. Targeted metabolomics identifies perturbations in amino acid metabolism that sub-classify patients with COPD. Mol Biosyst. 2012;8:3125–3133. doi: 10.1039/c2mb25194a.
    1. Pouw EM, Schols AM, Deutz NE, Wouters EF. Plasma and muscle amino acid levels in relation to resting energy expenditure and inflammation in stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;158:797–801. doi: 10.1164/ajrccm.158.3.9708097.
    1. Cruickshank-Quinn CI, et al. Metabolomics and transcriptomics pathway approach reveals outcome-specific perturbations in COPD. Sci Rep. 2018;8:17132. doi: 10.1038/s41598-018-35372-w.
    1. Cervenka Igor, Agudelo Leandro Z., Ruas Jorge L. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science. 2017;357(6349):eaaf9794. doi: 10.1126/science.aaf9794.
    1. Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004;4:762–774. doi: 10.1038/nri1457.
    1. Puccetti M, et al. Towards Targeting the Aryl Hydrocarbon Receptor in Cystic Fibrosis. Mediators Inflamm. 2018;2018:1601486. doi: 10.1155/2018/1601486.
    1. Paliogiannis Panagiotis, Fois Alessandro G., Sotgia Salvatore, Mangoni Arduino A., Zinellu Elisabetta, Pirina Pietro, Negri Silvia, Carru Ciriaco, Zinellu Angelo. Neutrophil to lymphocyte ratio and clinical outcomes in COPD: recent evidence and future perspectives. European Respiratory Review. 2018;27(147):170113. doi: 10.1183/16000617.0113-2017.
    1. Li P, Yin YL, Li D, Kim SW, Wu G. Amino acids and immune function. Br J Nutr. 2007;98:237–252. doi: 10.1017/S000711450769936X.
    1. Hurst JR, et al. Susceptibility to exacerbation in chronic obstructive pulmonary disease. N Engl J Med. 2010;363:1128–1138. doi: 10.1056/NEJMoa0909883.
    1. McDonald ML, et al. Quantitative computed tomography measures of pectoralis muscle area and disease severity in chronic obstructive pulmonary disease. A cross-sectional study. Ann Am Thorac Soc. 2014;11:326–334. doi: 10.1513/AnnalsATS.201307-229OC.
    1. Layman DK, Walker DA. Potential importance of leucine in treatment of obesity and the metabolic syndrome. J Nutr. 2006;136:319S–323S. doi: 10.1093/jn/136.1.319S.
    1. Ubhi BK, et al. Metabolic profiling detects biomarkers of protein degradation in COPD patients. Eur Respir J. 2012;40:345–355. doi: 10.1183/09031936.00112411.
    1. Hofford JM, et al. The nutritional status in advanced emphysema associated with chronic bronchitis. A study of amino acid and catecholamine levels. Am Rev Respir Dis. 1990;141:902–908. doi: 10.1164/ajrccm/141.4_Pt_1.902.
    1. Engelen MP, Wouters EF, Deutz NE, Menheere PP, Schols AM. Factors contributing to alterations in skeletal muscle and plasma amino acid profiles in patients with chronic obstructive pulmonary disease. Am J Clin Nutr. 2000;72:1480–1487. doi: 10.1093/ajcn/72.6.1480.
    1. Celli BR, et al. The body-mass index, airflow obstruction, dyspnea, and exercise capacity index in chronic obstructive pulmonary disease. N Engl J Med. 2004;350:1005–1012. doi: 10.1056/NEJMoa021322.
    1. Patel MJ, et al. Race and sex differences in small-molecule metabolites and metabolic hormones in overweight and obese adults. Omics. 2013;17:627–635. doi: 10.1089/omi.2013.0031.
    1. Darst BF, Koscik RL, Hogan KJ, Johnson SC, Engelman CD. Longitudinal plasma metabolomics of aging and sex. Aging (Albany NY) 2019;11:1262–1282. doi: 10.18632/aging.101837.
    1. Miller MR, et al. Standardisation of spirometry. Eur Respir J. 2005;26:319–338. doi: 10.1183/09031936.05.00034805.
    1. Sieren JP, et al. SPIROMICS Protocol for Multicenter Quantitative Computed Tomography to Phenotype the Lungs. Am J Respir Crit Care Med. 2016;194:794–806. doi: 10.1164/rccm.201506-1208PP.
    1. Gevenois PA, de Maertelaer V, De Vuyst P, Zanen J, Yernault JC. Comparison of computed density and macroscopic morphometry in pulmonary emphysema. Am J Respir Crit Care Med. 1995;152:653–657. doi: 10.1164/ajrccm.152.2.7633722.
    1. Woodruff PG, et al. Clinical Significance of Symptoms in Smokers with Preserved Pulmonary Function. N Engl J Med. 2016;374:1811–1821. doi: 10.1056/NEJMoa1505971.
    1. Bowler RP, et al. Prediction of acute respiratory disease in current and former smokers with and without COPD. Chest. 2014;146:941–950. doi: 10.1378/chest.13-2946.
    1. Nagana Gowda GA, Gowda YN, Raftery D. Expanding the limits of human blood metabolite quantitation using NMR spectroscopy. Anal Chem. 2015;87:706–715. doi: 10.1021/ac503651e.
    1. McHugh Cora, Flott Thomas, Schooff Casey, Smiley Zyad, Puskarich Michael, Myers Daniel, Younger John, Jones Alan, Stringer Kathleen. Rapid, Reproducible, Quantifiable NMR Metabolomics: Methanol and Methanol: Chloroform Precipitation for Removal of Macromolecules in Serum and Whole Blood. Metabolites. 2018;8(4):93. doi: 10.3390/metabo8040093.
    1. Lacy P, et al. Signal intensities derived from different NMR probes and parameters contribute to variations in quantification of metabolites. PLoS One. 2014;9:e85732. doi: 10.1371/journal.pone.0085732.
    1. Reinke Stacey N., Gallart-Ayala Héctor, Gómez Cristina, Checa Antonio, Fauland Alexander, Naz Shama, Kamleh Muhammad Anas, Djukanović Ratko, Hinks Timothy S.C., Wheelock Craig E. Metabolomics analysis identifies different metabotypes of asthma severity. European Respiratory Journal. 2017;49(3):1601740. doi: 10.1183/13993003.01740-2016.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate - a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B-Methodological. 1995;57:289–300.

Source: PubMed

3
订阅