Skip to main content
Erschienen in: Respiratory Research 1/2023

Open Access 01.12.2023 | Research

Proteomics and metabolomics profiling reveal panels of circulating diagnostic biomarkers and molecular subtypes in stable COPD

verfasst von: Zili Zhang, Jian Wang, Yuanyuan Li, Fei Liu, Lingdan Chen, Shunping He, Fanjie Lin, Xinguang Wei, Yaowei Fang, Qiongqiong Li, Juntuo Zhou, Wenju Lu

Erschienen in: Respiratory Research | Ausgabe 1/2023

Abstract

Background

Chronic obstructive pulmonary disease (COPD) is a complex and heterogeneous disease with high morbidity and mortality, especially in advanced patients. We aimed to develop multi-omics panels of biomarkers for the diagnosis and explore its molecular subtypes.

Methods

A total of 40 stable patients with advanced COPD and 40 controls were enrolled in the study. Proteomics and metabolomics techniques were applied to identify potential biomarkers. An additional 29 COPD and 31 controls were enrolled for validation of the obtained proteomic signatures. Information on demographic, clinical manifestation, and blood test were collected. The ROC analyses were carried out to evaluate the diagnostic performance, and experimentally validated the final biomarkers on mild-to-moderate COPD. Next, molecular subtyping was performed using proteomics data.

Results

Theophylline, palmitoylethanolamide, hypoxanthine, and cadherin 5 (CDH5) could effectively diagnose advanced COPD with high accuracy (auROC = 0.98, sensitivity of 0.94, and specificity of 0.95). The performance of the diagnostic panel was superior to that of other single/combined results and blood tests. Proteome based stratification of COPD revealed three subtypes (I–III) related to different clinical outcomes and molecular feature: simplex COPD, COPD co-existing with bronchiectasis, and COPD largely co-existing with metabolic syndrome, respectively. Two discriminant models were established using the auROC of 0.96 (Principal Component Analysis, PCA) and 0.95 (the combination of RRM1 + SUPV3L1 + KRT78) in differentiating COPD and COPD with co-morbidities. Theophylline and CDH5 were exclusively elevated in advanced COPD but not in its mild form.

Conclusions

This integrative multi-omics analysis provides a more comprehensive understanding of the molecular landscape of advanced COPD, which may suggest molecular targets for specialized therapy.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12931-023-02349-x.
Zili Zhang, Jian Wang, Yuanyuan Li and Fei Liu contributed equally to this work

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
COPD
Chronic obstructive pulmonary disease
PCA
Principal Component Analysis
CDH5
Cadherin 5
RRM1
Ribonucleotide reductase catalytic subunit M1
SUPV3L1
Suv3 like RNA helicase
KRT78
Keratin 78
CRD
Chronic respiratory disease
COPD-BE
COPD co-existing with bronchiectasis
COPD-MD
COPD co-existing with metabolic syndrome
MPV
Mean platelet volume
DEPs
Differentially expressed proteins
GO
Gene Ontology
KEGG
Kyoto Encyclopedia of Genes and Genomes
HP
Haptoglobin
HBB
Hemoglobin subunit beta
VCL
Vinculin
HBA1
Hemoglobin subunit alpha 1
CA1/2
Carbonic anhydrase 1/2
SOD1
Superoxide dismutase 1
FGA
Fibrinogen alpha chain
PRDX2
Peroxiredoxin 2
ALDOA
Aldolase, fructose-bisphosphate A
TNC
Tenascin C
CAT
Catalase
PRM
Parallel Reaction Monitoring
B4GAT1
Beta-1,4-glucuronyltransferase 1
GNPTG
N-Acetylglucosamine-1-phosphate transferase subunit gamma
ADAMTSL4
ADAMTS like 4
CFP
Complement factor properdin
EXTL2
Exostosin like glycosyltransferase 2
SAA1
Serum amyloid A1
CRP
C-reactive protein
ORM1/2
Orosomucoid 1/2
DEMs
Differentially expressed metabolites
WGCNA
Weighted gene co-expression network analysis
BALF
Bronchoalveolar lavage fluid
RAB26
Member RAS oncogene family
SERPINA1
Serpin family A member 1
GOLD
Global Initiative for Obstructive Lung Disease
PBMCs
Peripheral blood mononuclear cells

Introduction

Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by chronic inflammation, airway obstruction, and destruction of the parenchyma. It is the fourth leading cause of death globally, and is projected to be the third leading cause of mortality by 2030 [20]. Current therapies for patients with advanced COPD mainly treat symptoms such as chronic cough and excessive sputum production, as well as prevent disease progression. However, 46–91% of adults still suffer from persistent and disabling breathlessness at rest and on minimal exertion [25]. To date, no therapy has been developed for reducing disease progression and lower mortality rates. Therefore, additional approaches for accurate diagnoses of advanced COPD are urgently needed.
In 2011, the Society for Qualification of Biomarkers for COPD was established to accelerate research and development of biomarkers. To date, however, only a handful of biomarkers associated with COPD have been discovered [21]. Integrated multi-omics data analysis can provide insights into the pathological mechanisms of COPD. Analysis of the proteome can provide studying disease-related mechanisms and diagnostic biomarkers, which reveals disease phenotype [21]. Compared to traditional proteomic techniques, TMT-LC–MS/MS is a more comprehensive and efficient method for capturing and quantification of proteins, with a smaller sample requirement without offset. In addition, the metabolome, which is defined as the total collection of small molecular metabolites present in a given type of cell or organism, is the final downstream product of metabolism. Particularly, it provides an exact reflection of the current metabolic status of the organic body. To date, some progress has been made in the fields of functional proteomics and metabolomics. For example, researchers have applied proteomic approaches to identify novel biomarkers, such as plasma sRAGE for detecting presence and progression of emphysema [33], whereas others have adopted metabolomics approaches to identify potential disease severity markers or therapeutic candidates such as purines [8], sphingolipids [2], and glycerol phospholipids [4]. However, no discovery-based approach has yet resulted in validated clinical biomarkers. Although findings from these omics-centric studies have added to the existing knowledge base, there are several gaps that are yet to be filled. We hypothesize that integrating contemporary proteomics and metabolomics approaches can effectively evaluate metabolic pathways and diagnostic biomarkers in advanced COPD. Moreover, most of the previous multi-omics studies have focused on patients derived from European, American, and African populations [24]. Therefore, it is important to systematically analyze the metabolic and proteomic profile of Chinese patient cohorts to generate new insights for this region.
Patients with COPD are often predisposed to various co-morbidities, such as cardiovascular disease, metabolic syndrome, and bronchiectasis [15, 20, 22]. Additionally, smoking is a risk factor for such co-morbidities, with previous evidence showing that some smokers develop a predominately emphysema phenotype, characterized by alveolar damage, while others developing predominantly airway disease. Evidence from other studies has shown that proteases, inflammation, oxidative stress, immune defects, and infections play a role in the development and progression of COPD [25]. Since COPD is a heterogeneous disease, grading the severity and identifying phenotypes according to the concomitant diseases (i.e., subpopulations of subjects with similar disease characteristics) can expand our understanding of the biological mechanisms underlying the disease’s development and progression. This will facilitate accurate diagnoses of the disease. Particularly, lowering mortality rates in patients with advanced COPD relies on early and accurate diagnosis and differentiation of different subtypes using simple and objective diagnostic assessments. The heterogeneity of COPD also exists at the molecular level, and thus molecular sub-phenotyping is the first and crucial step in the identification and classification of these subgroups. Previous studies have shown that omics approaches, based on appropriate sample sizes, can not only efficiently reveal heterogeneity of these subtypes but also facilitate diagnosis and reveal the exact mechanisms underlying COPD subgroups [22, 24]. Proteomics techniques, based on mass spectrometry, have shown strong power in detecting disease phenotypes.
In this study, we hypothesized that changes in proteomic and metabolic profiles of patients with stable COPD would produce a unique pattern of molecules compared to those without COPD, and that these molecular profiles would change with disease complications. Therefore, we first performed quantitative shotgun proteomic analyses to investigate COPD-related proteins molecular portrait and reveal COPD-related functional modulation. Next, we applied a targeted proteomics approach to validate specific members of dysregulated proteins in another independent sample set. In addition, untargeted metabolomics was performed using the same participants as the proteome. Our findings not only reveal the profiles of COPD biomarkers and molecular subtypes, but also provide data that will guide future studies seeking to develop tools for clinical application.

Methods

Biospecimen collection and clinical data

The previously published design of GIRD COPD Biobank (Clinical trial www.​chictr.​org.​cn, number ChiCTR-CCC-12002950) was adopted in this study [19]. The GIRD COPD Biobank collection, which was established in 2010, comprises specimens from patients and controls for research purposes. All patients were diagnosed with COPD through pulmonary function testing and clinical symptoms. The enrolled patients and controls were aged between 40 and 80 years, and were permanent residents (lived in Guangzhou for years). Permission to access medical records was sought from each individual, after which his/her management information was retrieved before and after hospitalization. The participants were also requested to provide blood samples and other materials for research purposes. The cross-sectional analysis presented in this work is based on clinical and biomarker data obtained at baseline.
Inclusion criteria of patients were as follows: male subjects aged 55–75 years, had a Global Initiative for Obstructive Lung Disease (GOLD) stage 1–4, and were current or ex-smokers with a smoking history of greater than or equal to 10 packyears, as well the patients were all stable COPD without therapies including steroid, theophylline, antibiotics etc. at least 1 week before joining the group. The exclusion criteria were: patients suffering from lung disease except extensive bronchiectasis, such as cystic fibrosis, and pulmonary fibrosis; with other inflammatory diseases, or reported COPD exacerbation within 4 weeks of enrollment. Participants were assigned to the control group if they had normal spirometry, cancer-free without suffering from any lung disease, aged between 40 and 80 years, current or ex-smokers, and were permanent residents of Guangzhou (lived in Guangzhou for several years).
Finally, 70 advanced COPD patients and 70 healthy controls were recruited for multi-omics study. Additional 10 mild-to-moderate COPD and 11 healthy controls were enrolled for protein biomarker validation. For the validation of final metabolites, the results of 46 mild-to-moderate COPD and 48 healthy controls were obtained with the help of professor Zhou [36]. For proteomics analysis, subjects were divided into a discovery (comprising 40 COPD patents and 40 controls) and validation (30 COPD patients and 30 controls) groups. Metabolomics analyses were only performed on the discovery cohort. Blood samples were obtained from all participants before breakfast, and immediately processed according to a previously published standard protocol [19]. Briefly, blood was collected into serum separating tubes (SST, Vacutainer SST II Tube 8.5 mL, #368972; BD), manually inverted 10 times, then centrifuged for 10 min at 1300×g. Serum samples were aliquoted and stored at − 80 °C until proteomics analysis. Peripheral Blood Mononuclear cells (PBMCs) were isolated using lymphocyte separation medium as described previously [34]. The study design is shown in Fig. 1.
Furthermore, we collected each patient’s demographic, clinical manifestation, anthropometric information, individual and family disease history, and other co-morbidities. Moreover, information on other parameters, such as indoor living and working environment, dietary habits and smoking habits was collected. Family history of cancer was defined as any self-reported cancer in his/her first-degree relatives, such as parents, siblings, or children. A summary of the characteristics of all COPD patients and control subjects with complete data is presented in Table 1 in discovery stage. Each patient voluntarily provided a written informed consent prior to inclusion in the study and data collection. The study was approved by the Institutional Review Board of Guangzhou Medical University, Ethics Committee of the First Affiliated Hospital (approval number: GZMC 2009-08-1336), and was conducted in accordance with the principles of the Declaration of Helsinki.
Table 1
Characteristics of proteomics cohorts in discovery stage and in validation stage
 
Discovery stage
Pa
Validation stage
Pa
COPD
Controls
COPD
Controls
N
40
40
 
29
31
 
Male, %
100
100
1.0
100
100
1.0
Smoking, %
100
100
1.0
100
100
1.0
Packyears ≥ 30, %
100
100
1.0
100
100
1.0
Age (years), mean (SD)
64.6 (11.7)
63.7 (5.3)
0.656
   
Height (cm), mean (SD)
165.0 (5.2)
166.0 (5.9)
0.824
164 (4.9)
169 (5.9)
< 0.001
Weight (kg), mean (SD)
63.1 (12.5)
65.7 (8.2)
0.266
60.60 (9.3)
69.10 (9.4)
0.001
BMI (kg/m2), mean (SD)
23.8 (2.9)
23.1 (4.6)
 
22.40 (3.2)
23.90 (2.5)
0.054
Fan in kitchen, %
93.2
92.7
0.475
92.3
93.6
1.000
Good room ventilation, %
38.6
63.4
0.022
62.1
51.6
0.414
Often preserved food, %
18.8
7.1
0.106
10.3
3.2
0.346
Often cook, %
25.0
29.3
0.658
17.2
35.5
0.110
Comorbidity, %
 CRD
29.8
12.2
0.045
34.6
3.2
0.002
 Hypertension
42.6
31.7
0.294
34.6
22.6
0.314
 Diabetes
12.8
7.5
0.498
0
10
 Heart diseases
40.4
12.5
0.004
19.2
6.7
0.231
 Stroke
4.3
0
3.8
0
Family history, %
 Cancer
6.3
9.5
0.563
23.1
22.6
0.965
 RD without COPD
19.1
15.8
0.759
7.1
3.2
 Severe COPD, %
100
0
100
0
Pulmonary function, mean (SD)
 pre_FVC_%Pred
74.9 (28.1)
98.0 (15.0)
< 0.001
78.5 (16.3)
94.3 (10.4)
< 0.001
 pre_FEV1_%Pred
59.7 (32.1)
88.4 (26.1)
< 0.001
47.1 (14.3)
94.5 (9.6)
< 0.001
 pre_FEV1/FVC_%Pred
62.7 (15.9)
71.3 (15.5)
0.036
57.4 (15.8)
96.8 (9.7)
< 0.001
 post_FVC_%Pred
74.6 (24.6)
99.8 (12.5)
< 0.001
88.3 (14.6)
104 (13.0)
0.061
 post_FEV1_%Pred
57.1 (28.6)
90.8 (25.4)
< 0.001
53.8 (15.8)
101 (10.4)
< 0.001
 post_FEV1/FVC_%Pred
60.7 (15.2)
70.9 (15.4)
0.021
57.7 (15.4)
76.9 (6.3)
0.025
Bold values indicate significant differences
CRD chronic respiratory disease, severe COPD Global Initiative for Obstructive Lung Disease stage 3–4
aP-values for a two-sided χ2 test or t-test. Data are median (P25–P75), n (%)

Proteomics analysis

Protein extraction and trypsin digestion of identified proteomics

Cellular debris were first removed from serum samples via a 10-min centrifugation at 12,000×g at 4 °C, and the supernatant transferred to a new centrifuge tube. The top 12 high abundance proteins were then removed by Pierce™ Top 12 Abundant Protein Depletion Spin Columns Kit (Thermo Fisher), and the protein concentration determined using the BCA kit according to the manufacturer’s instructions. For digestion, the protein solution was reduced by treating it with 5 mM dithiothreitol for 30 min at 56 °C, then alkylated with 11 mM iodoacetamide for 15 min at room temperature in darkness. Next, the prote in sample was diluted by adding 100 mM TEAB to urea concentration less than 2 M. Trypsin, at 1:50 trypsin-to-protein mass ratio, was added for the first digestion overnight, followed by 1:100 trypsin-to-protein for a second 4 h-digestion. After tryps indigestion, peptide was desalted using Strata X C18 SPE columns (Phenomenex), vacuum-dried, then reconstituted in 0.5 M TEAB and labeled for TMT pro11 plexkit according to the manufacturer’s protocol.

Liquid chromatography–mass spectrometry (LC–MS/MS)

The tryptic peptides were fractionated into fractions by high pH reverse-phase HPLC using Thermo Betasil C18 column (5 μm particles, 10 mm ID, 250 mm length). The peptides were first separated with a gradient of 8% to 32% acetonitrile (pH 9.0) over 60 min into 60 fractions. Then, the peptides were combined into 6 fractions and dried by vacuum centrifuging. An electrospray, at a voltage of 2.0 kV, was applied with a m/z scan range of 350 to 1800 for full scan, while intact peptides were detected in the Orbitrap at a resolution of 70,000. The peptides were then selected for MS/MS using NCE setting as 28, while the fragments were detected in the Orbitrap at a resolution of 17,500. A data-dependent procedure, which alternated between one MS scan followed by 20 MS/MS scans with 15.0 s dynamic exclusion, was also applied. Automatic gain control (AGC) was set at 5E4, while the fixed first mass was set at 100 m/z.

Analysis of proteomics data

Differentially expressed proteins (DEPs) were identified using the empirical Bayesian algorithm implemented in the limma package in R software. Up-regulated and down-regulated proteins were defined by a fold change of ≥ 1.2 or ≤ 0.83 and a P-value < 0.05. We performed Gene Ontology (GO) annotation of the proteome, using the UniProt-GOA database (http://​www.​ebi.​ac.​uk/​GOA), then identified enriched pathways using the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Next, we used the “heatmap.2” function in “ggplot” package in R to perform hierarchical clustering and visualization of the DEPs. We used the GO terms to classify the proteins into three categories, namely biological process, cellular components, and molecular function. Accession numbers for all DEPs or their sequences were searched against the STRING database version 10.1 for protein–protein interactions (PPI). The STRING algorithm uses a metric called “confidence score” to define interaction confidence. Thus, we fetched all interactions with a confidence score of ≥ 0.7 (high confidence). The resulting interaction was among proteins was visualized using the “networkD3” package in R.
Principal Component Analysis (PCA) is a data reduction technique used to convert a large set of variables into a smaller set which still contains most of the original information. Principal components were extracted as a linear combination of the variables. This variance was then removed and a second linear combination was built, which iteratively explains the maximum proportion of the whole information. This is called the principal axis method, which leads to orthogonal (uncorrelated) factors. Furthermore, it involves the computation of eigenvalues and eigenvectors of covariance matrices. These eigenvectors were sorted in the descending order of their eigenvalues, followed by the actual data [13].

Validation using targeted proteomics analysis

For validation, 30 μL of serum, collected as earlier described, was treated with Pierce™ Top 12 Abundant Protein Depletion Spin Columns Kit (Thermo Fisher) according to the manufacturer’s instructions. The mixture was digested with trypsin in a similar fashion to the discovery study. All samples were analyzed via LC–MS, operated under the parallel-reaction monitoring (PRM) acquisition scheme. PRM data were analyzed using Skyline (v.3.6) to identify transitions and peak area integration, while protein intensities were Log2 transformed. Proteins with missing values, in more than 60% samples, were excluded, while the remaining missing values were considered to be low abundance due to limited MS sensitivity. Therefore, we replaced them using random numbers drawn from a normal distribution with a mean value 1.8× lower and a standard deviation 0.3× of the original data.

Metabolomics analyses

Untargeted metabolomics analysis using UPLC-Q-TOF/MS

Metabolic profiling of serum samples was performed on an Agilent 1290 Infinity LC system (Agilent Technologies, Santa-Clara, California, USA), coupled with an AB SCIEX Triple TOF 6600 System (AB SCIEX, Framingham, MA, USA). Chromatographic separation and aqueous phase of extracts used for both positive and negative models, was implemented on ACQUITY HSS T3 1.8 μm (2.1 × 100 mm) columns with a temperature of 25 °C. The mobile phases, comprising 0.1% formic acid in water (A) and 0.1% formic acid in acetonitrile (B), were used in the positive ionization mode, while 0.5 mM ammonium fluoride in water (C) and acetonitrile (D) were used in negative ionization mode. In the positive (negative) model, the elution gradient initially started with 1% B (D) for 1 min, linearly increased to 100% B (D) at 8 min, where it was maintained for 2 min, then returned to 1% B (D) for about 2 min of equilibrium. Delivery was achieved at a flow rate of 300 μL/min, and 2 μL aliquot of each sample injected onto the column. TOF/MS was performed on both positive and negative ion modes. We applied the information-dependent acquisition (IDA), an artificial intelligence-based production scan mode, for detection and identification of MS/MS spectra.

Metabolomics data analysis

The datasets were normalized and integrated using support vector regression, then uploaded into the Metabo Analyst software for further analysis (www. metaboanalyst.ca). Datasets from both positive and negative models were log-transformed and pareto-scaled. Next, they were subjected to principal component analysis (PCA) and partial least square discriminant analysis (PLS-DA). We calculated variable importance in the projection (VIP) value, for each variable in the PLS-DA model, to determine its contribution to the classification. Metabolites with the VIP value > 1 were further analyzed using the Student t-test at the univariate level to determine the significance of each metabolite. Differences at P-value < 0.05 were considered statistically significant. The secondary metabolites screened by metabolomics were analyzed using Spearman correlation. R language and Cytoscape software were jointly used to analyze the matrix heat map, hierarchical clustering, association network, and other variables.

Validation of expressions or regulatory roles of prioritized molecules upon mild-to-moderate COPD

The total RNA was extracted from PBMCs obtained from patients with COPD and healthy individuals using Trizol reagent (Invitrogen). It was reversely transcribed to cDNA using PrimeScript™ RT reagent Kit (TaKaRa, China). The qRT-PCR assay was performed on CFX96-C1000 system (Bio-Rad, CA) using SsoFast™ EvaGreen® supermix kit (Bio-Rad). Primers used for qRT-PCR were as follows: human CDH5: 5′-ATGAGATCGTGGTGGAAGCG-3′ (forward), 5′-TGTGTACTTGGTCTGGGTGA AG′ (reverse); human GAPDH: 5′-ACAACTTTG GTATCGTGGAAGG-3′ (forward), 5′-GCCATCACGCCACAGTTT C-3′ (reverse). The relative expression of each gene was normalized to GAPDH expression and calculated using the 2−△△Ct method. Validation of metabolites (theophylline and hypoxanthine) was performed on an Ultimate 3000 UHPLC system coupled with Q-Exactive MS (Thermo Scientific).

Statistical analysis

Data of quantitative and categorical traits were analyzed using the Mann–Whitney U and t tests, with P < 0.05 considered statistically significant. The combined values for diagnosing disease severity were calculated by binary logistic regression using a stepwise method (with a variable entered and removed if P < 0.05 and P > 0.1, respectively). The accuracy of each independent or combined indexes was determined using the auROC. The optimal threshold value was obtained by calculating the correct classification ratio (CCR). PCA seeks a linear combination of variables such that the principal components (PC) can be extracted. Loadings from the first and second PCs were used to form the weighted component scores (Y1, Y2) as a linear combination of the original 12 variables for each participant. Eigen equations showed by Y1, Y2 were combined to obtain a composite disease expression score (Y3 = |Y1 λ1| + |Y2 λ2|) where λi is the variance explained by each PC (eigenvalue) that accounts for most of the variation. All statistical analyses were performed using packages implemented in R (v3.2.0).

Results

Clinical characteristics of participants

Blood samples were collected from 70 patients with advanced COPD and 70 healthy controls from GIRD COPD Biobank (Table 1). There were statistically significant differences between COPD patients and healthy controls in terms of room ventilation, heart diseases, and pulmonary function (P < 0.05), but sex, smoking, pack years, age, height, weight, BMI, fan in kitchen, preserved food consumption, cooking, other comorbidities, and family history were not significantly different between the two groups (P > 0.05). Additional proteomics analysis was conducted using an independent validation set comprising COPD patents (n = 29) and controls (n = 31) (Table 1 on the validation stage). Results indicated that COPD patients had significantly lower mean height, weight, and pulmonary function than healthy individuals (Pmax = 0.001). Moreover, COPD patients, but not healthy controls, reported complications related to chronic respiratory disease (CRD) at admission (P = 0.02). Pulmonary function (except post-FVC_%Pred) and mean platelet volume (MPV) were significantly lower in COPD patients than in healthy individuals (Pmax = 0.025). Similarly, COPD patients exhibited significantly higher monocytes than healthy subjects (P = 0.027) (Table 2). Next, we used proteomics results to stratify COPD patients into 3 subgroups (Table 3): subtype I were mainly COPD without other respiratory diseases (simplex COPD, n = 19), subtype II largely for COPD co-existing with bronchiectasis (COPD-BE, n = 9), and subtype III focused on COPD co-existing with metabolic syndrome (COPD-MD, n = 12). Further analysis revealed that participants in the COPD-BE and COPD-MD groups had significantly lower room ventilation and COPD without chronic respiratory disease than the simplex COPD (P = 0.002 and 0.014, respectively), while those in the COPD-BE group had significantly lower pre_FEV1_%Pred relative to those in other groups (P = 0.025).
Table 2
Blood count of validation cohorts for targeted proteomics
 
COPDb
Controls
Pa
N
29
31
 
Male, %
100
100
1.0
Smoking, %
100
100
1.0
Pack_years ≥ 30, %
100
100
1.0
Severe COPD, %
100
0
White blood cells, WBC
7.55 (1.91)
6.76 (1.8)
0.11
Neutrophil ratio
62.15 (8.25)
59.67 (8.3)
0.25
Lymphocyte ratio
24.95 (7.54)
28.98 (7.52)
0.04
Monocyte ratio
8.77 (2.27)
7.77 (2.09)
0.09
Eosinophil ratio
3.47 (2.89)
2.88 (1.66)
0.34
Basophilic cell ratio
0.66 (0.24)
0.71 (0.23)
0.44
nucleated red cells ratio
0.21 (0.72)
0.10 (0.07)
0.42
Neutrophil count
4.59 (1.47)
4.05 (1.32)
0.15
Lymphocyte count
1.85 (0.8)
1.94 (0.78)
0.66
Monocyte countb
0.65 (0.28)
0.52 (0.15)
0.027
Eosinophil count
0.26 (0.27)
0.19 (0.13)
0.24
Basophil count
0.22 (0.94)
0.04 (0.05)
0.28
NRBC
5.59 (30.08)
0.01 (0.01)
0.31
Red blood cells, RBC
4.84 (1.0)
4.80 (0.51)
0.85
Hemoglobin, HGB
141.53 (16.42)
145.3 (11.13)
0.31
Hematocrit, HCT
3.29 (11.02)
3.19 (10.43)
0.97
Mean corpuscular volume, MCV
97.17 (47.81)
94.64 (8.52)
0.77
Mean hemoglobin content, MCH
29.45 (4.33)
30.54 (3.16)
0.27
Mean hemoglobin concentration, MCHC
326.9 (10.73)
322.4 (11.9)
0.13
RDW-SD
43.94 (4.89)
43.99 (3.39)
0.96
RDW-CV
14.14 (1.08)
13.56 (1.06)
0.04
Platelet, PLT
255.9 (57.97)
236.1 (60.46)
0.20
Mean platelet volume, MPV
8.14 (1.04)
8.76 (0.94)
0.019
PDW
16.50 (1.2)
16.19 (1.65)
0.41
PCT
0.21 (0.04)
0.20 (0.05)
0.97
CRD chronic respiratory disease, RDW-SD standard deviation of RBC distribution width, RDW-CV coefficient variation of RBC distribution width, PDW platelet distribution width, PCT thrombocytocrit, NRBC nucleated red cells absolute value, severe COPD Global Initiative for Obstructive Lung Disease stage 3–4
In bold is P < 0.05
aP values for a two-sided χ2 test or t-test. Data are median (P25–P75), n (%)
bCOPD patients exhibited significantly higher monocytes than healthy subjects
Table 3
Characteristics of proteomics-driven subtype cohorts by using the discovery proteomics data
 
COPD
Pa
COPD
COPD-BE
COPD-MD
N
19
9
12
 
Male, %
100
100
100
1.0
Smoking, %
100
100
100
1.0
Packyears ≥ 30, %
100
100
100
1.0
Age (years), mean (SD)
59.8
67.8
63.6
0.22
Height (cm), mean (SD)
167
164
165
0.54
Weight (kg), mean (SD)
87.3
53.1
85.7
0.17
BMI (kg/m2), mean (SD)
32.2
18.8
32.0
0.22
Fan in kitchen, %
17 (94.4)
8 (100)
12 (100)
1.00
Good room ventilation, %
13 (72.2)
1 (12.5)
2 (16.7)
0.002
Often preserved food, %
3 (16.7)
0
0
0.54
Often cook, %
5 (27.8)
3 (37.5)
3 (25)
0.88
Comorbidity, %
 CRD
6 (31.6)
3 (33.3)
4 (33.3)
0.99
 Hypertension
8 (42.1)
3 (33.3)
5 (41.7)
0.90
 Diabetes
4 (21.1)
1 (11.1)
1 (8.3)
0.85
 Heart diseases
6 (31.6)
3 (33.3)
5 (41.7)
0.84
 Stroke
2 (10.5)
0
0
0.71
Family history, %
 Cancer
3 (15.8)
0
0
0.30
 CRD without COPD
6 (42.8)
1.0 (11.1)
0
0.014
Severe COPD%
100
100
100
1.0
 pre_FVC_%Pred
81.1 (29.5)
53.2 (12.0)
63.4 (19.7)
0.19
 pre_FEV1_%Pred
69.8 (31.5)
24.1 (4.4)
41.0 (16.9)
0.025
 pre_FEV1/FVC_%Pred
81.8 (29.5)
53.2 (12.1)
63.4 (19.7)
0.19
 post_FVC_%Pred
77.5 (28.4)
64.0 (4.4)
71.7 (17.6)
0.69
 post_FEV1_%Pred
66.1 (29.1)
29.3 (7.3)
44.1 (18.1)
0.07
 post_FEV1/FVC_%Pred
77.5 (28.4)
64.0 (4.4)
71.7 (17.6)
0.69
Bold values indicate significant differences
CRD chronic respiratory disease, severe COPD Global Initiative for Obstructive Lung Disease stage 3–4, COPD-BE COPD co-existing with bronchiectasis, COPD-MD COPD co-existing with metabolic syndrome
aP-values for a two-sided χ2 test or oneway-ANOVA, or Fisher’s exact test, non-parametric test.as appropriate. Data are median (P25–P75), n (%)

Proteomic profiles and functional alterations related to COPD

Serum samples were obtained from 40 patients with advanced COPD and 40 healthy controls for TMT-labeled proteomic analysis. The proteomic patterns of serum from COPD patients were distinct from those of serum obtained from healthy controls. A total of 1432 proteins were identified and quantified. Quality control analysis was carried out, the lengths and mass errors of peptides, as well as coverage and sequence distribution of the proteins were calculated (Additional file 1: Fig. S1A–D). Consequently, 251 differentially expressed proteins (DEPs) were identified between the two groups, of which 151 and 100 were significantly up-regulated and down-regulated, respectively (fold change ≥ 1.2 or ≤ 0.83 and a P < 0.05) (Additional file 1: Fig. S1E). Moreover, 31.43% of these proteins were involved in extracellular matrix, whereas 29.29 and 18.57% among them regulated functions in the cytoplasm and nucleus, respectively (Additional file 1: Fig. S1F). The DEPs were divided into Q1–Q4 according to the multiple of fold change, and the heatmap of enrichment analysis (GO and KEGG) shown in Additional file 2: Fig. S2. In sum, the quality control analysis showed the data were acquired with a high degree of consistency and reproducibility, and the significantly up-regulated DEPs indicated a generally activated effect of biological processes in COPD.
The detailed data processing protocols for COPD and healthy controls are showed in Fig. 2A. In total, 251 dysregulated DEPs were identified between the two groups (Fig. 2B). For biological processes, these proteins were mainly involved in immune response, myeloid leukocyte activation, neutrophil mediated immunity, granulocyte activation, platelet activation, and homotypic cell–cell adhesion. For molecular functions, the proteins were primarily involved in seven function processes, namely identical protein binding, structural molecule activity, cadherin binding, oxidoreductase activity, structural constituent of cytoskeleton, tetrapyrrole binding, and heme binding. Most of these proteins were located in vesicle lumen and secretory granule (Fig. 2C). Results from KEGG pathway analyses revealed that these DEPs were significantly enriched in carbon metabolism, and glycolysis/gluconeogenesis. Heatmap analyses showed higher antioxidant activity and activated glycosaminoglycan binding in COPD compared to healthy controls (Fig. 2D). Among these DEPs, the final dysregulated proteins were selected (Fig. 2E), and validated by targeted proteomics according to differential significance levels, including ORM1, HP, HBB, VCL, TPIA, HBA1, CA2, SOD1, FGA, PRDX2, CDH5, ALDOA, CA1, TNC, CAT, and LRG1 (Fig. 2F). In final, 16 DEPs were selected associated with COPD compared with healthy controls.

Protein validation via targeted proteomics (PRM)

The COPD-related proteomics and functional alteration results from the discovery study were then used to develop protein marker panels for accurate prediction of severity of COPD. Thus, we analyzed members of upregulated functional groups based on top Pmin value and fold changes (max). Finally, 16 DEPs with confident quantitation data were validated in an additional cohort comprising 29 COPD and 31 healthy controls (Additional file 6: Table S1). Considering the challenge of quantifying dozens of protein candidates in parallel, we employed a median-throughput mass spectrometry-based approach as the Parallel Reaction Monitoring (PRM) for analysis of 176 tryptic peptides. Eventually, this targeted proteomic analysis approach detected 16 protein candidates with robust signal across the validation set. The trends of the marked proteins in COPD samples corroborated results from the discovery study (Fig. 2F). To sum to, the final 5 significantly dysregulated proteins were selected after validating via targeted proteomics, including ORM1, HP, HBB, VCL, and CDH5.

Proteomic subtypes of COPD and their association with clinical outcomes

Consensus clustering based on the 107 most variable proteins in COPD identified three proteomic subtypes (each disease normalized by health and SD > 0.5) (Fig. 3A). They were designated as subtype I (n = 19), subtype II (COPD-BE, n = 9), and subtype III (COPD-MD, n = 12). The resulting heatmap revealed that the DEPs were significantly enriched in metabolic pathways and complement and coagulation cascades in subtype I. Moreover, 5 highly expressed proteins, including B4GAT1, GNPTG, ADAMTSL4, CFP, and EXTL2 were identified. We found that the subtype II was enriched in metabolic pathways, biosynthesis of antibiotics, carbon metabolism, biosynthesis of amino acids, and glycolysis/gluconeogenesis, and the involved proteins included SOD1, PRDX2, CAT, PRDX6, HBB, GSTO1, and HBA1. For subtype III group, the complement and coagulation cascades were significantly enriched, and the following proteins were enriched: HP, LBP, SERPINA (1, 3), SAA1, CRP, ORM1, ORM2, and CRP. GO enrichment analysis was performed to annotate the putative functional implications of the grouped DEPs (Fig. 3B–D, F).
A tridimensional plot via PCA showed the configuration of indexes on COPD and COPD with co-morbidities in Fig. 3E. Plots of individual component scores for the first principal component (PC1) versus the second principal component (PC2) versus the third principal component (PC3) were provided. PC1, PC2, and PC3 showed clear separation of COPD from COPD subtypes. Combinations of PC1, PC2, and PC3 could explain 58.4% proportion of the whole variances. Based on the selected proteins panel, as indicated in Fig. 3G, ROC analysis of PCA and the combination of RRM1 + SUPV3L1 + KRT78 was calculated, and results showed that the auROC was 0.95 and 0.96, respectively. There was no significant difference between PCA analysis and the combination of RRM1 + SUPV3L1 + KRT78 (P > 0.05). In addition, basophil count showed the ability to distinguish COPD from COPD-BE or COPD-MD, while white blood cells and neutrophil ratio was able to distinguish COPD from COPD-BE, as well COPD-BE from COPD-MD (Fig. 3H).
In sum, COPD were subtyped into three based on their corresponding clinical outcomes. We also identified that both PCA analysis and the combination of RRM1 + SUPV3L1 + KRT78 could effectively differentiate COPD and COPD with co-morbidities.

Metabolomic profiles and functional alterations associated with COPD

A library of known metabolite standards (APPLIED PROTEINS TECHNOLOGY Co. Ltd) was employed to identify 210 differentially expressed metabolites (DEMs) in COPD compared to healthy controls. In addition, quality control analyses were carried out based on correlation distributions for total and separately metabolites (or by group). The EBAM plots, normalization, PLS-DA, and t test were conducted (Additional file 3: Fig. S3A, B). Results indicated that PLS-DA produced a model that could separate positive and negative metabolites. Heatmaps depicting clustering of total and selected metabolites in positive and negative modes, respectively, are shown in Fig. 4A, B. Notably, 44 differentially expressed metabolites between the two groups were identified, among which 15 and 29 were positive and negative metabolites, respectively. The functions of the selected metabolites were displayed on VIP and volcano plots, and these metabolites were palmitoylethanolamide, trans-Dehydroandrosterone, decanoyl-l-carnitine, betaine, pseudouridine, camphor, 1-stearoyl-2-hydroxy-sn-glycero, hypoxanthine, theophylline, l-isoleucine pregnenolone, androsterone sulfate, azelaic acid, sunitinib, bisindolymalemide1 (Fig. 4C, D). By using the complementary approach, the weighted gene co-expression network analysis (WGCNA), we identified several co-expression modules (Additional file 5: Fig. S5). The Betaine in MEcyan module was found to be significantly associated with COPD. Summary, 8 positive- and 6 negative-metabolites were selected by metabolomic analysis.

Integrated analyses of proteomics and metabolomics data

Correlation analysis

After appropriate sample quality control (QC) and normalization procedures, we performed PCA on the proteomics and metabolomics data. All datasets effectively distinguished COPD from healthy controls, with the best separation observed with the combined proteomics and metabolomics analysis (Fig. 5E). We found that a considerable number of proteins and metabolites were both involved in mineral absorption, proximal tubule bicarbonate reclamation, inflammatory mediator regulation, lysosome, neuroactive ligand-receptor interaction, cAMP signaling pathway, biosynthesis of amino acids, purine metabolism, fructose and mannose metabolism, glycolysis/gluconeogenesis Fig. 5C. However, P value of 0.05 as a cutoff, the significant enrichment pathways were enriched including both proteomics and metabolomics data in Fig. 5A, B. Heatmap analyses of the differentially expressed proteins and metabolites identified relatively strong or weak proteins-metabolites correlations. Proteins or metabolites with strong or weak correlations were detailed in Fig. 5F. The final differential proteins (DEPs) or DEMs were selected as the target proteins or metabolites. To analyze the interactions between them, the network between DEPs and DEMs was analyzed by Cytoscape, and the results detailed in Fig. 5G. In sum, enrichment analyses of DEPs and DEMs were performed to investigate the potential correlations between them, and the results showed that there were strong or weak correlations between these proteins or metabolites.

Establishment of diagnostic panels for COPD (diagnostic efficacy of single biomarkers)

Before the biomarkers were integrated, the profile of each biomarker was first analyzed separately (Fig. 5Ha, Hb). Subsequently, the ROC models were applied to calculate the auROC, specificity, and sensitivity of single biomarkers. The calculations were performed using the following formula: %sensitivity = [true-positive/(true-positive + false-negative)] * 100; %specificity = [true-negative/(true-negative + false-positive)] * 100. Thereafter, 7 positive and 7 negative metabolites, alongside 6 proteins that had shown significant changes in COPD patients (Pmax = 0.029) were individually subjected to ROC analysis, to evaluate their sensitivity and specificity and help discriminate COPD from healthy controls. As showed in Additional file 7: Table S2, results indicated that palmitoylethanolamide, which was used as a positive metabolite had a maximal auROC of 78.0%, with sensitivity and specificity of 68.0 and 72%, respectively. On the other hand, 1-Stearoyl-sn-glycerol (used as a negative metabolite) had an auROC of 78.0% and a sensitivity of 71.0% against controls. For the proteomics data, CDH5 recorded a maximum auROC of 85.0%, with a sensitivity and specificity of 80.0 and 78%, respectively. Results from blood routine tests showed that MPV had an auROC of 64.0%, with a sensitivity and specificity of 59.0 and 53.0% respectively, while monocytes recorded an auROC of 68.0%, with a sensitivity of 62.0%, and specificity of 63.0%. In sum, diagnostic efficacy of single biomarker was established based on metabolites, proteins, and blood routine test. The result indicated that palmitoylethanolamide, 1-Stearoyl-sn-glycerol, and CDH5 had the highest auROC values for positive metabolites, negative metabolites, and proteins, respectively.

Diagnostic capability of combined biomarkers

The data shown in Additional file 8: Table S3 and Fig. 5H indicate that analysis of predictive capability of a combination of 4 positive metabolites, namely palmitoylethanolamide, trans-dehydroandrosterone, decanoyl-l-carnitine, and betaine, obtained an auROC of 91.0%, with a sensitivity and specificity of 83.0 and 85.0%, respectively. When 4 negative metabolites (theophylline, l-isoleucine, 1-stearoyl-sn-glycerol, and hypoxanthine) were combined, an auROC of 95.9%, was obtained with a sensitivity of 90.0%, and specificity of 90.0%. On the other hand, combining scores from 3 positive metabolites (palmitoylethanolamide, decanoyl-l-carnitine, and betaine) with those from 2 negative ones (theophylline and hypoxanthine) resulted in ROC curve with an auROC of 97.0%, a sensitivity of 88.0%, and specificity of 93.0%. The same model was used to construct a logistic model using the 5 markers, dubbed diagnostic P5, and observed differential abundance in predicting serious COPD as follows:
$${\text{Y}}_{{({\text{COPD}} = 1|{\text{control}} = 0)}} = - 14.645 + \left( {0.41*{\text{palmitoylethanolamide}} + 1.41*{\text{decanoyl-L}}\;{\text{carnitine}} - 4.83*{\text{betaine}} + 0.15*{\text{theophylline}} + 1.17*{\text{hypoxanthine}}} \right)/10000.$$
Using this P5 score, advanced COPD participant can be distinguished predicted with high sensitivity and specificity, and the auROC reached 0.97 in our data set (Fig. 5Hc).
Combining scores from all proteins resulted in an auROC of 93.6%, with a sensitivity and specificity of 88.0 and 90.0%, respectively (Fig. 5Hd). The 3-protein (ORM1, CDH5, and PRDX2) based logistic model generated a dichotomous score, dubbed diagnostic P3, which allowed classification of each participant. The relationship between the probability score of a participant being positively diagnosed with advanced COPD and the log2 intensity value of each protein marker was defined as follows:
$${\text{Y}}_{{({\text{COPD}} = 1|{\text{control}} = 0)}} = - 10.323 + 2.354*{\text{ORM}}1 + 6.834*{\text{CDH}}5 + 1.694*{\text{PRDX}}2.$$
Combining scores from 3 metabolites and that of 1 protein resulted in a high auROC value of 98.0%, with a sensitivity of 94.0%, and specificity of 95.0% (Additional file 9: Table S4). The final logistic model, dubbed P4, comprised palmitoylethanolamide, theophylline, hypoxanthine, and CDH5 (all auROCmin > 0.724), and was expressed as follows:
$${\text{Y}}_{{({\text{COPD}} = 1|{\text{control}} = 0)}} = - 17.934 + \left( {0.46*{\text{palmitoylethanolamide}} + 0.13*{\text{theophylline}} + 0.77*{\text{hypoxanthine}}} \right)/10000 + 8.340*{\text{CDH}}5.$$
The scores from the P4 model had significantly higher power than scores from other models in predicting advanced COPD. The sensitivity, specificity, and auROC of P4 for COPD prediction were greatest (Fig. 5He). The highest Youden index (0.835), which indicates the model’s ability to correctly diagnose true serious COPD patients, was achieved at the cut-point. Taken together, results from the logistic model indicated that a combination of palmitoylethanolamide, theophylline, hypoxanthine, and CDH5 was the best signature of serum biomarkers for predicting advanced COPD.

Validation study of the final predicted model

The final predictors were further verified on mild-to-moderate COPD patients and healthy controls. For CDH5, it was found that its expression was not significantly different between COPD and controls (Fig. 5D). The clinical and demographic characteristics of participants are presented in Table 4. A total of 30 patients with COPD and 30 healthy controls were enrolled. Of note, there were statistically significant differences between COPD patients and controls in terms of pack_years, age, heart disease, and pulmonary function (P < 0.05), but no significant difference was found between the two groups in terms of sex, smoking, BMI, respiratory symptoms, chronic respiratory diseases, poison exposure, room ventilation, cook, other comorbidities, and family history of cancers (P > 0.05). Among the metabolites, theophylline was not significantly different between the two groups, but hypoxanthine showed significant differences in the validation cohort (data missing for palmitoylethanolamide) (Fig. 5D). The detailed clinical and demographic characteristics for participants was described in the previous study [36].
Table 4
Characteristics of validation cohorts on mild-to-moderate COPD for CDH5
 
Controls
COPDb
Pa
N
30
30
Male, %
96.67
83.33
0.20
Smoking, %
93.33
76.67
0.15
Pack_years, median (IQR)
25 (14, 40)
40 (20, 50)
0.01
Age (years), mean (SD)
59.97 ± 4.66
65.0 ± 7.00
< 0.01
BMI (kg/m2), mean (SD)
23.97 ± 3.86
22.36 ± 2.59
0.06
Cough without having a cold
7 (23.33)
14 (46.67)
0.06
Phlegm without having a cold
10 (33.33)
13 (43.33)
0.43
Chronic respiratory diseases
3 (10.00)
9 (30.00)
0.05
Poison exposure
14 (46.67)
14 (46.67)
1.00
Good room ventilation, %
19 (63.33)
19 (63.33)
1.00
Offen cook, %
10 (33.33)
15 (50.00)
1.00
Comorbidity, %
 Hypertension
9 (30.00)
8 (26.67)
0.77
 Diabetes
2 (6.67)
2 (6.67)
1.00
 Heart disease
0
5 (16.67)
0.02
 Stroke
2 (6.67)
1 (3.33)
0.55
Family history of cancer, %
8 (26.67)
11 (36.67)
0.41
Pulmonary function, mean (SD)
 FEV1_%Pred
100.51 ± 13.12
68.66 ± 6.90
< 0.01
 FEV1/FVC_%Pred
80.25 ± 3.68
62.23 ± 6.53
< 0.01
Bold values indicate significant differences
CRD chronic respiratory disease
aP-values for a two-sided χ2 test or t-test. Data are median (P25–P75), n (%)
bMild-to-moderate COPD, Global Initiative for Obstructive Lung Disease stage 1–2
In sum, theophylline and CDH5 had not significantly different between mild-to-moderate COPD patients and healthy controls.

Discussion

Globally, COPD kills more than 3 million people every year. Although several advances have been achieved in the symptomatic treatment and prevention of acute clinical cases, there are few interventions for ameliorating disease progression or decrease mortality. Therefore, it is important to identify biomarkers that can predict disease occurrence or aid in diagnose of advanced COPD. This will facilitate early intervention and prevent progression. In this study, we found that a combination of theophylline, palmitoylethanolamide, hypoxanthine, and CDH5 provides a high diagnostic accuracy. Proteomics facilitates the differentiation of COPD from COPD with co-morbidities. We also found that basophil count could effectively distinguish COPD from COPD-BE or COPD-MD. Moreover, hypoxanthine was still significantly different between mild-to-moderate COPD and controls.
In clinical practice, plasma or serum is the most widely used specimen for biomarker discovery because proteins/metabolites in the circulatory system likely reflect disease pathophysiology. Our dataset can be used to identify potential predictive biomarkers of advanced COPD. Theophylline and the other three methylxanthine derivatives (aminophylline, etophylline, and caffeine), are the first four compounds to have been approved for use in clinical practice [12]. Among them, as bronchodilators, theophylline is the most effective and is widely used for the treatment of asthma and COPD. Evidences showed that corticosteroids and theophylline, both in low doses, have synergistic and clinically useful anti-inflammatory effects in COPD [26]. The underlying molecular mechanisms suggest that this happens through theophylline increasing the activity of the nuclear enzyme histone deacetylase-2 (HDAC2), which is decreased in COPD, therefore preventing the anti-inflammatory effect of corticosteroids [1]. Scientists have identified that low-dose theophylline, especially below those which lead to bronchodilatation, can reverse corticosteroid insensitivity in COPD [9, 26]. Another study has demonstrated an effect for low-dose theophylline on the forced expiratory volume in one second (FEV1) as well as exacerbations [37]. The metabolic disposition of theophylline in humans was first reported by Brodie et al. [3]. Following a therapeutic dose, only 85% has been accounted for by measurement of known metabolites, and unchanged drug excreted in urine. Therefore, about 10% of theophylline administered to man appears in urine in an unchanged form. This would be one of the main sources of theophylline in the body, and the main reason for deviations between patients and controls. It may also explain why there were no differences in theophylline between mild-to-moderate COPD and control in the present study. In addition, as one of the methylxanthines, theophylline is also a natural and synthetic compound found in tea, most of which is metabolized by some types of bacteria and fungi, some of which exist in blood circulation in the human body [35]. However, the information about tea drinking was lacking in this study. This need to be investigated in the future.
Hypoxanthine is a product of ATP degradation, and its conversion to uric acid is facilitated by the enzyme xanthine oxidase, generating free oxygen radicals [5]. It is a metabolite that is involved in purine biosynthesis and nucleotide metabolism, and often serves as a biomarker. For instance, hypoxanthine is a potential marker for oxidative stress in cystic fibrosis [31]; a combination of eight metabolites including uric acid, stearic acid, threitol, acetylgalactosamine, heptadecanoic acid, aspartic acid, xanthosine and hypoxanthine were found to accurately diagnose asthma while discriminating between healthy control and asthma subgroups. In preschool children with cystic fibrosis, hypoxanthine concentrations were found to be elevated in BALF from lobes of the lung containing localized bronchiectasis and were correlated with neutrophil counts and important clinical outcomes [7, 32]. Elevated hypoxanthine concentrations in various body fluids are as a result of vital tissue hypoxia. For mild-to-moderate COPD, higher level of hypoxanthine has also been demonstrated, and this might explain that tissue hypoxia exists in COPD at early time. Suppressed serum hypoxanthine levels have been reported in lung cancer [14] and cystic fibrosis lung disease [17]. Increased conversion to uric acid during exacerbation, may result in a reduction in the concentration of hypoxanthine, generating superoxide and hydroxyl radicals in which cause cellular damage. However, this phenomenon needs to be investigated in COPD.
Vascular endothelial cadherin 5 (CDH5), an endothelial specific cell–cell adhesion molecule, plays important roles in the formation, maturation, and remodeling of the vascular wall [10]. RAB26 is a newly identified small GTPase involved in regulation of endothelial cell (EC) permeability [6]. It confers protective effects on EC permeability, which is in part dependent on autophagic targeting of active SRC, and the resultant CDH5 dephosphorylation maintains adherent junction stabilization. During inflammation, CDH5 phosphorylation at tyrosine residues induces opening of endothelial adherent junctions [30]. Post-translational modifications of CDH5 at tyrosine residues are involved in vascular permeability and leukocyte transmigration. Moreover, cell surface CDH5 phosphorylation is directly linked to EC barrier integrity. These results suggest that any change in CDH5 will impact endothelial barrier functions at multiple levels and CDH5 inhibition may lead to a marked increase in permeability [11, 27]. Enhanced permeability is an early step in the angiogenic process, enabling endothelial migration out of the primary vessel in order to format the tumor neovasculature in the next [18]. Moreover, induction of CDH5 during epithelial mesenchymal transformation accentuates breast cancer progression via TGF-β signaling, indicating that in certain tumor cells, CDH5 can induce cellular responses that counteract its inhibitory role in cell–cell contact growth in EC [16]. Therefore, CDH5 has two functions in angiogenesis and cancer progression. Smoking, a key factor that regulates COPD development, causes hypoxia, which is an important driver of angiogenesis which participates in the pathogenesis of COPD.
COPD is a heterogeneous condition that presents the opportunity for precision therapy based on more precise disease subtypes. Subtype directed therapies, such as inhaled corticosteroids for patients with frequent exacerbations, have had only moderate success. This is likely due to imprecise phenotype categorization, the limited number of drugs for treating COPD, and the generally modest effects of most of these drugs. It is, therefore, crucial to provide precise therapies for patients with specific COPD subtypes based on specific biomarkers. Since comorbidities have a tremendous impact on the prognosis and severity of COPD, the 2015 American Thoracic Society/European Respiratory Society (ATS/ERS) Research Statement on COPD urgently called for studies to elucidate on the pathological mechanisms involved in the association between COPD and its comorbidities. Since comorbidities have influence the clinical outcomes of COPD, identification of the mechanisms linking COPD to its comorbidities is key to developing effective therapies. Presently, it has not been established whether BE or MD is an independent co-existing condition or a direct consequence of progressive lung pathology in COPD patients. In this study, we developed a pipeline for proteomic dominated subtyping of COPD, which complements subtyping approaches based on clinical or imaging data [23, 29], as well clustering by omics in Chinese. In particular, based on proteomics results, COPD patients were grouped into three clusters according to prominent molecular features, including simplex COPD, COPD-BE, and COPD-MD. To further differentiate the disease subtypes, we identified that COPD-MD is highly involved in complement and coagulation cascades processes, and was enriched with various proteins, including HP, LBP, SERPINA1, SERPINA3, SAA1, ORM1, ORM2, and CRP. COPD-BE participates in complement and coagulation cascades processes, and is enriched with various proteins, including metabolic pathways, biosynthesis of antibiotics, carbon metabolism, biosynthesis of amino acids, and glycolysis/gluconeogenesis. Moreover, SOD1, PRDX2, CAT, PRDX6, HBB, GSTO1, and HBA1 were highly expressed in COPD-MD. Since advanced COPD possess unique metabolic pathways and typically express protein isoforms that may have special functions, proteomic approaches for studies of metabolic pathways are especially important.
This study has some limitations. First, no follow-up investigation of the same participants was carried out. Further multi-center and longitudinal studies are need to the prediction performance of the identified biomarkers in advanced COPD. Second, this was a retrospective study, therefore, laboratory tests might be underestimated in medical records, making it difficult to explore their effects on outcomes. Moreover, information on medication, disease control status, and disease phenotypes before admission were incomplete. The impact of these factors on disease expression should be further evaluated. Third, the study population was relatively small. Thus, large prospective studies should be performed to validate the present findings. Finally, although traditional methods, such as logistic regression used in this study, are often used to establish prediction models, it has been suggested that Artificial Intelligence (AI) based machine learning (ML) approaches may be more accurate than traditional logistic regression. This is because AI-based ML can overcome many of the disadvantages of conventional statistical approaches used for analyses of high-volume next generation sequencing data. For instance, ML does not require full details of sequencing measurements and can extract features from sequences [28]. Therefore, ML approaches should be considered in further studies.

Conclusion

An integrated array of proteins and metabolites, including theophylline, palmitoylethanolamide, hypoxanthine, and CDH5 showed the potential to diagnose advanced COPD patients with a high accuracy. Based on proteomics, advanced COPD patients were assigned into 3 subgroups. In particular, COPD-MD was found to be highly involved in complement and coagulation cascades processes, and HP, LBP, SERPINA1, SERPINA3, SAA1, ORM1, ORM2, and CRP were highly enriched.

Acknowledgements

We sincerely thank Professor Juntuo Zhou (Beihang University, Beijing, China) for his invaluable assistance in the validation of final metabolites on mild-to-moderate COPD.

Declarations

Each patient voluntarily provided a written informed consent prior to inclusion in the study and data collection. The study was approved by the Institutional Review Board of Guangzhou Medical University, Ethics Committee of the First Affiliated Hospital (approval number: GZMC 2009-08-1336), and was conducted in accordance with the principles of the Declaration of Helsinki.
Not applicable.

Competing interests

The authors declare that there is no conflict of interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Barnes PJ. Identifying molecular targets for new drug development for chronic obstructive pulmonary disease: what does the future hold? Semin Respir Crit Care Med. 2015;36(4):508–22.CrossRefPubMed Barnes PJ. Identifying molecular targets for new drug development for chronic obstructive pulmonary disease: what does the future hold? Semin Respir Crit Care Med. 2015;36(4):508–22.CrossRefPubMed
2.
Zurück zum Zitat Bowler RP, Jacobson S, Cruickshank C, Hughes GJ, Siska C, Ory DS, Petrache I, Schaffer JE, Reisdorph N, Kechris K. Plasma sphingolipids associated with chronic obstructive pulmonary disease phenotypes. Am J Respir Crit Care Med. 2015;191(3):275–84.CrossRefPubMedPubMedCentral Bowler RP, Jacobson S, Cruickshank C, Hughes GJ, Siska C, Ory DS, Petrache I, Schaffer JE, Reisdorph N, Kechris K. Plasma sphingolipids associated with chronic obstructive pulmonary disease phenotypes. Am J Respir Crit Care Med. 2015;191(3):275–84.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Brodie BB, Axelrod J, Reichenthal J. Metabolism of theophylline (1,3-dimethylxanthine) in man. J Biol Chem. 1952;194(1):215–22.CrossRefPubMed Brodie BB, Axelrod J, Reichenthal J. Metabolism of theophylline (1,3-dimethylxanthine) in man. J Biol Chem. 1952;194(1):215–22.CrossRefPubMed
4.
Zurück zum Zitat Chen Q, Deeb RS, Ma Y, Staudt MR, Crystal RG, Gross SS. Serum metabolite biomarkers discriminate healthy smokers from COPD smokers. PLoS ONE. 2015;10(12): e0143937.CrossRefPubMedPubMedCentral Chen Q, Deeb RS, Ma Y, Staudt MR, Crystal RG, Gross SS. Serum metabolite biomarkers discriminate healthy smokers from COPD smokers. PLoS ONE. 2015;10(12): e0143937.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Chevion S, Moran DS, Heled Y, Shani Y, Regev G, Abbou B, Berenshtein E, Stadtman ER, Epstein Y. Plasma antioxidant status and cell injury after severe physical exercise. Proc Natl Acad Sci USA. 2003;100(9):5119–23.CrossRefPubMedPubMedCentral Chevion S, Moran DS, Heled Y, Shani Y, Regev G, Abbou B, Berenshtein E, Stadtman ER, Epstein Y. Plasma antioxidant status and cell injury after severe physical exercise. Proc Natl Acad Sci USA. 2003;100(9):5119–23.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Dong W, He B, Qian H, Liu Q, Wang D, Li J, Wei Z, Wang Z, Xu Z, Wu G, Qian G, Wang G. RAB26-dependent autophagy protects adherens junctional integrity in acute lung injury. Autophagy. 2018;14(10):1677–92.CrossRefPubMedPubMedCentral Dong W, He B, Qian H, Liu Q, Wang D, Li J, Wei Z, Wang Z, Xu Z, Wu G, Qian G, Wang G. RAB26-dependent autophagy protects adherens junctional integrity in acute lung injury. Autophagy. 2018;14(10):1677–92.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Esther CR, Coakley RD, Henderson AG, Zhou Y-H, Wright FA, Boucher RC. Metabolomic evaluation of neutrophilic airway inflammation in cystic fibrosis. Chest. 2015;148(2):507–15.CrossRefPubMedPubMedCentral Esther CR, Coakley RD, Henderson AG, Zhou Y-H, Wright FA, Boucher RC. Metabolomic evaluation of neutrophilic airway inflammation in cystic fibrosis. Chest. 2015;148(2):507–15.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Ford PA, Durham AL, Russell RE, Gordon F, Adcock IM, Barnes PJ. Treatment effects of low-dose theophylline combined with an inhaled corticosteroid in COPD. Chest. 2010;137(6):1338–44.CrossRefPubMed Ford PA, Durham AL, Russell RE, Gordon F, Adcock IM, Barnes PJ. Treatment effects of low-dose theophylline combined with an inhaled corticosteroid in COPD. Chest. 2010;137(6):1338–44.CrossRefPubMed
12.
Zurück zum Zitat Jilani TN, Preuss CV, Sharma S. Theophylline. BTI - StatPearls. Jilani TN, Preuss CV, Sharma S. Theophylline. BTI - StatPearls.
13.
Zurück zum Zitat Krzanowski WJ, Kline P. Cross-validation for choosing the number of important components in principal component analysis. Multivar Behav Res. 1995;30(2):149–65.CrossRef Krzanowski WJ, Kline P. Cross-validation for choosing the number of important components in principal component analysis. Multivar Behav Res. 1995;30(2):149–65.CrossRef
14.
Zurück zum Zitat Kumar N, Shahjaman M, Mollah MNH, Islam SMS, Hoque MA. Serum and plasma metabolomic biomarkers for lung cancer. Bioinformation. 2017;13(6):202–8.CrossRefPubMedPubMedCentral Kumar N, Shahjaman M, Mollah MNH, Islam SMS, Hoque MA. Serum and plasma metabolomic biomarkers for lung cancer. Bioinformation. 2017;13(6):202–8.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Labaki WW, Rosenberg SR. Chronic obstructive pulmonary disease. Ann Internal Med. 2020;173(3):ITC17–32.CrossRef Labaki WW, Rosenberg SR. Chronic obstructive pulmonary disease. Ann Internal Med. 2020;173(3):ITC17–32.CrossRef
16.
Zurück zum Zitat Labelle M, Schnittler HJ, Aust DE, Friedrich K, Baretton G, Vestweber D, Breier G. Vascular endothelial cadherin promotes breast cancer progression via transforming growth factor beta signaling. Can Res. 2008;68(5):1388–97.CrossRef Labelle M, Schnittler HJ, Aust DE, Friedrich K, Baretton G, Vestweber D, Breier G. Vascular endothelial cadherin promotes breast cancer progression via transforming growth factor beta signaling. Can Res. 2008;68(5):1388–97.CrossRef
17.
Zurück zum Zitat Laguna TA, Reilly CS, Williams CB, Welchlin C, Wendt CH. Metabolomics analysis identifies novel plasma biomarkers of cystic fibrosis pulmonary exacerbation. Pediatr Pulmonol. 2015;50(9):869–77.CrossRefPubMedPubMedCentral Laguna TA, Reilly CS, Williams CB, Welchlin C, Wendt CH. Metabolomics analysis identifies novel plasma biomarkers of cystic fibrosis pulmonary exacerbation. Pediatr Pulmonol. 2015;50(9):869–77.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Le Guelte A, Dwyer J, Gavard J. Jumping the barrier: VE-cadherin, VEGF and other angiogenic modifiers in cancer. Biol Cell. 2011;103(12):593–605.CrossRefPubMed Le Guelte A, Dwyer J, Gavard J. Jumping the barrier: VE-cadherin, VEGF and other angiogenic modifiers in cancer. Biol Cell. 2011;103(12):593–605.CrossRefPubMed
19.
Zurück zum Zitat Lu W, Zheng Z, Chen X, Tan H, Wang J, Zhang Z, Zheng J, Chen R, Zhang C, Xu X, Chen Y, Yang Q, Xiong M, Guo M, Zhou Q, Tang C, Wang Y, Ye J, Li D, Shu J, Tan S, Xu C, Wang Y, Lai N, Yang K, Lu J, Ran P, Zhong N. Study design and interim outcomes of Guangzhou institute of respiratory disease COPD biobank. COPD. 2016;13(2):203–13.CrossRefPubMed Lu W, Zheng Z, Chen X, Tan H, Wang J, Zhang Z, Zheng J, Chen R, Zhang C, Xu X, Chen Y, Yang Q, Xiong M, Guo M, Zhou Q, Tang C, Wang Y, Ye J, Li D, Shu J, Tan S, Xu C, Wang Y, Lai N, Yang K, Lu J, Ran P, Zhong N. Study design and interim outcomes of Guangzhou institute of respiratory disease COPD biobank. COPD. 2016;13(2):203–13.CrossRefPubMed
20.
21.
Zurück zum Zitat Ran N, Pang Z, Gu Y, Pan H, Zuo X, Guan X, Yuan Y, Wang Z, Guo Y, Cui Z, Wang F. An updated overview of metabolomic profile changes in chronic obstructive pulmonary disease. Metabolites. 2019;9(6):111.CrossRefPubMedPubMedCentral Ran N, Pang Z, Gu Y, Pan H, Zuo X, Guan X, Yuan Y, Wang Z, Guo Y, Cui Z, Wang F. An updated overview of metabolomic profile changes in chronic obstructive pulmonary disease. Metabolites. 2019;9(6):111.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Regan EA, Hersh CP, Castaldi PJ, DeMeo DL, Silverman EK, Crapo JD, Bowler RP. Omics and the search for blood biomarkers in chronic obstructive pulmonary disease. insights from COPDGene. Am J Respir Cell Mol Biol. 2019;61(2):143–9.CrossRefPubMedPubMedCentral Regan EA, Hersh CP, Castaldi PJ, DeMeo DL, Silverman EK, Crapo JD, Bowler RP. Omics and the search for blood biomarkers in chronic obstructive pulmonary disease. insights from COPDGene. Am J Respir Cell Mol Biol. 2019;61(2):143–9.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Criner GJ, Frith P, Halpin DMG, Han M, López Varela MV, Martinez F, Montes de Oca M, Papi A, Pavord ID, Roche N, Sin DD, Stockley R, Vestbo J, Wedzicha JA, Vogelmeier C. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease: the GOLD science committee report 2019. Eur Respir J. 2019;53(5):1900164.CrossRefPubMed Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, Criner GJ, Frith P, Halpin DMG, Han M, López Varela MV, Martinez F, Montes de Oca M, Papi A, Pavord ID, Roche N, Sin DD, Stockley R, Vestbo J, Wedzicha JA, Vogelmeier C. Global strategy for the diagnosis, management, and prevention of chronic obstructive lung disease: the GOLD science committee report 2019. Eur Respir J. 2019;53(5):1900164.CrossRefPubMed
26.
Zurück zum Zitat Spears M, Donnelly I, Jolly L, Brannigan M, Ito K, McSharry C, Lafferty J, Chaudhuri R, Braganza G, Adcock IM, Barnes PJ, Wood S, Thomson NC. Effect of low-dose theophylline plus beclometasone on lung function in smokers with asthma: a pilot study. Eur Respir J. 2009;33(5):1010–7.CrossRefPubMed Spears M, Donnelly I, Jolly L, Brannigan M, Ito K, McSharry C, Lafferty J, Chaudhuri R, Braganza G, Adcock IM, Barnes PJ, Wood S, Thomson NC. Effect of low-dose theophylline plus beclometasone on lung function in smokers with asthma: a pilot study. Eur Respir J. 2009;33(5):1010–7.CrossRefPubMed
27.
Zurück zum Zitat Taddei A, Giampietro C, Conti A, Orsenigo F, Breviario F, Pirazzoli V, Potente M, Daly C, Dimmeler S, Dejana E. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat Cell Biol. 2008;10(8):923–34.CrossRefPubMed Taddei A, Giampietro C, Conti A, Orsenigo F, Breviario F, Pirazzoli V, Potente M, Daly C, Dimmeler S, Dejana E. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat Cell Biol. 2008;10(8):923–34.CrossRefPubMed
28.
Zurück zum Zitat Teng SY, Yew GY, Sukačová K, Show PL, Máša V, Chang J-S. Microalgae with artificial intelligence: a digitalized perspective on genetics, systems and products. Biotechnol Adv. 2020;44: 107631.CrossRefPubMed Teng SY, Yew GY, Sukačová K, Show PL, Máša V, Chang J-S. Microalgae with artificial intelligence: a digitalized perspective on genetics, systems and products. Biotechnol Adv. 2020;44: 107631.CrossRefPubMed
29.
Zurück zum Zitat Wan ES, Castaldi PJ, Cho MH, Hokanson JE, Regan EA, Make BJ, Beaty TH, Han MK, Curtis JL, Curran-Everett D, Lynch DA, DeMeo DL, Crapo JD, Silverman EK. Epidemiology, genetics, and subtyping of preserved ratio impaired spirometry (PRISm) in COPDGene. Respir Res. 2014;15:89.CrossRefPubMedPubMedCentral Wan ES, Castaldi PJ, Cho MH, Hokanson JE, Regan EA, Make BJ, Beaty TH, Han MK, Curtis JL, Curran-Everett D, Lynch DA, DeMeo DL, Crapo JD, Silverman EK. Epidemiology, genetics, and subtyping of preserved ratio impaired spirometry (PRISm) in COPDGene. Respir Res. 2014;15:89.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Wessel F, Winderlich M, Holm M, Frye M, Rivera-Galdos R, Vockel M, Linnepe R, Ipe U, Stadtmann A, Zarbock A, Nottebaum AF, Vestweber D. Leukocyte extravasation and vascular permeability are each controlled in vivo by different tyrosine residues of VE-cadherin. Nat Immunol. 2014;15(3):223–30.CrossRefPubMed Wessel F, Winderlich M, Holm M, Frye M, Rivera-Galdos R, Vockel M, Linnepe R, Ipe U, Stadtmann A, Zarbock A, Nottebaum AF, Vestweber D. Leukocyte extravasation and vascular permeability are each controlled in vivo by different tyrosine residues of VE-cadherin. Nat Immunol. 2014;15(3):223–30.CrossRefPubMed
31.
Zurück zum Zitat Wetmore DR, Joseloff E, Pilewski J, Lee DP, Lawton KA, Mitchell MW, Milburn MV, Ryals JA, Guo L. Metabolomic profiling reveals biochemical pathways and biomarkers associated with pathogenesis in cystic fibrosis cells. J Biol Chem. 2010;285(40):30516–22.CrossRefPubMedPubMedCentral Wetmore DR, Joseloff E, Pilewski J, Lee DP, Lawton KA, Mitchell MW, Milburn MV, Ryals JA, Guo L. Metabolomic profiling reveals biochemical pathways and biomarkers associated with pathogenesis in cystic fibrosis cells. J Biol Chem. 2010;285(40):30516–22.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Wolak JE, Esther CR, O’Connell TM. Metabolomic analysis of bronchoalveolar lavage fluid from cystic fibrosis patients. Biomarkers. 2009;14(1):55–60.CrossRefPubMed Wolak JE, Esther CR, O’Connell TM. Metabolomic analysis of bronchoalveolar lavage fluid from cystic fibrosis patients. Biomarkers. 2009;14(1):55–60.CrossRefPubMed
33.
Zurück zum Zitat Yonchuk JG, Silverman EK, Bowler RP, Agustí A, Lomas DA, Miller BE, Tal-Singer R, Mayer RJ. Circulating soluble receptor for advanced glycation end products (sRAGE) as a biomarker of emphysema and the RAGE axis in the lung. Am J Respir Crit Care Med. 2015;192(7):785–92.CrossRefPubMed Yonchuk JG, Silverman EK, Bowler RP, Agustí A, Lomas DA, Miller BE, Tal-Singer R, Mayer RJ. Circulating soluble receptor for advanced glycation end products (sRAGE) as a biomarker of emphysema and the RAGE axis in the lung. Am J Respir Crit Care Med. 2015;192(7):785–92.CrossRefPubMed
34.
Zurück zum Zitat Zhang H, Guan R, Zhang Z, Li D, Xu J, Gong Y, Chen X, Lu W. LncRNA Nqo1-AS1 attenuates cigarette smoke-induced oxidative stress by upregulating its natural antisense transcript Nqo1. Front Pharmacol. 2021;12: 729062.CrossRefPubMedPubMedCentral Zhang H, Guan R, Zhang Z, Li D, Xu J, Gong Y, Chen X, Lu W. LncRNA Nqo1-AS1 attenuates cigarette smoke-induced oxidative stress by upregulating its natural antisense transcript Nqo1. Front Pharmacol. 2021;12: 729062.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Zhou B, Ma C, Zheng C, Xia T, Ma B, Liu X. 3-Methylxanthine production through biodegradation of theobromine by Aspergillus sydowii PT-2. BMC Microbiol. 2020;20(1):269.CrossRefPubMedPubMedCentral Zhou B, Ma C, Zheng C, Xia T, Ma B, Liu X. 3-Methylxanthine production through biodegradation of theobromine by Aspergillus sydowii PT-2. BMC Microbiol. 2020;20(1):269.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Zhou J, Li Q, Liu C, Pang R, Yin Y. Plasma metabolomics and lipidomics reveal perturbed metabolites in different disease stages of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2020;15:553–65.CrossRefPubMedPubMedCentral Zhou J, Li Q, Liu C, Pang R, Yin Y. Plasma metabolomics and lipidomics reveal perturbed metabolites in different disease stages of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2020;15:553–65.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Zhou Y, Wang X, Zeng X, Qiu R, Xie J, Liu S, Zheng J, Zhong N, Ran P. Positive benefits of theophylline in a randomized, double-blind, parallel-group, placebo-controlled study of low-dose, slow-release theophylline in the treatment of COPD for 1 year. Respirology. 2006;11(5):603–10.CrossRefPubMed Zhou Y, Wang X, Zeng X, Qiu R, Xie J, Liu S, Zheng J, Zhong N, Ran P. Positive benefits of theophylline in a randomized, double-blind, parallel-group, placebo-controlled study of low-dose, slow-release theophylline in the treatment of COPD for 1 year. Respirology. 2006;11(5):603–10.CrossRefPubMed
Metadaten
Titel
Proteomics and metabolomics profiling reveal panels of circulating diagnostic biomarkers and molecular subtypes in stable COPD
verfasst von
Zili Zhang
Jian Wang
Yuanyuan Li
Fei Liu
Lingdan Chen
Shunping He
Fanjie Lin
Xinguang Wei
Yaowei Fang
Qiongqiong Li
Juntuo Zhou
Wenju Lu
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2023
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/s12931-023-02349-x

Weitere Artikel der Ausgabe 1/2023

Respiratory Research 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Neue S3-Leitlinie zur unkomplizierten Zystitis: Auf Antibiotika verzichten?

15.05.2024 Harnwegsinfektionen Nachrichten

Welche Antibiotika darf man bei unkomplizierter Zystitis verwenden und wovon sollte man die Finger lassen? Welche pflanzlichen Präparate können helfen? Was taugt der zugelassene Impfstoff? Antworten vom Koordinator der frisch überarbeiteten S3-Leitlinie, Prof. Florian Wagenlehner.

Schadet Ärger den Gefäßen?

14.05.2024 Arteriosklerose Nachrichten

In einer Studie aus New York wirkte sich Ärger kurzfristig deutlich negativ auf die Endothelfunktion gesunder Probanden aus. Möglicherweise hat dies Einfluss auf die kardiovaskuläre Gesundheit.

Intervallfasten zur Regeneration des Herzmuskels?

14.05.2024 Herzinfarkt Nachrichten

Die Nahrungsaufnahme auf wenige Stunden am Tag zu beschränken, hat möglicherweise einen günstigen Einfluss auf die Prognose nach akutem ST-Hebungsinfarkt. Darauf deutet eine Studie an der Uniklinik in Halle an der Saale hin.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.