Skip to main content

An integrated metabo-lipidomics profile of induced sputum for the identification of novel biomarkers in the differential diagnosis of asthma and COPD

A Publisher Correction to this article was published on 05 April 2024

This article has been updated

Abstract

Background

Due to their complexity and to the presence of common clinical features, differentiation between asthma and chronic obstructive pulmonary disease (COPD) can be a challenging task, complicated in such cases also by asthma–COPD overlap syndrome. The distinct immune/inflammatory and structural substrates of COPD and asthma are responsible for significant differences in the responses to standard pharmacologic treatments. Therefore, an accurate diagnosis is of central relevance to assure the appropriate therapeutic intervention in order to achieve safe and effective patient care. Induced sputum (IS) accurately mirrors inflammation in the airways, providing a more direct picture of lung cell metabolism in comparison to those specimen that reflect analytes in the systemic circulation.

Methods

An integrated untargeted metabolomics and lipidomics analysis was performed in IS of asthmatic (n = 15) and COPD (n = 22) patients based on Ultra-High-Pressure Liquid Chromatography-Mass Spectrometry (UHPLC-MS) and UHPLC–tandem MS (UHPLC-MS/MS). Partial Least Squares-Discriminant Analysis (PLS-DA) was applied to resulting dataset. The analysis of main enriched metabolic pathways and the association of the preliminary metabolites/lipids pattern identified to clinical parameters of asthma/COPD differentiation were explored. Multivariate ROC analysis was performed in order to determine the discriminatory power and the reliability of the putative biomarkers for diagnosis between COPD and asthma.

Results

PLS-DA indicated a clear separation between COPD and asthmatic patients. Among the 15 selected candidate biomarkers based on Variable Importance in Projection scores, putrescine showed the highest score. A differential IS bio-signature of 22 metabolites and lipids was found, which showed statistically significant variations between asthma and COPD. Of these 22 compounds, 18 were decreased and 4 increased in COPD compared to asthmatic patients. The IS levels of Phosphatidylethanolamine (PE) (34:1), Phosphatidylglycerol (PG) (18:1;18:2) and spermine were significantly higher in asthmatic subjects compared to COPD.

Conclusions

This is the first pilot study to analyse the IS metabolomics/lipidomics signatures relevant in discriminating asthma vs COPD. The role of polyamines, of 6-Hydroxykynurenic acid and of d-rhamnose as well as of other important players related to the alteration of glycerophospholipid, aminoacid/biotin and energy metabolism provided the construction of a diagnostic model that, if validated on a larger prospective cohort, might be used to rapidly and accurately discriminate asthma from COPD.

Graphical Abstract

Background

Asthma and chronic obstructive pulmonary disease (COPD) are the most common lung diseases, with COPD the third leading cause of death worldwide [1]. The global prevalence is approximately 3.6% for asthma and 3.9% for COPD [2]. Currently, differentiation of asthma and COPD is mainly based on clinical and lung function parameters. Although asthma and COPD are two etiological different diseases with different pathophysiology and different patient characteristics, patients can sometimes present similar clinical features of both diseases and invasive diagnostic methods are required to improve diagnostic accuracy [3]. In older subjects, the predicted forced expiratory volume in 1 s (FEV1) falls to 50% in both asthma and COPD patients over 60 years old, therefore it could be difficult to discriminate between COPD and asthma [4]. Sometimes, patients with asthma might develop COPD later in life as well as patients with COPD can show clinical characteristics usually observed in asthma [5]. To further complicate this picture, asthma and COPD can occur together for such patients, and, in clinical practice, the term asthma-COPD overlap (ACO) is used to describe these cases [6]. Therefore, differentiating between these multifactorial pulmonary diseases can be difficult, posing many problems in the clinical setting and management, especially in case of misdiagnosis that may lead to inadequate treatment. Finally, yet importantly, the problem of underdiagnoses in COPD and asthma lead patients to begin therapy very late with increased risk of acute exacerbations and early death [7, 8].

Metabolomics, as well as other MS-based ‘omics, with the combined use of high-throughput technologies and multivariate statistical approaches, represents a valuable tool in the study of biochemical and metabolic processes underlying lung respiratory diseases [9,10,11,12]. In fact, metabolism dysregulation plays an important role in the development of both acute and chronic lung diseases. Additionally, alterations in metabolism may influence not only the severity and the progression of diseases but also patient response to a specific therapy [13]. Asthma and COPD metabolomics made great strides in the past two decades, highlighting main metabolic arrangements and providing an accurate characterization of metabolic pathways associated to asthma and COPD [9, 10]. However, the use of metabolomics as diagnostic tool to be translated into clinical practice, still remains a research gap to be filled.

Recent progress in MS-based integrated metabolomics and lipidomics has opened new horizons in the field of biomarkers discovery and translational medicine by establishing a more inclusive pattern profiling of small molecules, which can better characterize metabolic functions [14, 15]. In particular, biomarkers discovery from integrated metabolomics and lipidomics data will not only help the stratification of different patient cohorts but will also provide a potent tool for comparative analysis with high diagnostic value that could improve patient management [16,17,18].

Up to date, only three untargeted LC-MS studies have explored the metabolomics differences between asthma and COPD based on serum [19] and urine [20, 21], which are two important and non-invasive samples reflecting analytes in the systemic circulation. It would be beneficial, however, to analyze specific biomarkers also at a cellular level in the more proximal site of inflammation. Induced sputum (IS) contains specific information related to inflammatory cells and specific mediators of these two respiratory disorders and closely reflects the pathophysiology of these two diseases; therefore, it is widely recognized as an ideal source of biomarkers [11, 12, 22].

Comparison of the wide arrays of metabolites and lipids abundances between asthma and COPD might therefore unmask specific inflammatory and immune response associated to asthma and COPD, enabling the establishment of diagnostic models to be translated into the differential clinical management of these two diseases.

In the present study, an UHPLC-MS and UHPLC-MS/MS based metabolomics and lipidomics untargeted approach was used to analyze for first time the metabolomics profiles of IS from asthmatic and COPD patients. In particular, we performed a multi-tiered analysis of the metabolomics and lipidomics data obtained from IS comparing these specific biosignatures between the two groups. This integrated approach, by comparing a wide array of metabolites and lipids of a such rich informative fluid of the respiratory airways, not only opens new horizons for the discovery of novel putative biomarkers which can better differentiate asthma from COPD, but also implements the patients’ choice for appropriate and specific therapies.

Materials and methods

Study population

A total of 15 asthmatic patients and 22 COPD patients were recruited in accordance with the principles of the Declaration of Helsinki, and the study was approved by the Institutional Ethical Committee Board of MAGNA GRAECIA UNIVERSITY and MATER DOMINI HOSPITAL (Approval number 219, date of approval:16 November 2016). All patients were enrolled after giving their informed written consent. For all participants an informed consent was provided before enrollment. Asthma and COPD were diagnosed according to the Global Initiative for Asthma guidelines [23] and the Global Initiative for Chronic Obstructive Lung Disease guidelines [24], respectively. Specifically, clinically diagnosed asthmatic subjects showed evidence of airway hyperresponsiveness with 20% drop in FEV1 after a provocative dose of methacholine < 2.5 mg. Subjects treated with oral corticosteroids and those affected by respiratory infection in one month before enrollement were excluded. COPD and asthmatic patients with important comorbidities including heart, liver, kidney pathology, or any other respiratory disease that could possibly influence this study were also excluded. The clinical characteristics of the subjects recruited in this study, including demographic characteristics, smoking status and spirometry function were collected and are summarized in Table 1.

Table 1 Clinical features of the asthmatic and COPD patients enrolled in the study

Induced sputum collection and processing

IS samples were collected and processed in accordance with a previous study by Terracciano et al. [11]. Sputum induction was carried out according to the guidelines of the European Respiratory Society Task Force, with a modification of the standard procedure of induction. Specifically, besides the administration of 4.5% hypertonic saline at 5 min intervals for a maximum of 20 min, participants were instructed to rinse their mouths with water and blow their noses after each inhalation to prevent contamination from saliva and postnasal drip. Only the mucoid components of sputum were selected in order to minimize the influence of salivary contamination. Samples were treated with PBS to which DTT had been added to a final concentration of 5 mM DTT. Then, a protease inhibitor cocktail (P8340 Sigma-Aldrich) was added to the sputum sample (22.5 mL/g of sputum). The tubes, containing the samples, were then placed on a bench roller for 30 min at room temperature and the contents were filtered through a 70 mm mesh and subsequently centrifuged at 400 g for 10 min at 4 °C, to separate the cells from the fluid phase. The supernatants were separated from the pellets and re-centrifuged at 12,000 g, for 10 min at 4 °C, aliquoted and stored at – 80 °C until use. Protein concentration in sputum samples was determined by BCA assay (Pierce BCA Protein Assay Kit, 23225, Thermo Fisher) and data normalization to protein concentration was performed.

Sample preparation for metabolites and lipids extraction

Metabolomics and lipidomics analysis were carried out at the University of Colorado Anschutz Medical Campus Metabolomics Core. First, IS samples were thawed and were treated with a solution of chilled 5:3:2 methanol:acetonitrile:water (v/v/v) at 1:25 ratio, vortexed for 30 min at 4 °C and centrifugated for 10 min at 18,000 rcf at 4 °C, in order to extract metabolites and lipids. After centrifugation, the supernatants were carefully collected and used for metabolomics and lipidomics analysis according to previous studies [18, 25, 26].

UHPLC-MS metabolomics

Metabolomics analyses of IS samples were conducted using a Vanquish UHPLC combined with a high-resolution Q Exactive mass spectrometer (Thermo Fisher, Bremen, Germany) in negative and positive polarity modes in accordance to previous studies [18, 25]. In the case of negative polarity mode, 5 μL of extracts were injected into the UHPLC system and resolved on a Kinetex C18 column (150 × 2.1 mm, 1.7 μm, Phenomenex, Torrance, CA, USA) at 450 μL/min through a 5 min gradient from 0 to 100% organic solvent B (mobile phases: A = 95% water, 5% acetonitrile, 1 mM ammonium acetate; B = 95% acetonitrile, 5% water, 1 mM ammonium acetate). Solvent gradient: 0–0.5 min 0% B, 0.5–1.1 min 0–100% B, 1.1–2.75 min hold at 100% B, 2.75–3 min 100–0% B, 3–5 min hold at 0% B. In the case of positive polarity mode, 5 μL of extracts were injected into the UHPLC system and resolved on a Kinetex C18 column (150 × 2.1 mm, 1.7 μm, Phenomenex, Torrance, CA, USA) at 450 μL/min through a 5 min gradient from 5 to 95% organic solvent B (mobile phases: A = water, 0.1% formic acid; B = acetonitrile, 0.1% formic acid). Solvent gradient: 0–0.5 min 5% B, 0.5–1.1 min 5–95% B, 1.1–2.75 min hold at 95% B, 2.75–3 min 95–5% B, 3–5 min hold at 5% B. The UHPLC system was coupled online with a Q Exactive mass spectrometer, which operated in Full MS mode with a resolution of 70,000 in the 60–900 m/z range. The instrument was set to a 4 kV spray voltage, 15 sheath gas, and 5 auxiliary gas, with negative or positive ion mode (separated runs). Calibration was conducted prior to analysis using the Pierce Positive and Negative Ion Calibration Solutions (Thermo Fisher Scientific). Samples were analyzed in randomized order and technical mixtures as quality controls were used to qualify instrument performance. Quality controls (generated by mixing 10 μL of all samples tested in this study) were injected every 10 runs to warrant technical reproducibility, as demonstrated by determining coefficients of variation (CV, calculated by dividing standard deviations for each metabolite by the mean across all tech mixes) for each reported metabolite for chromatographic retention times and peak areas. A coefficient of variation < 20% was used as threshold. Metabolomics data were collected using Thermo Fisher Scientific Xcalibur software (v.4.1.31.9). LC-MS raw files were converted to mzXML file format using MSconvert v.3.0.20315-7da487568 (ProteoWizard). Metabolite assignments were performed using accurate intact mass (sub-10 ppm), isotopologue distributions, and comparing retention time/spectra to an in-house standard compound library (MSMLS, IROA Technologies, NJ, USA) using MAVEN (Princeton, NJ, USA) (without performing MS/MS methods). The MAVEN software platform provides tools for peak picking (based on peak intensity, quality control consistency, retention time and 13C abundance), feature detection and metabolite assignment using the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database.

UHPLC-MS lipidomics

Lipidomics analyses of IS were performed using a Vanquish UHPLC system coupled online to a high-resolution Q Exactive mass spectrometer (Thermo Fisher, Bremen, Germany) in negative and positive polarity modes in accordance to previous studies [18, 26]. Samples were separated over an ACQUITY HSS T3 column (2.1 × 150 mm, 1.8 µm particle size (Waters, MA, USA). In the case of negative polarity mode, an aqueous phase (A) of 25% acetonitrile and 5 mM ammonium acetate and a mobile phase (B) of 50% isopropanol, 45% acetonitrile, and 5 mM ammonium acetate. Solvent gradient: 0–1 min 25% B and 0.3 mL/min, 1–2 min 25–50% B and 0.3 mL/min, 2–8 min 50–90% B and 0.3 mL/min, 8–10 min 90–99% B and 0.3 mL/min, 10–14 min hold at 99% B and 0.3 mL/min, 14–14.1 min 99–25% B and 0.3 mL/min, 14.1–16.9 min hold at 25% B and 0.4 mL/min, and 16.9 17 min hold at 25% B and resume flow of 0.3 mL/min. In the case of positive polarity mode, an aqueous phase (A) of 25% acetonitrile and 5 mM ammonium acetate and a mobile phase (B) of 90% isopropanol, 10% acetonitrile, and 5 mM ammonium acetate. Solvent gradient: 0–10 min 30–100% B and 0.325 mL/min; 10–12 min 100% B and 0.325 mL/min; 12–12.5 min 100–30% B and 0.4 mL/min; 12.5–14.9 min 30% B and 0.4 mL/min; 14.9 15 min 30% B and 0.325 mL/min. The Q Exactive mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) was operated independently in positive or negative ion mode, scanning in Full MS mode (2 μscans) from 150 to 1500 m/z with a resolution of 70,000, 4 kV spray voltage, 45 sheath gas, and 15 auxiliary gas. For discovery mode untargeted lipidomics, dd-MS2 was performed at 17,500 resolution, AGC target = 1 × 105, maximum IT = 50 ms, and stepped NCE of 25, 35 for positive mode, and 20, 24, and 28 for negative mode. Calibration was conducted prior to analysis using the Pierce Positive and Negative Ion Calibration Solutions (Thermo Fisher Scientific). Samples were analyzed in randomized order and technical mixture or quality controls (generated by mixing 10 μL of all samples tested in this study) were injected every 10 runs and were used to qualify instrument performance (CV < 20%).

Discovery mode analysis was performed with standard workflows using Compound Discoverer 2.1 SP1 (Thermo Fisher Scientific, San Jose, CA, USA). From these analyses, lipid IDs or unique chemical formulae were determined from high-resolution accurate intact mass, isotopic patterns, identification of eventual adducts (e.g., Na+ or K+, etc.) and MS2 fragmentation spectra against the KEGG pathway, HMDB, ChEBI, and ChEMBL databases. Additional untargeted lipidomics analyses were performed with the software LipidSearch 4.0 (Thermo Fisher, Bremen, Germany), which provides lipid identification on the basis of accurate intact mass, isotopic pattern, and fragmentation pattern to determine lipid class and acyl chain composition [26].

Statistical analysis and data integration

After the independent collection of metabolomics and lipidomics data from IS samples, distinct files for metabolomics and lipidomics were generated via MAVEN and Compound Discover, respectively, including a list of all the metabolites and lipids identified in the samples. Subsequently, these files were merged together in a single file and used to perform the subsequent integrated data analysis. Statistical analyses were conducted using a two-sided Student’s t-test on peak intensity and the results were considered statistically significant with p-values < 0.05. Multivariate analyses were performed using MetaboAnalyst (5.0) and encompassed Partial Least Square-Discriminant Analysis (PLS-DA), Orthogonal partial least squares discriminant analysis (OPLS-DA) and hierarchical clustering analyses. PLS-DA and OPLS-DA were multivariate supervised methods used to minimize data dimensionality and identify variables that exhibit the capacity to distinguish between different groups. Hierarchical Clustering Analyses was performed to visualize the top 50 metabolites and lipids through a heatmap, allowing for a clear representation of their patterns. Additionally, box plot analysis was performed by OriginLab® software (version 7.0, OriginLab Corporation, Northampton, MA, USA) and correlation analysis (Spearman) was carried out in JASP open-source statistical software (Version 0.18.1, https://jasp-stats.org). The correlation analysis aimed to provide valuable insights into the clinical relevance and the potential diagnostic significance of the identified metabolites and lipids. Lastly, ROC (Receiver Operating Characteristic) curve analysis was performed using MetaboAnalyst (5.0), to assess the diagnostic performance of biomarkers and evaluate their ability to discriminate between different groups or conditions.

Spearman’s correlation analysis

The relationship between significantly altered metabolites (asthma vs. COPD) and lung function parameters was explored using Spearman’s correlation analysis, conducted using JASP open-source statistical software (Version 0.18.1, https://jasp-stats.org). In order to investigate the extent to which the altered metabolites were linearly related to FEV1 and FEV1/FVC of asthmatic and COPD subjects a Spearman’s correlation analysis was performed.

Results

Clinical characteristics of study participants

The detailed clinical characteristics of the two groups of participants recruited in this study, including demographic characteristics, smoking status and spirometry function, are shown in Table 1.

Differences between patients with COPD and those with asthma were statistically significant in terms of age (p value < 0.001), FEV1% (p value < 0.0001), Forced Vital Capacity percentage predicted (FVC%) and FEV1/FVC (p value < 0.01). Specifically, the COPD patient group exhibited a higher average age and lower FEV1 and FVC% values compared to the asthmatic group. Furthermore, the COPD group comprised 6 current smokers, 15 ex-smokers, and 1 never-smoker. Conversely, among the asthmatic participants, 1 individual was a current smoker, 4 were ex-smokers, and 10 had never smoked. In terms of gender distribution, the majority of COPD patients were male (20 out of 22), while among the 15 asthmatic patients, 5 were male, and 10 were female.

Integrated metabolomics and lipidomics analysis revealed differences between asthmatic and COPD patients

In the present investigation, an untargeted integrated metabolomics and lipidomics analysis by UHPLC-MS was performed, in order to investigate the differences in induced sputum metabolomics and lipidomics patterns between asthmatic and COPD patients.

Metabolomics and lipidomics data from the two analyzed groups of patients, obtained by UHPLC-MS, were compared using multivariate statistical approaches, including PLS-DA and OPLS-DA. These multivariate analyses were used to gain meaningful information from the complex metabolomic datasets, reducing data dimensionality and obtaining a visual differentiation between asthmatic and COPD patients.

As shown in Fig. 1, a well-defined separation of groups was obtained by both the PLS-DA (Fig. 1A) and OPLS-DA (Fig. 1B) models. All COPD IS samples (green dots) were clustered on the right, whereas all asthma induced sputum samples (red dots) were clustered on the left, and no overlap was observed (Fig. 1A, B). This result suggested the presence of noteworthy differences in the IS metabolomic and lipidomic profiles between individuals with COPD and those suffering from asthma.

Fig. 1
figure 1

Metabolic and lipidomic data analysis of asthmatic and COPD patients using multivariate statistical methods. A The score plot generated by Partial Least Squares Discriminant Analysis (PLS-DA) and B the score plot produced by Orthogonal Partial Least Squares Discriminant Analysis (OPLS-DA) visually demonstrates a clear distinction in the metabolome and lipidome profiles between asthmatic and COPD patients. C The VIP score plot, generated using PLS-DA, highlights the top 15 metabolites and lipid compounds that exhibited differences between asthmatic and COPD patients. Colored boxes on the right side of the plot indicate the relative concentration of the corresponding compounds, with blue representing low concentration and red indicating high concentration. These PLS-DA, OPLS-DA, and VIP score plots were generated using the Metaboanalyst software

Figure 1C shows the Variable Importance in the Projection (VIP) plot that was used to identify key metabolites or lipids contributing to the distinction between the two groups. VIP plot was generated from the PLS-DA model and shows the top 15 compounds with a VIP score > 1, which are ranked based on their discriminatory power. Considering that variables with a VIP score > 1 are interpreted as being highly influential, principal compounds considered more important to the strength of segregation were Putrescine, Spermine, Phosphatidylethanolamine (PE) (34:1), Phosphatidylglycerol (PG) (18:1;18:2), AcCa(4:0), Phosphate, PE(36:3), PG(16:1;18:1), PG(18:1;18:1), Lysophosphatidylethanolamine (LPE) (18:1), Ceramide (Cer) (d18:1;16:0), Lysophosphatidylcholine (LPC) (20:1), Sphingomyelin (SM) (d32:1), Phosphatidylcholine (PC) (33:0), PC(O-28:0;21:2).

Variations in metabolite and lipid levels between asthmatic and COPD patients are shown in Fig. 2. In particular, a heatmap was used to display the relative abundances of the top 50 significant metabolites and lipids (Fig. 2A), represented with color intensity, between asthmatic and COPD individuals. In addition, by combining results from fold change (> 1.5 or < 0.67) (COPD/ASTHMA), considered significant cutoff values, and FDR-corrected p-values < 0.05, volcano plot analysis was conducted displaying metabolites and lipid compounds that significantly contributed to the distinction between the two groups (Fig. 2B).

Fig. 2
figure 2

Variations in metabolite and lipid levels between asthmatic and COPD patients. A The heatmap illustrates the relative abundances of the top 50 metabolites and lipids in asthmatic compared to COPD patients. Red indicates an increase in abundance, while blue a decrease. The columns and rows in the heatmap correspond to the experimental samples and the compounds, respectively. B The volcano plot highlights metabolites and lipid compounds that exhibit significant differences between the two groups, based on fold change (COPD/ASTHMA) and p-value. Specifically, blue dots (Fold Change < 0.67; p-value < 0.05) or red dots (Fold Change > 1.5; p-value < 0.05) indicate significantly downregulated or upregulated metabolites, respectively. Gray dots represent compounds that did not show significant differences. Both the heatmap and volcano plot were generated using the Metaboanalyst software

Subsequentially, combining results from VIP plot (VIP score > 1) and Volcano plot analysis (p values < 0.05 by Student t-test and fold change > 1.5 or < 0.67) 22 statistically significant compounds (including metabolites and lipids) were obtained as shown in the Table 2. In particular, 18 compounds showed a decreased concentration and 4 showed an increased concentration in COPD when compared to asthmatics patients. In order to more comprehensively assess the differences between the two groups, a box plot analysis of peak intensities (autoscale normalized, i.e., mean-centered and divided by the standard deviation of each variable) for each significant compound was also performed and results are displayed in Figs. 3 and 4.

Table 2 List of differential metabolites and lipids between asthmatic and COPD patients
Fig. 3
figure 3

Box plot analysis of main discriminant compounds. Box plot of the peak intensities for the 18 statistically downregulated compounds in COPD patients compared to asthmatics. The p-values were calculated by Student t-test and the asterisks show the level of significance between the two groups; * p-values < 0.05, ** p-values < 0.01, *** p-values < 0.001. Box plot analysis was performed by OriginLab® software

Fig. 4
figure 4

Box plot analysis of main discriminant compounds. Box plot of the peak intensities for the four statistically upregulated compounds in COPD patients compared to asthmatics. The p-values were calculated by Student t-test and the asterisks show the level of significance between the two groups; * p-values < 0.05. Box plot analysis was performed by OriginLab® software

Table 2 also lists the corresponding pathways that each identified compound was involved in, based on the knowledge derived from Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway database. The differentially expressed compounds found between asthmatic and COPD patients belonged to Glycerophospholipid and sphingolipid metabolism, Arginine and Proline metabolism, Tryptophan metabolism, Energy metabolism, FA biosynthesis, ABC transporters and Biotin metabolism (Table 2).

Validating the statistically significant compounds as potential biomarkers of differentiation between asthma and COPD by ROC analysis

The differentially expressed metabolites and lipids were examined by ROC curves, aimed at evaluating the diagnostic performances of potential biomarkers, including sensitivity and specificity (Figs. 5 and 6). The area under the ROC curve (AUC) was used to detect the accuracy and efficiency of this method in distinguishing asthma and COPD patients. Our data revealed that, among all the differentially expressed compounds, 18 exhibited the highest AUC (AUC > 0.7). In particular, PE (34:1), Spermine, Putrescine and PG(18:1;18:2) had AUC values around 0.8, indicating a good discriminating capability between asthmatic and COPD patients (Fig. 5). Other compounds, including AcCa (4:0), 6-Hydroxykynurenic acid, D-Rhamnose, Hexanoic acid, lysophosphatidylglycerol (LPG) (18:1), LPE (O-18:2), LPE (18:1), L-lysine, PC (33:0), PG (18:1;18:1), PG (16:1;18:1), Pentanoate, PE (36:3) and PC (38:3) exhibited AUC values around or greater than 0.7, showing moderate abilities in the differentiation between the two group (Fig. 6A–N). Subsequently, a cumulative ROC curve was created by combining the compounds previously mentioned. This cumulative ROC curve demonstrated an AUC of 0.849 and showed a strong sensitivity (0.992) and a reasonable specificity (0.616) as depicted in Fig. 6O. This combination of potential biomarkers proved to be a valuable tool that could well distinguish between asthmatic and COPD individuals.

Fig. 5
figure 5

Roc curve analysis of statistically significant compounds. AD ROC curves were constructed for compounds that exhibited the most effective diagnostic performance and a good ability to distinguish between these two groups, as measured by an Area under the Curve (AUC) around 0.8. ROC curves were acquired by Metaboanalyst software

Fig. 6
figure 6

Roc curve analysis of statistically significant compounds. AN ROC curves constructed for compounds that exhibited the most effective diagnostic performance and a moderate ability to distinguish between these two groups, as measured by an Area under the Curve (AUC) around or greater than 0.7. O Cumulative ROC curve constructed by combining all compounds that showed good/ moderate diagnostic capability. ROC and cumulative ROC curves were acquired by Metaboanalyst software

Associations between IS metabolites and lipids and lung functions by spearman correlation analysis

Lung functions FEV1 and FVC were significantly lower in the COPD compared to asthma group showing a p value < 0.0001 and p value < 0.01 respectively (Table 1).

FEV1/FVC ratio was significantly lower in the COPD compared to asthma group (p value < 0.01).

Metabolites and lipids compounds were correlated with lung function parameters (FEV1, FVC, FEV1/FVC) by Spearman correlation analysis (Fig. 7 and Additional file 1: Table S1).

Fig. 7
figure 7

Spearman correlation analysis between compounds and lung function parameters. Each panel shows the most significant metabolites and lipids correlated to FEV1(%) (A), FVC(%) (B) and FEV1/FVC (C). The x-axis indicates the Spearman’s correlation coefficient and the y-axis indicates the significance of the correlation (− log10 of the p-value)

Figure 7 shows the most significant compounds found to be correlated with FEV1, FVC and FEV1/FVC. In particular, FEV1(%) correlated positively, especially with Hexanoic acid, some fatty acids (FA) and PG species, Palmitic, Lauric and Myristic acid, and negatively with some triglycerides (TG) species, homocarnosine, inosine monophosphate (IMP), histidine, proline, citrulline, ornithine and quinolinic acid (Fig. 7A). FVC (%) showed a positive correlation with some LPE, LPC species, Palmitic acid, phosphate and glutathione disulfide. On the contrary, D-Rhamnose, carnosine, quinolinic acid, mannitol, thymidine and some amino acids including lysine, alanine, histidine, phenylalanine, aspartate correlated negatively with FVC (%) (Fig. 7B). Finally, FEV1/FVC exhibited a positive correlation with some TG, Lysospingomyelin (LSM) species, thymidine, fumarate and hexanoic acid, while a negative correlation was observed for some Diacylglycerols (DG), PC, LPC, FA species and 4-Pyridoxate (Fig. 7C).

Discussion

Asthma and COPD both originate from complex interactions between environmental agents and genetic factors. In COPD, mainly caused by tobacco smoke and airborne pollutants, airflow limitation is usually poorly reversible and persistently progressive. In asthma, bronchial obstruction is extremely variable and often reversible, as it is mainly due to the pathogenic actions of respiratory viruses and aeroallergens [12, 27]. A common feature shared by asthma and COPD is their remarkable heterogeneity, expressed as multiple phenotypes characterized by different clinical, functional, and pathobiologic patterns [28, 29].

IS supplies information about both inflammatory cells and mediators present in the airways, potentially important for phenotypic characterization of patients with chronic respiratory disorders, such as COPD and asthma [11, 12]. It is well accepted that recognition of specific metabolic profiles in COPD and asthma may improve patient management strategies, including diagnosis, offering new potential targets for future tailored therapy thus providing higher patients’ probabilities for survival [30, 31]. The present investigation provides a comprehensive comparative view of the metabolic and lipidomic profile of IS samples in asthmatic and COPD patients, offering not only new insights for investigation into the mechanism of disease and identification of novel diagnostic biomarkers, but also supplying additional information to complement previous research on asthma and COPD. Until now, only a limited number of reports analyzed IS, focusing on metabolomics and lipidomics differences between asthmatic and healthy individuals, or between COPD patients and controls, never directly comparing asthma and COPD patients [9, 32, 33]. In the majority of cases, the comparison of metabolomic and lipidomic profiles between asthma and COPD has been performed using other types of bodily fluids, such as serum and urine, which do not accurately reflect the lung's specific pathophysiological state [19,20,21]. Only one study has been recently reported, in which metabolomics and lipidomics analyses were performed on a proximal but invasive sample such as cultured bronchial epithelial cells (BECs) comparing healthy individuals to patients with severe asthma and, for comparison, patients with COPD [34]. Other reports used exhaled breath condensate (EBC) samples to distinguish between the metabolome of these two diseases [35, 36]. However, this specific sample may contain low concentrations of biomarkers due to dilution by the water vapor of exhaled air and is difficult to normalize [33].

Concerning asthma metabolomics, it is well established that metabolomic biosignatures in EBC, urine, and blood could distinguish asthma and asthma phenotypes [10]. Many studies found a significant alteration of lipid metabolism components such as PE, PG and SM in asthma [37,38,39]. Interestingly, in a sputum metabolomics study, which compared 20 asthmatic patients against 15 healthy subjects, glycolysis/gluconeogenesis, glycerophospholipid, and inositolphosphate metabolism pathways associated with asthma [40]. It is widely accepted that glycerophospholipid metabolism was the most significantly perturbed pathway in experimental allergic asthma [40, 41].

Metabolomics and lipidomics studies related to COPD in different kinds of samples have shown that the alteration of lipid and amino acid metabolism, energy production pathways, the imbalance of oxidations and antioxidations oxidative stress and protein malnutrition may lead to local and systemic inflammation and, may contribute to the development and progression of COPD [9, 42].

In line with reported studies, our dataset has highlighted the alteration of many of the metabolic pathways (Table 2 and Fig. 8) found in previous studies. To the best of our knowledge, this is the first UHPLC-MS based approach that screens and compares the metabolomic and lipidomic profile between asthma and COPD patients in IS samples. The use of our integrated and untargeted metabolomics/lipidomics approach by UHPLC-MS has shown significant differences and alterations in specific metabolic and lipid pathways prominently associated with asthma or COPD (Fig. 8). In particular, a total of 22 compounds, including 14 lipids species and 8 metabolites, were identified to be significantly different in the IS from the two groups (Fig. 3 and Fig. 4) and well differentiate asthmatic from COPD patients (Fig. 5 and Fig. 6). The analyzed data seem to strongly confirm that asthmatic patients show a typical IS metabolomics and lipidomics profile distinct from that found in COPD subjects. Levels of lipid metabolic intermediates such as PE and PG were found significantly increased in IS of asthmatic in comparison to COPD patients (Fig. 3). Interestingly, a significant dysregulation in arginine and proline metabolism pathways, evidenced by a strikingly elevated concentration of two polyamines spermine and putrescine in asthmatic vs COPD, was found (Fig. 3). In this differential metabolic pattern, deduced by our data, the observed decreased level of AcCa (4:0) and Lysine in the IS of asthmatic compared to COPD patients (Fig. 4) was noteworthy. These findings seem to fit well with the established main features on asthma and COPD pathophysiology, as it will be discussed in the following sections. Asthma pathobiology, involving activation of both innate and adaptive immune systems that stimulate chronic airway inflammation in response to allergens and several external agents, results in altered pathways of glycerophospholipids, arginine and proline, as well as tryptophan metabolism (Fig. 8). On the other hand, in COPD, altered pathways seems more prominently related to biotin and amino acids as well as energy metabolism, indicating the importance of inflammatory-immune processes with increased catabolism and alterations in the energy production (Fig. 8).

Fig. 8
figure 8

Main metabolic pathways and related analytes with significant variation of their levels in the induced sputum of COPD and asthmatic patients

Dysregulated metabolites between COPD and asthmatic patients

Aminoacid metabolism

Our results showed elevated levels of L-Lysine in IS of COPD compared to asthmatic patients (Fig. 4) and a significantly negative correlation between this metabolite and a lung function parameters (FVC%) was also found (Fig. 7 and Additional file 1:Table S1). Lysine is involved in biotin metabolism, as a product of biotin protein turnover during carboxylation reactions [43]. Importantly, it plays a pivotal role in the synthesis of carnitine, which is essential for fatty acids metabolism [44].

In accordance with our data, Khamis et al. found an increase of lysine levels in the urine samples of COPD compared to asthmatic patients [21]. Furthermore, other studies demonstrated that lysine was associated with worsening severity of COPD exacerbations [45, 46], suggesting a potential implication of this amino acid in this pathology.

Interestingly, lysine was dysregulated in ACO patients when compared with both asthma and COPD [47]. In particular, the mentioned study demonstrated that lysine levels are significantly lower in ACO patients as compared to asthma and COPD.

To the best of our knowledge, this is the first report to find significantly different levels of L-lysine between asthma and COPD patients in IS samples. Additionally, the negative correlation we found between this metabolite and the lung parameter FVC (%) (Fig. 7B and Additional file 1: Table S1), implies that high levels of lysine are associated with a decrease in FVC (%). Diminished FVC (%) values are typically observed in individuals diagnosed with COPD (Table 1). Consequently, our findings are consistent with the increased lysine levels observed in COPD patients (Fig. 4), suggesting a strong association of this amino acid with the development of COPD.

It is tempting to speculate that the lower levels of lysine found in asthmatic patients might be due to the higher metabolic demand in these subjects to generate acetyl-CoA. Concomitantly, acylcarnitine levels result lower in asthmatic compared to COPD patients (Figs. 4 and 8) because needed to supply the production of acetyl-CoA (Fig. 8). Therefore, a convergent regulatory synergistic mechanism could be involved in the production of acetyl-CoA providing its involvement in the TCA cycle (Fig. 8). Obviously, the significance of this finding warrants further investigations.

Polyamines biosynthesis

In our study, altered levels of two polyamines, spermine and putrescine, are observed between asthmatic and COPD patients (Fig. 3). As already stated, spermine and putrescine showing AUC values around 0.8 indicated a good discriminating capability to correctly estimate a random subject as asthmatic or COPD (Fig. 5).

Polyamines are naturally occurring small-sized super-cations that interact with intracellular macromolecules carrying negative charges. In mammals, polyamines are synthesized from the decarboxylation of arginine- and proline-derived ornithine by the enzyme ornithine decarboxylase (ODC) (Fig. 8) [48].

Changes in polyamine levels are associated to various pathological conditions, including asthma [49].

Polyamines have a significant role in lung physiology, as they are specifically accumulated in the epithelial cells of the lung [50].

Increased levels of polyamines have been reported in the blood of individuals with asthma and in the lungs of asthma mouse models [51, 52]. Polyamines were found to enhance the pathological aptitude of inflammatory cells, like mast cells and granulocytes, either stimulating the release of pro-inflammatory substances from these cells or extending their lifetime [53, 54].

North and colleagues detected elevated levels of putrescine and spermine in sputum samples of asthmatic patients after an allergen challenge [55]. They also demonstrated that endogenous concentrations of these polyamines were significantly increased in the lungs of a murine model of ovalbumin-induced allergic inflammation. In particular, their data showed that, when relevant concentrations of spermine were administered into the airways, airway responsiveness was enhanced both in naïve mice and in mice with allergic inflammation [55]. It is worth of note that an increased biosynthesis of polyamines exacerbates lung diseases; in fact, the synthetic pathway of these species competes for L-arginine at the expenses of NO production, leading in this way to bronchoconstriction (Fig. 8). By the way, exhaled NO is produced from arginine and several studies highlight the association with asthma and asthma severity [56, 57].

Our study demonstrated that spermine and putrescine were some of the most important metabolites for group segregation as a statistically significant increase of these two polyamines was observed in asthmatic compared to COPD patients (Figs. 3 and 5). Specifically, the asthma group showed higher levels of spermine and putrescine, confirming previous literature data.

Our results are the first to demonstrate different levels of these polyamines between asthma and COPD groups in IS samples. These findings might suggest that spermine and putrescine are strongly associated with airway hyperresponsiveness that typically occurs in asthmatic individuals [27]. Given the previous observation, it is tempting to hypothesize that their synthetic pathway is correlated to a lower level of arginine (see Fig. 8). Low arginine levels are associated with impaired smooth muscle relaxation [58]. In line with this, we observed a slight decrease in arginine levels in asthmatic subjects compared to COPD patients (Additional file 2: Fig. S1 and Additional file 3: Table S2). A similar trend of arginine levels was also found in urine samples in a metabolomic comparative analysis between asthma and COPD subjects [20]. Interestingly, cytrulline, which is formed as by-product together with NO from arginine (Fig. 8), was found among the seven top metabolite negatively associated with FVC (-log (p value) > 3) (Fig. 7 and Additional file 1: Table S1) in line with the above mentioned hypothesis.

Fatty acid biosynthesis related compounds

A statistically significant decrease of Hexanoic acid was found in the COPD group compared to the asthmatic one (Fig. 3). A significantly positive correlation between Hexanoic acid and lung function parameters (FEV1% and FEV1/FVC) was also observed (Fig. 8 and Table S1).

Hexanoic acid (or caproic acid) is a short-chain fatty acid (SCFAs) produced through bacterial fermentation of dietary fiber within the gut [59, 60]. Increasing evidence indicates the existence of a metabolic and immune connection between the gut and the respiratory system [61, 62] and suggests SCFAs as the key components in establishing this connection. The association between lung and gut was confirmed by the identification of SCFAs in the sputum of patients with cystic fibrosis [63]. Interestingly, SCFAs: (i) contribute to the regulation of immune-metabolic tone in the lung [64]; (ii) to have anti-inflammatory effects in asthma by downregulating allergic immune response and inflammasome pathway [65]; (iii) participate in maintaining the integrity of damaged airway epithelium by upregulating the expression of dense contact proteins such as ZO-1 [66]. This is particularly significant considering the existence of dysfunction in airway epithelial barrier and tight cell contacts associated with smoking and COPD [67]. Consequently, SCFAs might have a potential clinical significance in repairing airways barrier function [66].

Interestingly, a in study by Li et al. the level of seven SCFAs (including caproic acid) in the stool samples of COPD patients and healthy individuals was compared, showing a lower level of total SCFAs in the COPD group compared to healthy [68]. Although in this study the authors analyzed fecal samples, their findings may support the data from our study on IS samples, given the existence of a connection between the gut and the lungs.

Tryptophan metabolism related compound

6-Hydroxykynurenic acid was another metabolite found to be statistically significantly decreased in COPD compared to the asthmatic group (Fig. 3). This metabolite is a derivative of kynurenic acid that takes part to the tryptophan metabolism.

Tryptophan is an essential amino acid with important functions including cellular protein synthesis and the formation of the cytoskeleton [69] and it can be metabolized via different pathways in several cell types.

Tryptophan and its metabolites have been investigated in the context of inflammatory and respiratory diseases, revealing alterations in their levels during infections, as a consequence of the immune response [70]. In the lung, tryptophan catabolism leads to the accumulation of metabolites of the kynurenine pathway by the activity of the enzyme indoleamine-pyrrole 2,3-dioxygenase (IDO), thus resulting in immune tolerance and an anti-inflammatory effect. The induction of immune tolerance has been linked to lung cancer and HIV infection [71]. IDO, predominantly expressed in macrophages and dendritic cells, has also been shown to act as an important T cell immunomodulator [72].

Alteration in tryptophan metabolism was also observed in the sputum of COPD patients, attributed to a decrease in hydroxy-indolacetic acid levels [73], which could potentially lead to a diminished lung defense against neutrophilic inflammation and epithelial apoptosis. Ubhi and colleagues found decreased tryptophan levels in the serum of COPD patients with emphysema [74]. Noteworthy, Maneechotesuwan et al. observed a decrease in the activity of IDO in the sputum of COPD patients, and this reduction was associated to the severity of the disease. This finding suggests that the decline in IDO activity within sputum contributes to an environment that promotes neutrophilic inflammation [75]. Interestingly, IDO contributes to the suppression of intracellular reactive oxygen species (ROS) in acute lung allograft injury and improves the resistance of lung cells to oxidative stress [76]. Therefore, our data might suggest a decrease of IDO activity within IS of COPD compared to the asthmatic patients, which resulted in the observed lower levels of 6-hydroxykynurenic acid in COPD compared to asthma. The hypothesized decrease of IDO activity in COPD might also suggest a concomitant mechanism associated to increased expression of ROS resulting in major oxidative stress in COPD.

It is well known, that tryptophan and its metabolites can interact and/or generate ROS. This is an important topic, as alterations in the kynurenine pathway metabolites, oxidative stress, energetic deficit, cell death and inflammation may converge in a network of pathogenic mechanism [77]. Up to date, this pathway has attracted an intense research interest in the fields of aging and brain diseases [78, 79]. The identification of this novel metabolite in IS by our approach may open new avenues for exploring the involvement of this pathway also in chronic inflammatory lung diseases and this topic surely warrants future investigations.

Other compounds

In this investigation, a significantly altered level of phosphate was found between COPD and asthmatic patients (Fig. 3).

In its ionized form, inorganic phosphate, is involved in common cellular processes, such as regulating energy metabolism, promoting bone mineralization, facilitating membrane transport and enabling intracellular signaling [80]. Furthermore, respiratory muscle contractility, electrolyte transport and the response to inflammation can be affected by phosphate [81]. Recent investigations showed that alteration in phosphate levels can promote adverse outcomes in many diseases [82, 83]. Few studies have suggested that there is a correlation between serum phosphate levels and the severity of COPD. For instance, Stroda et al. showed that serum phosphate levels are lower in COPD patients compared to controls [84]. Farah and colleagues also suggested that hypophosphatemia was related to an increase of COPD exacerbation, duration of hospitalization and mortality rate [85]. On the contrary, other studies revealed that high level of serum phosphate was associated with mortality in COPD patients [81, 86].

According to our current knowledge, the present study is the first to show a significant decrease of phosphate levels in the IS of COPD compared to asthmatic patients; additionally, phosphate shows a positive correlation with FVC (%) (Figs. 3, 7B and Additional file 1: Table S1), suggesting a potential application of this metabolite in the differentiation between these two lung conditions.

Another metabolite able to discriminate COPD from asthmatic patients was d-Rhamnose.

Rhamnose is a naturally occurring deoxy sugar. Its D form (d-Rhamnose) is a component of cell surface polysaccharides in some species of bacteria such as Pseudomonas aeruginosa [87], a pathogenic bacteria that frequently cause infections, especially in individuals with compromised immune systems. These bacteria are a primary cause of lung infections in patients with cystic fibrosis and are linked to a significant rate of illness [88]. Pseudomonas aeruginosa has also been isolated from bronchial secretions in patients with COPD [89, 90] and asthma [91]. It has also been correlated with a high frequency of exacerbation, increased respiratory symptoms, rapidly worsening of lung function, poor quality of life and increased mortality [92].

To the best of our knowledge, our study is the first to identify d-Rhamnose as a potential discriminative biomarker between asthma and COPD in IS. Therefore, the upregulation of this metabolite found in COPD group might suggest a higher prevalence of Pseudomonas aeruginosa infection in COPD patients compared to asthmatic. This elevated level of D-Rhamnose in COPD patients in comparison to asthma is confirmed by the strong negative correlation between the metabolite and FVC(%) observed in our investigation (Fig. 7B and Additional file 1: Table S1). Specifically, as d-Rhamnose levels increase, FVC (%) values decrease. Notably, diminished values of this lung parameter were consistently observed in COPD patients (Table 1). Several studies support the hypothesis that bacterial colonization of the airways induces inflammation and impairs lung function (FEV1) [93]. Therefore, the metabolomics approach performed in this study might support future research to more accurately characterize the immune-phenotyping airway microbiota of both asthma and COPD, which in turn, could address the development of novel targeted immuno-therapeutics.

Dysregulated lipid species between COPD and asthmatic patients

Glycerophospholipid related metabolism

Glycerophospholipids play significant roles in cell membranes as major constituents, serving as storage materials for bioactive compounds. Recent research has highlighted the involvement of glycerophospholipids in the development of lung infections [94], asthma and COPD [19, 46, 95, 96]. These lipids are also constituents of lung surfactants, which can be compromised by smoke exposure [97]. They are released by airway epithelial cells into the alveoli to minimize alveolar surface tension and prevent the invasion of pathogens.

In our investigation, reduced levels of different PC [PC (33:0), PC (32:2), PC (38:3)], PE [PE (34:1), PE (36:3)] and LPE [LPE (18:1), LPE (O-18:2)] species were found in COPD group compared to asthma (Fig. 3).

In a study by Cruickshank-Quinn et al. based on an integrated transcriptome/metabolome analysis, the authors demonstrated a gradual decrease of some glycerophospholipids species, including PC in association with declining respiratory function in blood samples [46]. Kilk and colleagues found reduced level of some PCs, including PC (38:3), in the EBC of patients with COPD compared to control individuals [98]. Halper-Stromberg et al. revealed significant reductions in PE, PC in the bronchoalveolar lavage fluid (BALF) of COPD patients.

In the study by Gai et al., a decrease level of LPE was found in acute exacerbation stage of COPD patients and LPE (18:1) was among these lipids [97].

These findings suggest a correlation between LPE, PE, PC and COPD. Alteration of lipid metabolism associated to alveolar surfactants in COPD patients [46] might offer a promising avenue for future studies aimed at restoring alveolar surfactants and potentially treating COPD.

Dysregulation of glycerophospholipids metabolism was also found in patients with asthma. For instance, Jiang and colleagues observed a higher level in plasma of some PEs species including PE(38:1), with structural similarity to PE(34:1) (Fig. 3), in asthmatic compared to healthy controls and a positive correlation with the severity of the condition [37]. In another study, increased levels of LPC have been reported in the BALF of patients with asthma [99]. Ravi et al., also found upregulation of PCs, LPCs and LPEs in the bronchial epithelial cells from patients with severe asthma compared with controls [34].

In the present work, we found that various phosphatidylglycerol (PG), including PG (18:1;18:2), PG (18:1;18:1), PG (16:1;18:1), and LPG, including LPG (18:1), were reduced in COPD group compared to asthmatic. PGs play a role as pulmonary surfactants, which are crucial for supporting efficient gas exchange and maintaining optimal respiratory function. Additionally, they are actively involved in organizing the surfactant complex, modulating the immune response, and contributing to the body's defense mechanisms [100].

In individuals with COPD, it has been observed that surface lipid levels decreased by 60%, and there was a notable reduction in PG levels in the BALF [101]. In a work by Liu et al., PGs levels were decreased in serum of COPD patients with exacerbation compared to healthy and this reduction could potentially be attributed to airway damage resulting from insufficient surfactant function [100].

In our study a positive correlation was found between IS levels of LPC (18:3) and FVC% (Fig. 7 and Additional file 1: Table S1). As expected, significantly diminished levels of several GLP in subjects with COPD in comparison to asthmatic can lead to a more reduced lung functions in COPD patients.

To the best of our knowledge, although several above-mentioned investigations have already pointed out the alterations of glycerophospholipids metabolism in asthma and COPD patients compared to healthy individuals, only two studies including the present and the study by Ravi et al. [34], have pointed out on glycerophospholipids dysregulations in more specific and less systemic specimen. Targeted comparative lipidomics analyses on IS samples might drive future investigations to identify the exact role of GPL in chronic lung diseases as this sampling methods contains less confounding factors in comparison to other systemic bio-fluids.

Sphingolipid related metabolism

Sphingolipids play a crucial role as structural elements within cellular membranes [102]. Dysregulation in the sphingolipid metabolism has been observed in lung diseases [103, 104]. In particular, alterations in ceramide and sphingomyelin levels have been implicated in COPD. Ceramides were found to be increased in lung tissue of smokers and patients with COPD and have been associated with the induction of pulmonary vascular cell apoptosis caused by tobacco smoke [105]. Telenga and colleagues, identified significantly higher levels of 28 ceramides, including Cer(d18:1;16:0), in sputum from smokers with COPD compared to smokers without COPD [95].

On the other hand, low levels of SM have been associated with COPD severity [103].

Dysregulation in sphingomyelins and ceramides levels has also been reported in asthma. In fact, some SM and Cer species were found to be increased and decreased, respectively, in asthmatic patients compared to healthy individuals [37].

In our investigation, we found a statistically significant reduced level of SM(d32:1) and an increase in Cer(d18:1_16:0) in COPD patients compared to asthmatics. This increase in Cer and decrease in SM observed in our study could lead us to hypothesize an unbalance in production of SM starting from Cer, maybe due to a deficit in SM synthase expression in patients with COPD or alternatively to an increased degradation of SM due to an upregulation of sphingomyelinases activity.

To the best of our knowledge, our study is the first to evaluate the differential sphingolipids profiling between asthma and COPD in IS samples.

Energy metabolism related compounds

One of the other lipid compounds found to be differentially altered between the two analyzed groups was AcCa (4:0), or butyryl-l-carnitine. In particular, in our study it was more elevated in COPD compared to asthmatics patients. AcCa (4:0) is an acylcarnitine formed when an acyl group is transferred from coenzyme A to a molecule of L-carnitine.

L-carnitine and acylcarnitines are essential for oxidative catabolism of the fatty acids and changes in carnitine and acylcarnitine profiles can cause alteration in energy production, leading to deficiencies in fatty acid oxidation and complications in mitochondrial metabolism [106].

Several studies related to COPD have found changes in the level of acylcarnitines [107,108,109,110].

Kim et al. used LC-MS on both serum and urine samples in a study of 59 TB-related COPD patients, 70 smoking-related COPD patients, and 39 healthy controls (including never-smokers and smokers) and found differences in the acyl carnitines [109].

Similar results were shown in a COPDGene discovery cohort of 839 subjects and a SPIROMICS.

replication cohort of 446 subjects using LC-MS on serum samples [107]. The differences in carnitines in COPD Gene and SPIROMICS were more pronounced in women compared to men. In particular, in this investigation, propionylcarnitine (C3) showed a positive association with COPD status; on the contrary medium chain, decanoylcarnitine (C10), cis-4-decenoylcarnitine (C10:1), laurylcarnitine(C12) and myristoleoylcarnitine (C14:1) showed a negative correlation. Interestingly, serum 2-methylbutyrylcarnitine (C5) was found among the three top metabolite positively associated with FEV1 in a large cross-sectional study (n = 4742) of three population cohorts with 393 COPD cases [111].

Naz et al. observed an elevation in circulating levels of acylcarnitines in COPD, suggesting a greater energy demand which is reflected in the increased transfer of acetyl coenzyme A to the tricarboxylic acid (TCA) cycle [110]. The upregulation of the TCA cycle results in higher ATP production and the elevated levels of extracellular ATP in the airway have been associated with the development of COPD. This is due to its role in recruiting and activating inflammatory cells, thereby causing inflammation and tissue deterioration [112]. However, an elevated endobronchial ATP concentration has been also detected in models of asthma [113]. An increase of endobronchial ATP both in asthma and COPD would support the pathophysiologic similarities between these two diseases as already established from genetic investigations supporting this concept [114].

Reinke and colleagues, in a large-scale MS-based investigation of the urinary metabolome in adult patients with asthma, with the aim to delineate disease- and oral corticosteroids-associated metabolic differences in asthma, found that short-chain carnitines represented the strongest metabolic signature associated with asthma severity. Specifically, they found a decreased abundance of acetyl − carnitine (C2) and propionyl − carnitine (C3:0) in relation to disease severity which was independent of oral corticosteroids treatment [115].

In our investigation, the level of the acylcarnitine (butyryl-l-carnitine) resulted to be significant increased in COPD group compared to asthmatic (Fig. 4). This result might suggest that, in line with what was previously hypothesized [110] the increased levels of acylcarnitine (butyryl-l-carnitine) in IS of COPD subjects may possibly be associated to the higher demand of production of acetyl-CoA in request to an higher energy consumption in COPD compared to asthmatic patients. This hypothesis would fit with the concomitant increased level of Lysine in COPD subjects compared to the asthmatic as already observed previously [See section “Aminoacid metabolism” and Fig. 8), which would contribute to acetyl-CoA production via Acetoacetate (Fig. 8).

Limitations of the study

The small group size and the fact that both asthma and COPD are multifactorial conditions characterized not only by a great heterogeneity but also by an important phenotypic complexity [116] may possibly limit the accuracy of this preliminary panel of putative diagnostic biomarkers to distinguish asthma from COPD. Future studies are required to confirm the selected pattern of metabolites and lipids in larger sample cohorts in order to increase the confidence and the statistical power of the analysis, which could provide a better understanding of different metabolism in asthma and COPD. At the same time, other novel metabolites and lipid intermediates differentiated in asthma and COPD could be more accurately revealed in a larger cohort.

Several factors may be causative of metabolome fluctuation including sex, age and smoking status. The mismatched age between Asthma and COPD patients as well as the sex and smoking status differences (Table 1) make the interpretation of the observed dysregulated metabo-lipidomics profile challenging because it is difficult distinguishing the differences related to aging, sex and tobacco exposure from those related to the specific disease. Nonetheless, in clinical practice, subjects suffering of COPD commonly include smokers and aged individuals. Therefore, it is quite challenging exploring these confounding factors also on larger cohorts. Further validation on stratified population subsets for both asthmatic and COPD subjects might be also an important goal to reach in future studies for better evaluate phenotypic variation and severity degrees also in relation to metabolomics dataset and lung functions.

The fact that no healthy or other control group have been included is another limitation of this study. Induction of sputum may cause such discomfort especially in healthy individuals with less expectorate fluids or in those with bronchiectasis with less expectorate fluids and consequently this is critical issue in heathy subjects sampling procedure. However, the highly specificity of the IS in the analyzed groups, which accurately reflects inflammatory changes at the site of tissue damage, provides more accurate dataset about both inflammatory cells and mediators present in the airways better than other fluid such as serum or urine which instead provide confounding systemic information.

Conclusions

Despite intensive research efforts involving huge economic and human resources, molecular and cellular mechanisms underlying the pathology and differentiation between COPD and asthma are not yet well understood. In this study, with the aim to more accurately differentiate asthma and COPD, an untargeted metabolomics and lipidomics approach has been performed based on UHPLC-MS and MS/MS analysis of IS, a highly specific fluid reflecting the closest features of the respiratory tract. Therefore, the resulting molecular signatures extrapolated by this pilot study might reflect specific inflammatory and immune response specifically associated to asthma or COPD. Our comparative metabolomics analysis revealed 22 metabolites and lipids which differ in a statistically significant manner in the IS of asthmatic and COPD subjects. Our findings demonstrated a prominent group segregation for PE (34:1), PG (18:1;18:2), spermine and putrescine. Many altered metabolic pathways have been identified including glycerophospholipids, arginine and proline, biotin and aminoacids, tryptophan and energy metabolism, among those, which prominently segregated asthma and COPD. Further targeted analysis will be helpful to validate these IS biomarkers. If validated on a larger prospective cohort, the metabolite panel of biomarkers identified in the IS in the present study might suggest novel strategies to address more accurate asthma and COPD diagnosis, stratification and interventions.

Availability of data and materials

All data supporting the findings of this study are available within the paper and its Supplementary Information. However, the individual data of patients participating to this study are not openly available due to reasons of sensitivity and are available from the corresponding author upon reasonable request. The datasets used and/or analyzed during the current study are available from the corresponding author upon reasonable request.

Change history

Abbreviations

ACO:

Asthma-COPD overlap

AUC:

Area under the curve

BALF:

Bronchoalveolar lavage fluid

BECs:

Bronchial epithelial cells

COPD:

Chronic obstructive pulmonary disease

Cer:

Ceramide

EBC:

Exhaled breath condensate

FEV1%:

Forced expiratory volume in 1 s percentage

FVC%:

Forced vital capacity percentage predicted

GPC:

Glycerophosphatidylcholine

GPE:

Glycerophosphoethanolamines

GPG:

Glycerylphosphorylglycerols

IDO:

Indoleamine-pyrrole 2,3-dioxygenase

IS:

Induced sputum

KEGG:

Kyoto encyclopedia of genes and genomes

KYN:

Kynurenine

LC–MS:

Liquid chromatography-mass spectrometry

LPE:

Lysophosphatidylethanolamine

LPG:

Lysophosphatidylglycerol

MS:

Mass spectrometry

OPLS-DA:

Orthogonal partial least squares discriminant analysis

PC:

Phosphatidylcholine

PE:

Phosphatidylethanolamine

PG:

Phosphatidylglycerol

PLS-DA:

Partial least squares discriminant analysis

PCA:

Principal component analysis

ROC:

Receiver operating characteristic

SCFA:

Short-chain fatty acid

SD:

Standard deviation

SM:

Sphingomyelin

TRP:

Tryptophan

UHPLC:

Ultra-high-pressure liquid chromatography

VIP:

Variable importance plots

References

  1. James SLAD, Abate D, Abate KH, Abay SM, Abbafati C, Abbasi N, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 354 diseases and injuries for 195 countries and territories, 1990–2017: a systematic analysis for the global burden of disease study 2017. Lancet. 2018;392(10159):1789–858.

    Article  Google Scholar 

  2. Mattila T, Vasankari T, Kauppi P, Mazur W, Härkänen T, Heliövaara M. Mortality of asthma, COPD, and asthma-COPD overlap during an 18 year follow up. Respir Med. 2023;207: 107112.

    Article  PubMed  Google Scholar 

  3. Hogg JC. Pathophysiology of airflow limitation in chronic obstructive pulmonary disease. Lancet. 2004;364(9435):709–21.

    Article  PubMed  Google Scholar 

  4. Postma DS, Rabe KF. The asthma-COPD overlap syndrome. N Engl J Med. 2015;373(13):1241–9.

    Article  CAS  PubMed  Google Scholar 

  5. Marcon A, Locatelli F, Dharmage SC, Svanes C, Heinrich J, Leynaert B, et al. The coexistence of asthma and COPD: risk factors, clinical history and lung function trajectories. Eur Respir J. 2021. https://doi.org/10.1183/13993003.04656-2020.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Slats A, Taube C. Asthma and chronic obstructive pulmonary disease overlap: asthmatic chronic obstructive pulmonary disease or chronic obstructive asthma? Ther Adv Respir Dis. 2016;10(1):57–71.

    Article  PubMed  Google Scholar 

  7. Çolak Y. Undiagnosed (or unrecognised) COPD and asthma: does active case-finding identify clinically impaired patients with treatment potential. Am J Respir Crit Care Med. 2023. https://doi.org/10.1164/rccm.202310-1793ED.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Çolak Y, Afzal S, Nordestgaard BG, Vestbo J, Lange P. Prognosis of asymptomatic and symptomatic, undiagnosed COPD in the general population in Denmark: a prospective cohort study. Lancet Respir Med. 2017;5(5):426–34.

    Article  PubMed  Google Scholar 

  9. Dasgupta S, Ghosh N, Bhattacharyya P, Roy Chowdhury S, Chaudhury K. Metabolomics of asthma, COPD, and asthma-COPD overlap: an overview. Crit Rev Clin Lab Sci. 2023;60(2):153–70.

    Article  PubMed  Google Scholar 

  10. Kelly RS, Dahlin A, McGeachie MJ, Qiu W, Sordillo J, Wan ES, et al. Asthma metabolomics and the potential for integrative omics in research and the clinic. Chest. 2017;151(2):262–77.

    Article  PubMed  Google Scholar 

  11. Terracciano R, Preianò M, Palladino GP, Carpagnano GE, Barbaro MP, Pelaia G, et al. Peptidome profiling of induced sputum by mesoporous silica beads and MALDI-TOF MS for non-invasive biomarker discovery of chronic inflammatory lung diseases. Proteomics. 2011;11(16):3402–14.

    Article  CAS  PubMed  Google Scholar 

  12. Pelaia G, Terracciano R, Vatrella A, Gallelli L, Busceti MT, Calabrese C, Stellato C, Savino R, Maselli R. Application of proteomics and peptidomics to COPD. Biomed Res Int. 2014;2014: 764581.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Gao D, Zhang L, Song D, Lv J, Wang L, Zhou S, et al. Values of integration between lipidomics and clinical phenomes in patients with acute lung infection, pulmonary embolism, or acute exacerbation of chronic pulmonary diseases: a preliminary study. J Transl Med. 2019;17(1):162.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Qiu S, Cai Y, Yao H, Lin C, Xie Y, Tang S, et al. Small molecule metabolites: discovery of biomarkers and therapeutic targets. Signal Transduct Target Ther. 2023;8(1):132.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Chen L, Lu W, Wang L, Xing X, Chen Z, Teng X, et al. Metabolite discovery through global annotation of untargeted metabolomics data. Nat Method. 2021;18(11):1377–85.

    Article  Google Scholar 

  16. Luan H, Gu W, Li H, Wang Z, Lu L, Ke M, et al. Serum metabolomic and lipidomic profiling identifies diagnostic biomarkers for seropositive and seronegative rheumatoid arthritis patients. J Transl Med. 2021;19(1):500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wang R, Li B, Lam SM, Shui G. Integration of lipidomics and metabolomics for in-depth understanding of cellular mechanism and disease progression. J Genet Genom. 2020;47(2):69–83.

    Article  CAS  Google Scholar 

  18. Correnti S, Preianò M, Fregola A, Gamboni F, Stephenson D, Savino R, et al. Seminal plasma untargeted metabolomic and lipidomic profiling for the identification of a novel panel of biomarkers and therapeutic targets related to male infertility. Front Pharmacol. 2023;14:1275832.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liang Y, Gai XY, Chang C, Zhang X, Wang J, Li TT. Metabolomic profiling differences among asthma, COPD, and healthy subjects: A LC-MS-based metabolomic analysis. Biomed Environ Sci. 2019;32(9):659–72.

    CAS  PubMed  Google Scholar 

  20. Adamko DJ, Nair P, Mayers I, Tsuyuki RT, Regush S, Rowe BH. Metabolomic profiling of asthma and chronic obstructive pulmonary disease: a pilot study differentiating diseases. J Allerg Clin Immunol. 2015;136(3):571-80.e3.

    Article  Google Scholar 

  21. Khamis MM, Holt T, Awad H, El-Aneed A, Adamko DJ. Comparative analysis of creatinine and osmolality as urine normalization strategies in targeted metabolomics for the differential diagnosis of asthma and COPD. Metabolomics. 2018;14(9):115.

    Article  PubMed  Google Scholar 

  22. D’Amato M, Iadarola P, Viglio S. Proteomic analysis of human sputum for the diagnosis of lung disorders: where are we today? Int J Mol Sci. 2022. https://doi.org/10.3390/ijms23105692.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Global Strategy for Asthma Management and Prevention. https://ginasthma.org/wp-content/uploads/2019/06/GINA-2019-main-report-June-2019-wms.pdf. Accessed on 14 December 2023.

  24. Global Strategy for Prevention, Diagnosis and Management of COPD. https://goldcopd.org/wp-content/uploads/2023/03/GOLD-2023-ver-1.3-17Feb2023_WMV.pdf. Accessed on 14 December 2023.

  25. Nemkov T, Reisz JA, Gehrke S, Hansen KC, D’Alessandro A. High-throughput metabolomics: isocratic and gradient mass spectrometry-based methods. Method Mol Biol. 2019;1978:13–26.

    Article  CAS  Google Scholar 

  26. Reisz JA, Zheng C, D’Alessandro A, Nemkov T. Untargeted and semi-targeted lipid analysis of biological samples using mass spectrometry-based metabolomics. Method Mol Biol. 2019;1978:121–35.

    Article  CAS  Google Scholar 

  27. Barnes PJ. Cellular and molecular mechanisms of asthma and COPD. Clin Sci. 2017;131(13):1541–58.

    Article  CAS  Google Scholar 

  28. Han MK, Agusti A, Calverley PM, Celli BR, Criner G, Curtis JL, et al. Chronic obstructive pulmonary disease phenotypes: the future of COPD. Am J Respir Crit Care Med. 2010;182(5):598–604.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Wenzel SE. Complex phenotypes in asthma: current definitions. Pulm Pharmacol Ther. 2013;26(6):710–5.

    Article  CAS  PubMed  Google Scholar 

  30. Nickler M, Ottiger M, Steuer C, Huber A, Anderson JB, Müller B, et al. Systematic review regarding metabolic profiling for improved pathophysiological understanding of disease and outcome prediction in respiratory infections. Respir Res. 2015;16:125.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Sim S, Choi Y, Park HS. Potential metabolic biomarkers in adult asthmatics. Metabolites. 2021. https://doi.org/10.3390/metabo11070430.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Pulik K, Mycroft K, Korczyński P, Ciechanowicz AK, Górska K. Metabolomic analysis of respiratory epithelial lining fluid in patients with chronic obstructive pulmonary disease—a systematic review. Cells. 2023. https://doi.org/10.3390/cells12060833.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Sinha A, Desiraju K, Aggarwal K, Kutum R, Roy S, Lodha R, et al. Exhaled breath condensate metabolome clusters for endotype discovery in asthma. J Transl Med. 2017;15(1):262.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ravi A, Goorsenberg AWM, Dijkhuis A, Dierdorp BS, Dekker T, van Weeghel M, et al. Metabolic differences between bronchial epithelium from healthy individuals and patients with asthma and the effect of bronchial thermoplasty. J Allerg Clin Immunol. 2021;148(5):1236–48.

    Article  CAS  Google Scholar 

  35. Maniscalco M, Paris D, Melck DJ, Molino A, Carone M, Ruggeri P, et al. Differential diagnosis between newly diagnosed asthma and COPD using exhaled breath condensate metabolomics: a pilot study. Eur Respir J. 2018;51(3):1701825.

    Article  PubMed  Google Scholar 

  36. Fens N, Roldaan AC, van der Schee MP, Boksem RJ, Zwinderman AH, Bel EH, et al. External validation of exhaled breath profiling using an electronic nose in the discrimination of asthma with fixed airways obstruction and chronic obstructive pulmonary disease. Clin Exp Allerg. 2011;41(10):1371–8.

    Article  CAS  Google Scholar 

  37. Jiang T, Dai L, Li P, Zhao J, Wang X, An L, et al. Lipid metabolism and identification of biomarkers in asthma by lipidomic analysis. Biochim Biophys Acta Mol Cell Biol Lipid. 2021;1866(2): 158853.

    Article  CAS  Google Scholar 

  38. Ntontsi P, Ntzoumanika V, Loukides S, Benaki D, Gkikas E, Mikros E, et al. EBC metabolomics for asthma severity. J Breath Res. 2020;14(3): 036007.

    Article  CAS  PubMed  Google Scholar 

  39. Ried JS, Baurecht H, Stückler F, Krumsiek J, Gieger C, Heinrich J, et al. Integrative genetic and metabolite profiling analysis suggests altered phosphatidylcholine metabolism in asthma. Allergy. 2013;68(5):629–36.

    Article  CAS  PubMed  Google Scholar 

  40. Tian M, Chen M, Bao YL, Xu CD, Qin QZ, Zhang WX, et al. Sputum metabolomic profiling of bronchial asthma based on quadruple time-of-flight mass spectrometry. Int J Clin Exp Pathol. 2017;10(10):10363–73.

    PubMed  PubMed Central  Google Scholar 

  41. Quinn KD, Schedel M, Nkrumah-Elie Y, Joetham A, Armstrong M, Cruickshank-Quinn C, et al. Dysregulation of metabolic pathways in a mouse model of allergic asthma. Allergy. 2017;72(9):1327–37.

    Article  CAS  PubMed  Google Scholar 

  42. Ran N, Pang Z, Gu Y, Pan H, Zuo X, Guan X, et al. An updated overview of metabolomic profile changes in chronic obstructive pulmonary disease. Metabolites. 2019;9(6):111.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Pacheco-Alvarez D, Solórzano-Vargas RS, Del Río AL. Biotin in metabolism and its relationship to human disease. Arch Med Res. 2002;33(5):439–47.

    Article  CAS  PubMed  Google Scholar 

  44. Vaz FM, Wanders RJ. Carnitine biosynthesis in mammals. Biochem J. 2002;361(Pt 3):417–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Halper-Stromberg E, Gillenwater L, Cruickshank-Quinn C, O’Neal WK, Reisdorph N, Petrache I, et al. Bronchoalveolar lavage Fluid from COPD patients reveals more compounds associated with disease than matched plasma. Metabolites. 2019;9(8):157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Cruickshank-Quinn CI, Jacobson S, Hughes G, Powell RL, Petrache I, Kechris K, et al. Metabolomics and transcriptomics pathway approach reveals outcome-specific perturbations in COPD. Sci Rep. 2018;8(1):17132.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ghosh N, Choudhury P, Subramani E, Saha D, Sengupta S, Joshi M, et al. Metabolomic signatures of asthma-COPD overlap (ACO) are different from asthma and COPD. Metabolomics. 2019;15(6):87.

    Article  CAS  PubMed  Google Scholar 

  48. Sagar NA, Tarafdar S, Agarwal S, Tarafdar A, Sharma S. Polyamines: functions, metabolism, and role in human disease management. Med Sci. 2021;9(2):44.

    CAS  Google Scholar 

  49. Jain V. Role of polyamines in asthma pathophysiology. Med Sci. 2018;6(1):4.

    Google Scholar 

  50. Hoet PH, Nemery B. Polyamines in the lung: polyamine uptake and polyamine-linked pathological or toxicological conditions. Am J Physiol Lung Cell Mol Physiol. 2000;278(3):L417-33.

    Article  CAS  PubMed  Google Scholar 

  51. Kurosawa M, Shimizu Y, Tsukagoshi H, Ueki M. Elevated levels of peripheral-blood, naturally occurring aliphatic polyamines in bronchial asthmatic patients with active symptoms. Allergy. 1992;47(6):638–43.

    Article  CAS  PubMed  Google Scholar 

  52. Zimmermann N, King NE, Laporte J, Yang M, Mishra A, Pope SM, et al. Dissection of experimental asthma with DNA microarray analysis identifies arginase in asthma pathogenesis. J Clin Invest. 2003;111(12):1863–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kurosawa M, Uno D, Kobayashi S. Naturally occurring aliphatic polyamines-induced histamine release from rat peritoneal mast cells. Allerg. 1991;46(5):349–54.

    Article  CAS  Google Scholar 

  54. Ilmarinen P, Moilanen E, Erjefält JS, Kankaanranta H. The polyamine spermine promotes survival and activation of human eosinophils. J Allerg Clin Immunol. 2015;136(2):482-4.e11.

    Article  CAS  Google Scholar 

  55. North ML, Grasemann H, Khanna N, Inman MD, Gauvreau GM, Scott JA. Increased ornithine-derived polyamines cause airway hyperresponsiveness in a mouse model of asthma. Am J Respir Cell Mol Biol. 2013;48(6):694–702.

    Article  CAS  PubMed  Google Scholar 

  56. Zuo L, Koozechian MS, Chen LL. Characterization of reactive nitrogen species in allergic asthma. Ann Allerg Asthma Immunol. 2014;112(1):18–22.

    Article  CAS  Google Scholar 

  57. Ghosh S, Erzurum SC. Nitric oxide metabolism in asthma pathophysiology. Biochim Biophys Acta. 2011;1810(11):1008–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Maarsingh H, Leusink J, Zaagsma J, Meurs H. Role of the l-citrulline/l-arginine cycle in iNANC nerve-mediated nitric oxide production and airway smooth muscle relaxation in allergic asthma. Eur J Pharmacol. 2006;546(1–3):171–6.

    Article  CAS  PubMed  Google Scholar 

  59. Wong JM, de Souza R, Kendall CW, Emam A, Jenkins DJ. Colonic health: fermentation and short chain fatty acids. J Clin Gastroenterol. 2006;40(3):235–43.

    Article  CAS  PubMed  Google Scholar 

  60. Rajendiran E, Ramadass B, Ramprasath V. Understanding connections and roles of gut microbiome in cardiovascular diseases. Can J Microbiol. 2021;67(2):101–11.

    Article  CAS  PubMed  Google Scholar 

  61. Enaud R, Prevel R, Ciarlo E, Beaufils F, Wieërs G, Guery B, et al. The gut-lung axis in health and respiratory diseases: a place for inter-organ and inter-kingdom crosstalks. Front Cell Infect Microbiol. 2020;10:9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Anand S, Mande SS. Diet, Microbiota and Gut-Lung Connection. Front Microbiol. 2018;9:2147.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Ghorbani P, Santhakumar P, Hu Q, Djiadeu P, Wolever TM, Palaniyar N, et al. Short-chain fatty acids affect cystic fibrosis airway inflammation and bacterial growth. Eur Respir J. 2015;46(4):1033–45.

    Article  CAS  PubMed  Google Scholar 

  64. Liu Q, Tian X, Maruyama D, Arjomandi M, Prakash A. Lung immune tone via gut-lung axis: gut-derived LPS and short-chain fatty acids’ immunometabolic regulation of lung IL-1β, FFAR2, and FFAR3 expression. Am J Physiol Lung Cell Mol Physiol. 2021;321(1):L65-l78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yoon HJ, Park MK, Lee H, Park TS, Park DW, Moon JY, et al. Effects of respiratory short-chain fatty acids on bronchial inflammation in asthma. World Allerg Organ J. 2020;13(8):100204.

    Article  Google Scholar 

  66. Richards LB, Li M, Folkerts G, Henricks PAJ, Garssen J, van Esch B. Butyrate and propionate restore the cytokine and house dust mite compromised barrier function of human bronchial airway epithelial cells. Int J Mol Sci. 2020;22(1):65.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Tatsuta M, Kan OK, Ishii Y, Yamamoto N, Ogawa T, Fukuyama S, et al. Effects of cigarette smoke on barrier function and tight junction proteins in the bronchial epithelium: protective role of cathelicidin LL-37. Respir Res. 2019;20(1):251.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Li N, Dai Z, Wang Z, Deng Z, Zhang J, Pu J, et al. Gut microbiota dysbiosis contributes to the development of chronic obstructive pulmonary disease. Respir Res. 2021;22(1):274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Li C, Zhao H. Tryptophan and its metabolites in lung cancer: basic functions and clinical significance. Front Oncol. 2021;11: 707277.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Naz S, Bhat M, Ståhl S, Forsslund H, Sköld CM, Wheelock ÅM, et al. Dysregulation of the tryptophan pathway evidences gender differences in COPD. Metabolites. 2019;9(10):212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Gulcev M, Reilly C, Griffin TJ, Broeckling CD, Sandri BJ, Witthuhn BA, et al. Tryptophan catabolism in acute exacerbations of chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2016;11:2435–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nat Rev Immunol. 2004;4(10):762–74.

    Article  CAS  PubMed  Google Scholar 

  73. Yan Z, Chen B, Yang Y, Yi X, Wei M, Ecklu-Mensah G, et al. Multi-omics analyses of airway host-microbe interactions in chronic obstructive pulmonary disease identify potential therapeutic interventions. Nat Microbiol. 2022;7(9):1361–75.

    Article  CAS  PubMed  Google Scholar 

  74. Ubhi BK, Cheng KK, Dong J, Janowitz T, Jodrell D, Tal-Singer R, et al. Targeted metabolomics identifies perturbations in amino acid metabolism that sub-classify patients with COPD. Mol Biosyst. 2012;8(12):3125–33.

    Article  CAS  PubMed  Google Scholar 

  75. Maneechotesuwan K, Kasetsinsombat K, Wongkajornsilp A, Barnes PJ. Decreased indoleamine 2,3-dioxygenase activity and IL-10/IL-17A ratio in patients with COPD. Thorax. 2013;68(4):330–7.

    Article  PubMed  Google Scholar 

  76. Liu H, Liu L, Fletcher BS, Visner GA. Novel action of indoleamine 2,3-dioxygenase attenuating acute lung allograft injury. Am J Respir Crit Care Med. 2006;173(5):566–72.

    Article  CAS  PubMed  Google Scholar 

  77. Reyes Ocampo J, Lugo Huitrón R, González-Esquivel D, Ugalde-Muñiz P, Jiménez-Anguiano A, Pineda B, et al. Kynurenines with neuroactive and redox properties: relevance to aging and brain diseases. Oxid Med Cell Longev. 2014;2014: 646909.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Savonije K, Weaver DF. The role of tryptophan metabolism in alzheimer’s disease. Brain Sci. 2023;13(2):292.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Sorgdrager FJH, Naudé PJW, Kema IP, Nollen EA, Deyn PP. Tryptophan metabolism in inflammaging from biomarker to therapeutic target. Front Immunol. 2019;10:2565.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kalantar-Zadeh K, Ganz T, Trumbo H, Seid MH, Goodnough LT, Levine MA. Parenteral iron therapy and phosphorus homeostasis: a review. Am J Hematol. 2021;96(5):606–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Li S, Huang Q, Nan W, He B. Association between serum phosphate and in-hospital mortality of patients with AECOPD: a retrospective analysis on eICU database. Heliyon. 2023;9(9): e19748.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Jung SY, Kwon J, Park S, Jhee JH, Yun HR, Kim H, et al. Phosphate is a potential biomarker of disease severity and predicts adverse outcomes in acute kidney injury patients undergoing continuous renal replacement therapy. PLoS ONE. 2018;13(2): e0191290.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Xu H, Evans M, Gasparini A, Szummer K, Spaak J, Ärnlöv J, et al. Outcomes associated to serum phosphate levels in patients with suspected acute coronary syndrome. Int J Cardiol. 2017;245:20–6.

    Article  PubMed  Google Scholar 

  84. Stroda A, Brandenburg V, Daher A, Cornelissen C, Goettsch C, Keszei A, et al. Serum phosphate and phosphate-regulatory hormones in COPD patients. Respir Res. 2018;19(1):183.

    Article  PubMed  PubMed Central  Google Scholar 

  85. Farah R, Khamisy-Farah R, Arraf Z, Jacobson L, Makhoul N. Hypophosphatemia as a prognostic value in acute exacerbation of COPD. Clin Respir J. 2013;7(4):407–15.

    Article  CAS  PubMed  Google Scholar 

  86. Campos-Obando N, Lahousse L, Brusselle G, Stricker BH, Hofman A, Franco OH, et al. Serum phosphate levels are related to all-cause, cardiovascular and COPD mortality in men. Eur J Epidemiol. 2018;33(9):859–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Giraud MF, Naismith JH. The rhamnose pathway. Curr Opin Struct Biol. 2000;10(6):687–96.

    Article  CAS  PubMed  Google Scholar 

  88. Melamed J, Kocev A, Torgov V, Veselovsky V, Brockhausen I. Biosynthesis of the Pseudomonas aeruginosa common polysaccharide antigen by d-Rhamnosyltransferases WbpX and WbpY. Glycoconj J. 2022;39(3):393–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Eklöf J, Sørensen R, Ingebrigtsen TS, Sivapalan P, Achir I, Boel JB, et al. Pseudomonas aeruginosa and risk of death and exacerbations in patients with chronic obstructive pulmonary disease: an observational cohort study of 22 053 patients. Clin Microbiol Infect. 2020;26(2):227–34.

    Article  PubMed  Google Scholar 

  90. Jacobs DM, Ochs-Balcom HM, Noyes K, Zhao J, Leung WY, Pu CY, et al. Impact of pseudomonas aeruginosa isolation on mortality and outcomes in an outpatient chronic obstructive pulmonary disease cohort. Open Forum Infect Dis. 2020;7(1):ofz546.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Zhang Q, Illing R, Hui CK, Downey K, Carr D, Stearn M, et al. Bacteria in sputum of stable severe asthma and increased airway wall thickness. Respir Res. 2012;13(1):35.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Garcia-Clemente M, de la Rosa D, Máiz L, Girón R, Blanco M, Olveira C, et al. Impact of pseudomonas aeruginosa infection on patients with chronic inflammatory airway diseases. J Clin Med. 2020;9(12):3800.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Kayongo A, Robertson NM, Siddharthan T, Ntayi ML, Ndawula JC, Sande OJ, et al. Airway microbiome-immune crosstalk in chronic obstructive pulmonary disease. Front Immunol. 2022;13:1085551.

    Article  CAS  PubMed  Google Scholar 

  94. Mander A, Langton-Hewer S, Bernhard W, Warner JO, Postle AD. Altered phospholipid composition and aggregate structure of lung surfactant is associated with impaired lung function in young children with respiratory infections. Am J Respir Cell Mol Biol. 2002;27(6):714–21.

    Article  CAS  PubMed  Google Scholar 

  95. Telenga ED, Hoffmann RF, Ruben tK, Hoonhorst SJ, Willemse BW, van Oosterhout AJ, et al. Untargeted lipidomic analysis in chronic obstructive pulmonary disease uncovering sphingolipids. Am J Respir Crit Care Med. 2014;190(2):155–64.

    Article  CAS  PubMed  Google Scholar 

  96. Chen H, Li Z, Dong L, Wu Y, Shen H, Chen Z. Lipid metabolism in chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2019;14:1009–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Gai X, Guo C, Zhang L, Zhang L, Abulikemu M, Wang J, et al. Serum glycerophospholipid profile in acute exacerbation of chronic obstructive pulmonary disease. Front Physiol. 2021;12: 646010.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Kilk K, Aug A, Ottas A, Soomets U, Altraja S, Altraja A. Phenotyping of chronic obstructive pulmonary disease based on the integration of metabolomes and clinical characteristics. Int J Mol Sci. 2018;19(3):666.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Kang YP, Lee WJ, Hong JY, Lee SB, Park JH, Kim D, et al. Novel approach for analysis of bronchoalveolar lavage fluid (BALF) using HPLC-QTOF-MS-based lipidomics: lipid levels in asthmatics and corticosteroid-treated asthmatic patients. J Proteome Res. 2014;13(9):3919–29.

    Article  CAS  PubMed  Google Scholar 

  100. Liu X, Zhang H, Si Y, Du Y, Wu J, Li J. High-coverage lipidomics analysis reveals biomarkers for diagnosis of acute exacerbation of chronic obstructive pulmonary disease. J Chromatogr B Analyt Technol Biomed Life Sci. 2022;1201–1202: 123278.

    Article  PubMed  Google Scholar 

  101. Agudelo CW, Kumley BK, Area-Gomez E, Xu Y, Dabo AJ, Geraghty P, et al. Decreased surfactant lipids correlate with lung function in chronic obstructive pulmonary disease (COPD). PLoS ONE. 2020;15(2): e0228279.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hannun YA, Obeid LM. Principles of bioactive lipid signalling: lessons from sphingolipids. Nat Rev Mol Cell Biol. 2008;9(2):139–50.

    Article  CAS  PubMed  Google Scholar 

  103. Kim J, Suresh B, Lim MN, Hong SH, Kim KS, Song HE, et al. Metabolomics reveals dysregulated sphingolipid and amino acid metabolism associated with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2022;17:2343–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Yang Y, Uhlig S. The role of sphingolipids in respiratory disease. Ther Adv Respir Dis. 2011;5(5):325–44.

    Article  CAS  PubMed  Google Scholar 

  105. Bowler RP, Jacobson S, Cruickshank C, Hughes GJ, Siska C, Ory DS, et al. Plasma sphingolipids associated with chronic obstructive pulmonary disease phenotypes. Am J Respir Crit Care Med. 2015;191(3):275–84.

    Article  PubMed  PubMed Central  Google Scholar 

  106. McCann MR, George De la Rosa MV, Rosania GR, Stringer KA. l-Carnitine and acylcarnitines: mitochondrial biomarkers for precision medicine. Metabolites. 2021;11(1):51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Gillenwater LA, Kechris KJ, Pratte KA, Reisdorph N, Petrache I, Labaki WW, et al. Metabolomic profiling reveals sex specific associations with chronic obstructive pulmonary disease and emphysema. Metabolites. 2021;11(3):161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Callejón-Leblic B, Pereira-Vega A, Vázquez-Gandullo E, Sánchez-Ramos JL, Gómez-Ariza JL, García-Barrera T. Study of the metabolomic relationship between lung cancer and chronic obstructive pulmonary disease based on direct infusion mass spectrometry. Biochimie. 2019;157:111–22.

    Article  PubMed  Google Scholar 

  109. Kim DJ, Oh JY, Rhee CK, Park SJ, Shim JJ, Cho JY. Metabolic fingerprinting uncovers the distinction between the phenotypes of tuberculosis associated COPD and smoking-induced COPD. Front Med. 2021;8: 619077.

    Article  Google Scholar 

  110. Naz S, Kolmert J, Yang M, Reinke SN, Kamleh MA, Snowden S, et al. Metabolomics analysis identifies sex-associated metabotypes of oxidative stress and the autotaxin-lysoPA axis in COPD. Eur Respir J. 2017;49(6):1602322.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Yu B, Flexeder C, McGarrah RW 3rd, Wyss A, Morrison AC, North KE, et al. Metabolomics identifies novel blood biomarkers of pulmonary function and COPD in the general population. Metabolites. 2019;9(4):61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Lommatzsch M, Cicko S, Müller T, Lucattelli M, Bratke K, Stoll P, et al. Extracellular adenosine triphosphate and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010;181(9):928–34.

    Article  CAS  PubMed  Google Scholar 

  113. Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, Muskens F, et al. Extracellular ATP triggers and maintains asthmatic airway inflammation by activating dendritic cells. Nat Med. 2007;13(8):913–9.

    Article  CAS  PubMed  Google Scholar 

  114. Hunninghake GM, Cho MH, Tesfaigzi Y, Soto-Quiros ME, Avila L, Lasky-Su J, et al. MMP12, lung function, and COPD in high-risk populations. N Engl J Med. 2009;361(27):2599–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Reinke SN, Naz S, Chaleckis R, Gallart-Ayala H, Kolmert J, Kermani NZ, et al. Urinary metabotype of severe asthma evidences decreased carnitine metabolism independent of oral corticosteroid treatment in the U-BIOPRED study. Eur Respir J. 2022;59(6):2101733.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Terracciano R, Pelaia G, Preianò M, Savino R. Asthma and COPD proteomics: current approaches and future directions. Proteom Clin Appl. 2015;9(1–2):203–20.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

S.C. is supported by a fellowship from the Ph.D. Program in Life Sciences (XXXVI cycle), University “Magna Græcia” of Catanzaro, Italy.

Funding

This research received funding from Department of Medical and Surgical Sciences, Magna Græcia University, 88100 Catanzaro, Italy.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: RS, RT. Investigation: SC, FG, DS, ADA. Statistical analysis and interpretation: RT, RS, SC, MP, FG, DS, ADA, GP,CP. Writing original draft preparation: SC, FG, DS, CP, RT. Writing—review and editing: MP, RS, ADA, RT. Resources: CP, GP,RS, ADA, RT. Supervision: RS, ADA, RT. All authors contributed to the article and approved the submitted version.

Corresponding authors

Correspondence to Serena Correnti or Rosa Terracciano.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the Ethics Committee of MAGNA GRAECIA UNIVERSITY and MATER DOMINI HOSPITAL (Approval number 219, date of approval:16 November 2016). All patients gave written informed consent prior to their participation in the study, in accordance with the tenets of the Declaration of Helsinki.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

The original version of this article was revised: Correction in marking one of the corresponding authors. It is now: Rosa Terracciano4; It should be: Rosa Terracciano4*.

Supplementary Information

Additional file1: Table S1.

Spearman’s correlation analysis between compounds and lung functions.

Additional file2: Figure S1.

Heatmap illustrating the relative abundance levels of all the metabolites and lipids in asthmatic compared to COPD patients. Red indicates an increase in abundance, while blue a decrease. The columns and rows in the heatmap correspond to the experimental samples and the compounds, respectively.

Additional file3: Table S2.

Peak intensities values of metabolites and lipids in all the asthmatic and COPD patients analyzed in this study.

Rights and permissions

Open Access This 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.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Correnti, S., Preianò, M., Gamboni, F. et al. An integrated metabo-lipidomics profile of induced sputum for the identification of novel biomarkers in the differential diagnosis of asthma and COPD. J Transl Med 22, 301 (2024). https://doi.org/10.1186/s12967-024-05100-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12967-024-05100-2

Keywords