Role of Lipidomics in Respiratory Tract Infections: A Systematic Review of Emerging Evidence
Abstract
1. Introduction
2. Methods
2.1. Search Strategy
2.2. Inclusion and Exclusion Criteria
2.3. PRISMA Process
2.4. Quality Assessment
2.5. Data Extraction
2.6. Data Synthesis
3. Results
3.1. Study Characteristics and Analytical Platforms
3.2. Identified Lipid Biomarkers in LRTIs
3.3. Role of Lipidomics in Pathophysiological Insights
3.4. Lipidomic Alterations by Pathogen Type
3.5. Temporal Dynamics and Prognostic Implications
3.6. Integrative Models and Clinical Translation
4. Discussion
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Georgakopoulou, V.E. Insights from respiratory virus co-infections. World J. Virol. 2024, 13, 98600. [Google Scholar] [CrossRef] [PubMed]
- Georgakopoulou, V.E.; Lempesis, I.G.; Tarantinos, K.; Sklapani, P.; Trakas, N.; Spandidos, D.A. Atypical pneumonia (Review). Exp. Ther. Med. 2024, 28, 424. [Google Scholar] [CrossRef]
- Hanage, W.P.; Schaffner, W. Burden of Acute Respiratory Infections Caused by Influenza Virus, Respiratory Syncytial Virus, and SARS-CoV-2 with Consideration of Older Adults: A Narrative Review. Infect. Dis. Ther. 2025, 14 (Suppl. S1), 5–37. [Google Scholar] [CrossRef]
- Liapikou, A.; Torres, A. The clinical management of lower respiratory tract infections. Expert Rev. Respir. Med. 2016, 10, 441–452. [Google Scholar] [CrossRef] [PubMed]
- Lim, W.S. Pneumonia—Overview. Encycl. Respir. Med. 2022, 185–197. [Google Scholar] [CrossRef]
- Wenk, M.R. Lipidomics of host-pathogen interactions. FEBS Lett. 2006, 580, 5541–5551. [Google Scholar] [CrossRef]
- Han, X.; Gross, R.W. Shotgun lipidomics: Multidimensional MS analysis of cellular lipidomes. Expert Rev. Proteom. 2005, 2, 253–264. [Google Scholar] [CrossRef]
- Anh, N.K.; Thu, N.Q.; Tien, N.T.N.; Long, N.P.; Nguyen, H.T. Advancements in Mass Spectrometry-Based Targeted Metabolomics and Lipidomics: Implications for Clinical Research. Molecules 2024, 29, 5934. [Google Scholar] [CrossRef]
- Cajka, T.; Fiehn, O. Comprehensive analysis of lipids in biological systems by liquid chromatography-mass spectrometry. TrAC Trends Anal. Chem. 2014, 61, 192–206. [Google Scholar] [CrossRef]
- Wu, Z.; Shon, J.C.; Liu, K.H. Mass Spectrometry-based Lipidomics and Its Application to Biomedical Research. J. Lifestyle Med. 2014, 4, 17–33. [Google Scholar] [CrossRef] [PubMed]
- Schenck, E.J.; Plataki, M.; Wheelock, C.E. A Lipid Map for Community-acquired Pneumonia with Sepsis: Observation Is the First Step in Scientific Progress. Am. J. Respir. Crit. Care Med. 2024, 209, 903–904. [Google Scholar] [CrossRef]
- Rischke, S.; Gurke, R.; Zielbauer, A.S.; Ziegler, N.; Hahnefeld, L.; Köhm, M.; Kannt, A.; Vehreschild, M.J.; Geisslinger, G.; Rohde, G.; et al. Proteomic, metabolomic and lipidomic profiles in community acquired pneumonia for differentiating viral and bacterial infections. Sci. Rep. 2025, 15, 1922. [Google Scholar] [CrossRef]
- Chen, L.; Zheng, Y.; Zhao, L.; Zhang, Y.; Yin, L.; He, Y.; Ma, X.; Xu, Y.; Gao, Z. Lipid profiles and differential lipids in serum related to severity of community-acquired pneumonia: A pilot study. PLoS ONE 2021, 16, e0245770. [Google Scholar] [CrossRef]
- Shan, J.; Qian, W.; Shen, C.; Lin, L.; Xie, T.; Peng, L.; Xu, J.; Yang, R.; Ji, J.; Zhao, X. High-resolution lipidomics reveals dysregulation of lipid metabolism in respiratory syncytial virus pneumonia mice. RSC Adv. 2018, 8, 29368–29377. [Google Scholar] [CrossRef]
- Virgiliou, C.; Begou, O.; Ftergioti, A.; Simitsopoulou, M.; Sdougka, M.; Roilides, E.; Theodoridis, G.; Gika, H.; Iosifidis, E. Untargeted Blood Lipidomics Analysis in Critically Ill Pediatric Patients with Ventilator-Associated Pneumonia: A Pilot Study. Metabolites 2024, 14, 466. [Google Scholar] [CrossRef]
- Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
- Wells, G.A.; Shea, B.; O’Connell, D.; Peterson, J.; Welch, V.; Losos, M.; Tugwell, P. The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. Available online: https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp (accessed on 18 September 2025).
- Castañé, H.; Iftimie, S.; Baiges-Gaya, G.; Rodríguez-Tomàs, E.; Jiménez-Franco, A.; López-Azcona, A.F.; Garrido, P.; Castro, A.; Camps, J.; Joven, J. Machine learning and semi-targeted lipidomics identify distinct serum lipid signatures in hospitalized COVID-19-positive and COVID-19-negative patients. Metabolism 2022, 131, 155197. [Google Scholar] [CrossRef]
- Chouchane, O.; Schuurman, A.R.; Reijnders, T.D.Y.; Peters-Sengers, H.; Butler, J.M.; Uhel, F.; Schultz, M.J.; Bonten, M.J.; Cremer, O.L.; Calfee, C.S.; et al. The Plasma Lipidomic Landscape in Patients with Sepsis due to Community-acquired Pneumonia. Am. J. Respir. Crit. Care Med. 2024, 209, 973–986. [Google Scholar] [CrossRef] [PubMed]
- Saballs, M.; Parra, S.; Martínez, N.; Amigo, N.; Cabau, L.; Iftimie, S.; Pavon, R.; Gabaldó, X.; Correig, X.; Paredes, S.; et al. Lipidomic and metabolomic changes in community-acquired and COVID-19 pneumonia. J. Lipid Res. 2024, 65, 100622. [Google Scholar] [CrossRef] [PubMed]
- Kassa-Sombo, A.; Verney, C.; Pasquet, A.; Vaidie, J.; Brea, D.; Vasseur, V.; Cezard, A.; Lefevre, A.; David, C.; Piver, E.; et al. Lipidomic signatures of ventilator-associated pneumonia in COVID-19 ARDS patients: A new frontier for diagnostic biomarkers. Ann. Intensive Care 2025, 15, 78. [Google Scholar] [CrossRef]
- Ma, X.; Chen, L.; He, Y.; Zhao, L.; Yu, W.; Xie, Y.; Yu, Y.; Xu, Y.; Zheng, Y.; Li, R.; et al. Targeted lipidomics reveals phospholipids and lysophospholipids as biomarkers for evaluating community-acquired pneumonia. Ann. Transl. Med. 2022, 10, 395. [Google Scholar] [CrossRef]
- Schuurman, A.R.; Chouchane, O.; Butler, J.M.; Peters-Sengers, H.; Joosten, S.; Brands, X.; Haak, B.W.; Otto, N.A.; Uhel, F.; Klarenbeek, A.; et al. The shifting lipidomic landscape of blood monocytes and neutrophils during pneumonia. JCI Insight 2024, 9, e164400. [Google Scholar] [CrossRef]
- Humes, S.T.; Iovine, N.; Prins, C.; Garrett, T.J.; Lednicky, J.A.; Coker, E.S.; Sabo-Attwood, T. Association between lipid profiles and viral respiratory infections in human sputum samples. Respir. Res. 2022, 23, 177. [Google Scholar] [CrossRef]
- Zheng, Y.; Ning, P.; Luo, Q.; He, Y.; Yu, X.; Liu, X.; Chen, Y.; Wang, X.; Kang, Y.; Gao, Z. Inflammatory responses relate to distinct bronchoalveolar lavage lipidome in community-acquired pneumonia patients: A pilot study. Respir. Res. 2019, 20, 82. [Google Scholar] [CrossRef]
- Sun, G.; Wang, Q.; Shan, X.; Kuerbanjiang, M.; Ma, R.; Zhou, W.; Sun, L.; Li, Q. Metabolomics and lipidomics of plasma biomarkers for tuberculosis diagnostics using UHPLC-HRMS. Front. Cell. Infect. Microbiol. 2025, 15, 1526740. [Google Scholar] [CrossRef]
- Zhang, C.; Cai, M.; Cai, H.; Chen, X. Plasma lipidomic analysis reveals distinct lipid alterations in patients with pulmonary tuberculosis. Eur. J. Med. Res. 2025, 30, 566. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Yu, Q.; Wei, H.; Hu, X.; Wang, H.; Wei, M.; Xu, J.; Zhao, J. Integration of lipidomics and metabolomics reveals plasma and urinary profiles in children with Mycoplasma pneumoniae pneumonia. Biomed. Chromatogr. 2023, 37, e5702. [Google Scholar] [CrossRef]
- Tomalka, J.A.; Owings, A.; Galeas-Pena, M.; Ziegler, C.G.K.; Robinson, T.O.; Wichman, T.G.; Laird, H.; Williams, H.B.; Ghaliwal, N.S.; Everman, S.; et al. Enhanced production of lipid mediators in plasma and activation of DNA damage pathways in PBMCs are correlated with the severity of ancestral SARS-CoV-2 infection. FASEB J. 2025, 39, e70600. [Google Scholar] [CrossRef] [PubMed]
- de Almeida, R.A.M.; Souza, L.A.; de Oliveira, M.A.; do Carmo, A.P.; Oliveira, L.L.; Arêas, G.P.T.; Ferreira, C.R.; Catharino, R.R. Diagnostic screening of COVID-19 based on multiomics data by high-resolution mass spectrometry. Sci. Rep. 2025, 15, 11849. [Google Scholar] [CrossRef]
- Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Alexiou, A.; Papadakis, M.; Batiha, G.E. Receptor-Dependent Effects of Sphingosine-1-Phosphate (S1P) in COVID-19: The Black Side of the Moon. Mol. Cell. Biochem. 2023, 478, 2271–2279. [Google Scholar] [CrossRef]
- Uchida, T.; Shirasawa, M.; Ware, L.B.; Kojima, K.; Hata, Y.; Makita, K.; Mednick, G.; Matthay, M.A. Receptor for Advanced Glycation End-Products Is a Marker of Type I Cell Injury in Acute Lung Injury. Am. J. Respir. Crit. Care Med. 2006, 173, 1008–1015. [Google Scholar] [CrossRef] [PubMed]
- Ware, L.B.; Koyama, T.; Zhao, Z.; Janz, D.R.; Wickersham, N.; Bernard, G.R.; Matthay, M.A. Biomarkers of Lung Epithelial Injury and Inflammation Distinguish Severe Sepsis Patients with Acute Respiratory Distress Syndrome. Crit. Care 2013, 17, R253. [Google Scholar] [CrossRef] [PubMed]
- Ishikawa, N.; Hattori, N.; Yokoyama, A.; Kohno, N. Utility of KL-6/MUC1 in the Clinical Management of Interstitial Lung Diseases. Respir. Investig. 2012, 50, 3–13. [Google Scholar] [CrossRef]
- de Souza Xavier Costa, N.; da Costa Sigrist, G.; Schalch, A.S.; Belotti, L.; Dolhnikoff, M.; da Silva, L.F.F. Lung Tissue Expression of Epithelial Injury Markers Is Associated with Acute Lung Injury Severity but Does Not Discriminate Sepsis from ARDS. Respir. Res. 2024, 25, 129. [Google Scholar] [CrossRef]
- Dennis, E.A.; Cao, J.; Hsu, Y.H.; Magrioti, V.; Kokotos, G. Phospholipase A2 Enzymes: Physical Structure, Biological Function, Disease Implication, Chemical Inhibition, and Therapeutic Intervention. Chem. Rev. 2011, 111, 6130–6185. [Google Scholar] [CrossRef]
- Petrache, I.; Natarajan, V.; Zhen, L.; Medler, T.R.; Richter, A.T.; Cho, C.; Hubbard, W.C.; Berdyshev, E.V. Ceramide Upregulation Causes Pulmonary Cell Apoptosis and Emphysema-Like Disease in Mice. Nat. Med. 2005, 11, 491–498. [Google Scholar] [CrossRef] [PubMed]
First Author | Year | Country | Study Design | Population | Sample Type | Analytical Platform | Lipid Classes Analyzed | Main Findings | NOS |
---|---|---|---|---|---|---|---|---|---|
Castané [18] | 2022 | Spain | Retrospective post hoc cohort study | 126 hospitalized COVID-19 patients, 45 hospitalized COVID-19-negative patients (infectious/inflammatory), 50 healthy controls | Serum | Semi-targeted lipidomics by UHPLC-QTOF-MS, machine learning (Monte Carlo, PCA, PLS-DA) | Acylcarnitines, lysophospholipids (LPC, LPE), phosphatidylcholines (PC), oxylipins (e.g., 9/13-HODE, 15-HETE), bile acids, long-chain TGs | COVID-19 and non-COVID inflammatory patients shared a common lipid signature characterized by elevated acylcarnitines and LPE, and decreased 9/13-HODE and 15-HETE. However, specific discrimination between COVID-19 and other conditions was achieved via decreased LPC(22:6-sn2), increased PC(36:1), and changes in secondary bile acids. Arachidonic acid levels were markedly decreased in both COVID-19 and other infectious groups. Machine learning identified oxylipins, carnitines, and phospholipids as key discriminatory features. Alterations in β-oxidation and fatty acid metabolism were prominent. The ratio of LPC22:6-sn2/PC36:1 achieved an AUC > 0.95. No association was found with ICU admission or mortality. | 8/9 |
Chen [13] | 2021 | China | Cross-sectional pilot study | 28 CAP patients (13 severe, 15 non-severe), 20 matched non-CAP controls | Serum | Untargeted UHPLC-MS/MS lipidomics, OPLS-DA, ROC, multivariate regression | Glycerophospholipids (PC, PE), sphingolipids (SM, HexCer), lysophospholipids (LPC, LPE), diacylglycerols, cholesterol esters, free fatty acids | Lipid profiles differed significantly across NC, NSCAP, and SCAP groups. CAP patients exhibited reduced LPC and PE levels, increased Hex2Cer and cholesterol esters. Four lipids (PC[16:0_18:1], PC[18:2_20:4], PC[36:4], PC[38:6]) outperformed CURB-65 and PSI in ROC analysis for disease severity. PC(18:2_20:4) had AUC 0.954, PC(38:6) had AUC 0.959. PC(18:2_20:4) and PC(36:4) correlated negatively with FiO2 and PCT; PC(16:0_18:1) positively with PCT. Lower PC levels were linked to longer hospital stay and higher 30-day mortality. Combined phospholipid biomarkers showed potential for disease monitoring, diagnosis, and prognosis in CAP. | 7/9 |
Chouchane [19] | 2024 | Netherlands | Prospective cohort study | 169 ICU patients with CAP sepsis, 51 noninfected ICU controls, 48 outpatient controls; plus two validation cohorts (CAPSOD, EARLI) | Plasma | Untargeted lipidomics via HPLC-MS (1833 lipid species across 33 classes), data validated in external cohorts | Cholesterol esters, triacylglycerols, phospholipids (PC, PE, LPC), sphingomyelins, ceramides, sulfatides, plasmalogens, lysolipids | Patients with sepsis due to CAP exhibited a profound shift in the plasma lipidome compared to both healthy and noninfected ICU controls. 58% of lipid species were decreased, while 6% increased. Cholesterol esters and lysophospholipids showed strong inverse associations with SOFA score and systemic inflammation. Recovery of specific lipids such as cholesterol esters by Day 4 in ICU was linked to lower 30-day mortality (e.g., OR = 0.84 per 10% increase). The lipidomic profile showed partial recovery over time, and a specific TG-rich pattern distinguished CAP from other ICU patients. LPC and Chol-E emerged as key prognostic lipids. Lipid class patterns were validated across CAPSOD and EARLI cohorts. Results support lipidomics as a biomarker and prognostic tool in CAP-associated sepsis. | 9/9 |
Rischke [12] | 2025 | Germany | Prospective cohort study | 69 patients with community-acquired pneumonia (CAP): 43 viral (incl. COVID-19), 26 bacterial | Plasma (baseline, day 3, day 7) | LC-MS/MS (MxP Quant 500), Olink proteomics, lipid network enrichment (LINEX2), PLS-DA, PCA | Phosphatidylcholines, ether-PCs, lysophosphatidylcholines (LPC), triglycerides, diglycerides, bile acids (GCA, TCA, TCDCA), oxylipins (EpOME, DiHOME) | Distinct plasma lipidomic profiles differentiated viral from bacterial CAP. Bacterial CAP was characterized by elevated PCs and PC-ethers, while viral CAP showed increased TGs, DGs (especially FA 18:2-containing), and linoleic acid–derived oxylipins (12,13-EpOME, 9,10-DiHOME). Proteomic markers like TRAIL, LAG-3, and LAMP3 were elevated in viral CAP, while CLEC4D and EN-RAGE were elevated in bacterial CAP. Integrated clustering of lipidomic, metabolomic, and proteomic analytes supported co-expression of pathogen-specific patterns. PLS-DA and hierarchical clustering identified robust discriminatory features. Findings indicate the potential of lipidomics and multi-omics in pathogen-specific diagnosis and individualized treatment strategies in CAP. | 8/9 |
Saballs [20] | 2024 | Spain | Prospective observational study | 71 CAP patients, 75 COVID-19 pneumonia patients, 75 healthy controls (age- and sex-matched) | Serum | 1H NMR spectroscopy (Liposcale® assay), BUME extraction, LipSpin, multivariate analysis (PLS-DA, random forest, ROC) | Phosphatidylcholine, lysophosphatidylcholine, PUFA, DHA, esterified/free cholesterol, triglycerides, HDL/LDL/VLDL subclasses, glycoproteins | Both CAP and COVID-19 pneumonia patients exhibited hypolipidemia with reduced levels of HDL-c, phosphatidylcholine, lysophosphatidylcholine, PUFA, and DHA. Severity was associated with increased VLDL-c, IDL-c, LDL-tg/LDL-c, triglycerides, and glycoproteins (GlycA, GlycB, GlycF), along with decreased HDL particles and esterified cholesterol. COVID-19 patients showed more pronounced alterations. A lipidomic-metabolomic model based on PC, glycerophospholipids, creatine, glutamate, isoleucine, alanine, and glycoproteins achieved AUC = 0.935 for etiology classification and 0.931 for severity. Metabolites linked to inflammation and energy metabolism (lactate, glucose, creatine, BCAAs, glutamate) were elevated. These alterations reflect lipid-mediated immune modulation, metabolic reprogramming, and potential for diagnostic/prognostic biomarker development. | 7/9 |
Kassa-Sombo [21] | 2025 | France | Prospective observational cohort study | 39 patients with COVID-19 ARDS (26 with VAP, 13 without VAP), matched controls | Tracheal aspirate | Untargeted lipidomics via UHPLC-HRMS (Q-Exactive MS), data processed with Workflow4Metabolomics/XCMS | Phosphatidylcholines, phosphatidylethanolamines, sphingomyelins, ether-linked PCs and PEs, lysophospholipids | Significant alterations in the tracheal aspirate lipidome were observed in VAP versus non-VAP COVID-19 ARDS patients (p = 0.003). Among 272 identified lipids, PCs were most frequently dysregulated, with 17 upregulated and 6 downregulated. SM(34:1) and PC(O-34:1) were the most predictive biomarkers for VAP, showing AUROC of 0.85 and 0.83, respectively. Eight key lipids were identified via multivariate analyses (PCA, PLS-DA, OPLS-DA), all upregulated in VAP. Combined lipid biomarkers modestly improved diagnostic performance (AUROC up to 0.86). These results suggest that lipidomics of tracheal aspirates offers potential for accurate VAP diagnostics and highlights lipid remodeling at the site of infection. | 8/9 |
Ma [22] | 2022 | China | Prospective multi-center cohort study | 58 patients with CAP (30 NSCAP, 28 SCAP), 11 healthy controls | Serum | Targeted LC-MS/MS, qRT-PCR validation, GEO database transcriptome analysis | Phosphatidylcholine (PC), lysophosphatidylcholine (LPC), phosphatidylethanolamine (PE), lysophosphatidylethanolamine (LPE) | LPC levels were decreased, while PE, PC, PC/LPC, and PE/LPE ratios were increased in CAP. PE combined with CURB-65 predicted severity (AUC = 0.848), while PC/LPC ratio improved 30-day mortality prediction (AUC = 0.838). PC(36:4) and LPC(18:2/0:0) emerged as species-level biomarkers. Expression of LPCAT2 was upregulated and LPCAT1 downregulated in SCAP; LPCAT2 positively correlated with inflammatory genes, LPCAT1 negatively. Lipid ratios (PC/LPC, PE/LPE) and PE were positively correlated with CRP, neutrophil percentage, and PSI, and negatively with albumin and lymphocytes. Alterations in Lands cycle enzymes support metabolic dysregulation during infection. | 8/9 |
Schuurman [23] | 2024 | Netherlands | Case–control cohort study | 48 CAP patients (baseline and 1-month follow-up), 25 matched noninfectious controls | Isolated blood monocytes and neutrophils | Untargeted HPLC-MS lipidomics with transcriptomics integration | Sphingolipids (SM, ceramides, S1P), phospholipids (PC, PE), lysophospholipids (LPC, LPE), fatty acids, diacylglycerols (DG), triglycerides (TG), BMPs | Pneumonia significantly altered the lipidomic landscape of monocytes and neutrophils, with distinct profiles. Monocyte lipid changes were mostly decreases in PC, PE, and SM species and resolved after 1 month. In contrast, neutrophils showed persistent changes with increased PC, PE, DG, BMP, and polyunsaturated TGs. Sphingolipid metabolism, particularly S1P signaling, was upregulated. Transcriptomic analysis confirmed upregulation of key enzymes (SPHK1, UGCG, SMPD1). Functional validation showed that inhibiting SPT and SPHK1 blunted cytokine production in both cell types. The study demonstrated a mechanistic link between altered lipid profiles and immune function during CAP. | 9/9 |
Virgiliou [15] | 2024 | Greece | Prospective pilot cohort study | 20 critically ill pediatric patients (12 high VAP suspicion, 8 low suspicion) | Plasma (blood samples at 4 timepoints: Days 1, 3, 6, 12) | Untargeted LC-HRMS (UPLC-TIMS-TOF/MS), multivariate + univariate analysis, MS-DIAL, Lipostar2 | Phosphatidylcholines (PC), lysophosphatidylcholines (LPC), sphingomyelins (SM), triglycerides (TG), diglycerides (DG), cholesterol esters (CE), carnitines | Untargeted lipidomics revealed 144 blood lipid species in critically ill pediatric patients with VAP suspicion. PCs, SMs, TGs, and DGs were significantly altered between high and low mCPIS groups. High suspicion group showed increased levels of PCs and TGs over time. Discriminatory lipids (e.g., PC 32:2, TG 48:3, SM 40:1) had AUC > 0.75. Specific lipid profiles were associated with culture-confirmed pathogens, including S. aureus and K. pneumoniae. Multivariate models (OPLS-DA) distinguished both VAP severity and pathogen type. Phospholipid shifts correlated with inflammatory markers and may indicate pathogen-specific metabolic remodeling. The study supports lipidomics as a promising diagnostic tool for early VAP identification and microbial stratification in pediatric ICU settings. | 7/9 |
Humes [24] | 2022 | USA | Cross-sectional study | 30 patients with confirmed viral respiratory infections: Influenza A H1-2009 (n = 9), Influenza A H3 (n = 11), Rhinovirus (n = 10) | Induced sputum | Untargeted LC–HRMS (Thermo Q-Exactive Orbitrap, UHPLC), Bayesian profile regression, multinomial logistic regression | >600 lipid species across 27 lipid classes (positive mode) and 23 classes (negative mode). Most abundant: TG, PE, PC, SM, ether-PC, ether-PE. Differential lipids included Acylcarnitines (10:1, 16:1, 18:2), DGs (16:0_18:0, 18:0_18:0), LPC (12:0, 20:5), PE (18:0_18:0), TGs (various species). | Three lipid clusters were identified. Cluster 1 (high TG/DG) associated with Influenza A H3, while Cluster 3 (low lipid levels) associated with Rhinovirus. Odds of H3 infection were 15× higher vs. Rhinovirus in lipid-rich cluster (OR 15.0, 95% CI 1.03–218.3; p = 0.047). Distinct lipid signatures suggest pathogen-specific metabolic remodeling. Findings support sputum lipidomics as a tool for differentiating viral respiratory infections. | 7/9 |
Zheng [25] | 2019 | China | Multicenter observational cohort (pilot) | 52 patients with community-acquired pneumonia (CAP); 68 controls (35 healthy, 33 with non-infectious lung disease); bronchoscopy performed within 72 h of admission; 30-day follow-up | BALF | Untargeted lipidomics via HPLC-MS (UHPLC-Q Exactive Orbitrap), PCA, k-means clustering, correlation and ROC analyses | Fatty acids (SFA, MUFA, PUFA), acylcarnitines, sphingolipids (SM, GM3, LacCer, sphingosine), neutral lipids (TG, DG), phospholipids (PC, PE, PI, PG, PS), lysolipids (LPC, LPE, LPG) | CAP patients showed markedly increased lactosylceramides (>10-fold), MUFA/PUFA, PE, and DG (~2-fold), with decreased sphingosine, PG, and lysolipids (LPC, LPE, LPG). Lipid clusters correlated with inflammation and severity: SM(d34:1) inversely with macrophages (adj r = −0.462), PE(18:1p/20:4) positively with PMNs (adj r = 0.541). Certain fatty acids discriminated severe CAP from non-severe (AUC ~0.74–0.76). Findings support BALF lipidomics as a marker of CAP severity and immune response. | 8/9 |
Sun [26] | 2025 | China | Case–control study with training (72 TB vs. 78 HCs) and validation set (30 TB vs. 30 HCs) | 102 active TB patients, 108 healthy controls | Plasma | Untargeted metabolomics and lipidomics by UHPLC-HRMS; machine learning (LASSO, Random Forest, XGBoost) | Broad plasma metabolites including lipids (bile acids, amino acids, phospholipids, oxylipins) | Identified 282 differential metabolites in training set, 214 validated. KEGG enrichment: lipid metabolism pathways. Seven core metabolites (Angiotensin IV, glycochenodeoxycholic acid, methyl indole-3-acetate, dulcitol, Asp-Phe, benzamide, carbadox). Angiotensin IV had very high diagnostic accuracy (AUC = 0.999 training; 0.991 validation; sensitivity 98.6–100%, specificity 96.7–100%). Lipid dysregulation (bile acid metabolism, fatty acid metabolism) central to TB profile. | 8/9 |
Zhang [27] | 2025 | China | Case–control study | 50 newly diagnosed pulmonary TB patients vs. 50 age- and sex-matched healthy controls | Plasma | LC-MS/MS lipidomics (untargeted) | TAGs, ceramides (CER), hexosylceramides (HCER), phosphatidylethanolamines (PE), phosphatidylcholines (PC), total cholesterol, triglycerides, HDL, LDL | PTB patients had significantly lower total cholesterol, triglycerides, HDL, LDL, TAG, CER, and HCER, while PE and PC were significantly elevated (p < 0.05). From 633 detected lipids, 61 were differentially expressed. Three lipid species (CER(24:0) H, HCER(d18:0/22:0) H, and PE(18:1/18:1)) demonstrated strong diagnostic potential (AUC > 0.75). Lipid alterations correlated weakly/moderately with age and glucose but not with BMI, sex, or | 8/9 |
Chen [28] | 2023 | China | Prospective observational study | 30 children with Mycoplasma pneumoniae infections (17 bronchitis, 13 pneumonia) and 35 healthy controls | Plasma, urine | Untargeted lipidomics (UHPLC-LTQ-Orbitrap XL MS), metabolomics (GC–MS) | Lysophosphatidylethanolamines (LPE), lysophosphatidylcholines (LPC), phosphatidylcholines (PC), phosphatidylethanolamines (PE), phosphatidylinositols (PI), triglycerides (TG) | Identified 14 differential lipids and >50 plasma/urine metabolites associated with infection. Perturbations in lysoPE, LPC, PI, and TG reflected membrane damage and altered energy metabolism. Pathways affected included amino acid, nucleotide, and energy metabolism. A 13-metabolite plasma panel (e.g., L-hydroxyproline, creatine, inosine, citric acid, cystine, glycine) discriminated bronchitis vs. pneumonia with AUC = 0.927 and infection vs. healthy with AUC = 1. Several metabolites correlated with neutrophil/lymphocyte ratios and disease severity. | 8/9 |
Tomalka [29] | 2025 | USA | Case–control, multi-omics study | 10 hospitalized COVID-19 patients, 10 matched healthy controls; extended analysis on 58 NP swab samples (ancestral, Delta, Omicron) | Plasma, PBMCs, nasopharyngeal swabs | GC–MS fatty acid panel, bulk RNA-seq (PBMCs), scRNA-seq (nasopharyngeal) | Fatty acids and eicosanoid precursors (AA, EPA, DHA, DPA, linoleic acid, dihomo-γ-linolenic acid), pro-resolving mediators (lipoxins, resolvins) | COVID-19 patients showed significantly increased AA, EPA, DHA, and DPA, all correlating with WHO disease severity (rho 0.70–0.74). Elevated eicosanoid precursors were linked to severe disease (WHO score > 5). Multi-omics revealed positive correlations of inflammatory lipids (notably EPA) with cell cycle progression, DNA damage, and ER stress pathways in PBMCs. Severe cases had reduced innate immune and interferon signaling. NP swabs confirmed upregulation of lipid mediator synthesis genes (ALOX5, ALOX15, PLA2G4A), especially in goblet and ciliated cells, across SARS-CoV-2 variants. Findings implicate lipid mediators in driving both inflammation and resolution, with potential prognostic/therapeutic value. | 8/9 |
de Almeida [30] | 2025 | Brazil | Case–control, multiomics diagnostic study | 239 serum samples (119 COVID-19 positive, 120 negative by ELISA); additional proteomic analysis on 300 samples (150 positive, 150 negative) | Serum | Direct infusion ESI(±)-Orbitrap MS for lipidomics; MALDI(+)-TOF MS for proteomics; machine learning (PCA, PLS-DA, SVM) | Glycerophospholipids (PC, PE, PS, PI, PG, PA, LPC, LPG, PIP), glycerolipids (DG, MG, TG), sphingolipids (SM), sterol lipids (Chol, CE), fatty acyls | Identified 16 key lipids in negative-ion mode (e.g., PS, sterols, fatty acids) and 18 in positive-ion mode (e.g., LPC, PC, SM, DG). COVID-19 positive patients showed enrichment in PS, PI, and PA, while controls showed higher PCs. SVM models achieved 96.7–100% sensitivity and 82–97% specificity, with accuracies up to 98.9%. Proteomic MALDI-TOF profiles also achieved ~99% accuracy. Demonstrated lipidomic/proteomic spectral signatures as powerful high-throughput COVID-19 diagnostics with near-zero false negatives. | 8/9 |
Lipid Biomarker (or Ratio) | Putative Biological Function in LRTIs | Diagnostic/Prognostic Performance (Study) |
---|---|---|
PC(18:2_20:4), PC(36:4), PC(38:6) | Structural phosphatidylcholines; membrane fluidity; inflammatory remodeling | Lower in severe CAP; correlated with FiO2 (–) and PCT; predicted severity better than CURB-65/PSI (AUC 0.954–0.959); lower levels linked to prolonged hospitalization and higher 30-day mortality [13] |
PC/LPC and PE/LPE ratios | Reflect Lands’ cycle activity (LPCAT1/2); membrane repair and inflammatory remodeling | Higher in CAP; correlated with CRP, neutrophils, PSI; PC/LPC ratio predicted 30-day mortality (AUC 0.838) and improved CURB-65 for severity [22] |
LPC(22:6-sn2)/PC(36:1) | Resolution lipid vs. structural PC; inflammatory discrimination | Discriminated COVID-19 from other infectious/inflammatory admissions (AUC > 0.95) [18] |
SM(34:1) and PC(O-34:1) | Sphingomyelin and ether-PC; markers of epithelial/surfactant injury | Best tracheal aspirate predictors of VAP in COVID-19 ARDS (AUROC 0.85 and 0.83), outperforming CRP/PCT [21] |
Cholesteryl esters (CE) | Lipid transport; immune/metabolic recovery | Day-4 rise in CE associated with lower 30-day mortality in CAP sepsis (OR 0.84 per 10% increase) [19] |
Oxylipins (12,13-EpOME; 9,10-DiHOME) | Linoleic acid-derived mediators; vascular and leukocyte signaling | Enriched in viral vs. bacterial CAP; contributed to pathogen-specific stratification [12] |
Lactosylceramides (LacCer) | Sphingolipid signaling; immune activation | >10-fold increase in BALF in CAP; tracked inflammatory cell patterns and severity [25] |
Acylcarnitines | Indicators of mitochondrial β-oxidation dysfunction and energy stress | Elevated in COVID-19 and infectious controls; discriminatory in machine-learning models [18,24] |
S1P axis (increase in SPHK1) | Neutrophil/monocyte activation; cytokine production | Immune-cell lipidomics showed SPHK1-driven S1P signaling; inhibition reduced cytokine release [23] |
NMR glycoproteins (GlycA/B/F) | Markers of systemic inflammation | Higher with pneumonia severity; included in high-AUC etiology/severity models [20] |
AA, EPA, DHA, DPA (eicosanoid precursors) | Pro- and pro-resolving mediator pools; immunometabolic reprogramming | Increased with COVID-19 severity (ρ ≈ 0.70–0.74 vs. WHO score) [29] |
Specific lipids in TB (e.g., CER(24:0)H, HCER(d18:0/22:0)H, PE(18:1/18:1)) | Sphingolipid and PE dysregulation; reflect chronic inflammation and immune evasion | Strong diagnostic classifiers in pulmonary TB (AUC > 0.75) [27] |
Mycoplasma pneumoniae perturbations (LPE, LPC, PI, TG) | Reflect membrane damage and altered energy metabolism | Differentiated bronchitis vs. pneumonia and correlated with severity in children [28] |
Dimension | Bacterial VAP | Bacterial CAP | Viral VAP (Non-COVID-19) | Viral CAP (Non-COVID-19) | COVID-19 | Tuberculosis | Mycoplasma Pneumoniae |
---|---|---|---|---|---|---|---|
Representative cohorts and samples | Pediatric ICU VAP suspicion; plasma; serial samples Days 1–12 [15] | CAP cohorts incl. sepsis; serum/plasma; multicenter and ICU [13,19,22] | — | Influenza A, Rhinovirus; sputum [24] | Hospitalized patients; serum/tracheal aspirate [18,20,21,29,30] | Multicenter CAP-like cohorts; plasma [26,27] | Children with M. pneumoniae infections; plasma/urine [28] |
Major lipid pattern | PCs, SMs, TGs, DGs altered; higher PCs and TGs in high mCPIS [15] | ↑ PCs and ether-PCs; ↓ LPC; CE and LPC inversely tracked SOFA/inflammation; TG-rich signature in sepsis [13,19,22] | — | ↑ TGs, DGs, oxylipins in influenza; rhinovirus lipid-depleted [24] | ↓ LPC(22:6-sn2), ↑ PC(36:1), acylcarnitines ↑; hypolipidemia with ↓ HDL-c/PC/LPC; ↑ GlycA/B/F [18,20,29,30] | ↑ PE, PC; ↓ total cholesterol, TAGs, ceramides; diagnostic lipid classifiers identified [26,27] | Perturbations in LPC, LPE, PI, TG reflecting infection severity [28] |
Signature lipids | PC 32:2 ↑, TG 48:3 ↑, SM 40:1 ↑; AUC > 0.75 [15] | PC(16:0_18:1), PC(18:2_20:4), PC(36:4), PC(38:6) ↓ with severity; CE recovery prognostic [13,19,22] | — | Cluster-specific acylcarnitines, DGs, TGs, LPCs [24] | SM(34:1), PC(O-34:1) best VAP markers; LPC(22:6-sn2)/PC(36:1) ratio AUC > 0.95 [18,21] | CER(24:0)H, HCER(d18:0/22:0)H, PE(18:1/18:1) diagnostic [27] | Differential LPE, LPC, PI species [28] |
Diagnostic/prognostic readouts | Discriminated VAP severity and pathogen type; AUC > 0.75 for select lipids [15] | PC species outperformed CURB-65/PSI (AUC ~0.95); CE recovery was associated with lower 30-day mortality (OR 0.84) [13,19,22] | — | Influenza A H3 odds 15× higher vs. Rhinovirus in lipid-rich cluster [24] | High diagnostic accuracy (AUC up to 0.95–0.99); lipid panels outperformed CRP/PCT [18,20,21,29,30] | High AUC (>0.75–0.99) for TB classifiers [26,27] | Plasma/urine metabolite-lipid panels AUC 0.927–1.0 [28] |
Inflammatory mediators | Lipid shifts correlated with inflammatory markers [15] | Correlated with CRP, PCT, PSI [13,22] | — | Not reported [24] | GlycA/B/F glycoproteins correlated with severity; CRP/PCT comparison [20,21] | Correlation with immune cell profiles, inflammation [26,27] | Correlated with neutrophil/lymphocyte ratio [28] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Georgakopoulou, V.E.; Dodos, K.; Pitiriga, V.C. Role of Lipidomics in Respiratory Tract Infections: A Systematic Review of Emerging Evidence. Microorganisms 2025, 13, 2190. https://doi.org/10.3390/microorganisms13092190
Georgakopoulou VE, Dodos K, Pitiriga VC. Role of Lipidomics in Respiratory Tract Infections: A Systematic Review of Emerging Evidence. Microorganisms. 2025; 13(9):2190. https://doi.org/10.3390/microorganisms13092190
Chicago/Turabian StyleGeorgakopoulou, Vasiliki E., Konstantinos Dodos, and Vassiliki C. Pitiriga. 2025. "Role of Lipidomics in Respiratory Tract Infections: A Systematic Review of Emerging Evidence" Microorganisms 13, no. 9: 2190. https://doi.org/10.3390/microorganisms13092190
APA StyleGeorgakopoulou, V. E., Dodos, K., & Pitiriga, V. C. (2025). Role of Lipidomics in Respiratory Tract Infections: A Systematic Review of Emerging Evidence. Microorganisms, 13(9), 2190. https://doi.org/10.3390/microorganisms13092190