Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter November 6, 2023

Targeted quantitative lipidomic uncovers lipid biomarkers for predicting the presence of compensated cirrhosis and discriminating decompensated cirrhosis from compensated cirrhosis

  • Yongbin Zeng , Li Zhang , Zhiyi Zheng , Jingyi Su , Ya Fu , Tianbin Chen , Kun Lin , Can Liu , Huanhuan Huang , Qishui Ou EMAIL logo and Yongjun Zeng EMAIL logo

Abstract

Objectives

This study aimed to characterize serum lipid metabolism and identify potential biomarkers for compensated cirrhosis (CC) predicting and decompensated cirrhosis (DC) discrimination using targeted quantitative lipidomics and machine learning approaches.

Methods

Serum samples from a cohort of 120 participants was analyzed, including 90 cirrhosis patients (45 CC patients and 45 DC patients) and 30 healthy individuals. Lipid metabolic profiling was performed using targeted LC-MS/MS. Two machine learning methods, least absolute shrinkage and selection operator (LASSO), and random forest (RF) were applied to screen for candidate metabolite biomarkers.

Results

The metabolic profiling analysis showed a significant disruption in patients with CC and DC. Compared to the CC group, the DC group exhibited a significant upregulation in the abundance of glycochenodeoxycholic acid (GCDCA), glyco-ursodeoxycholic acid (GUDCA), glycocholic acid (GCA), phosphatidylethanolamine (PE), N-acyl-lyso-phosphatidylethanolamine (LNAPE), and triglycerides (TG), and a significant downregulation in the abundance of ceramides (Cer) and lysophosphatidylcholines (LPC). Machine learning identified 11 lipid metabolites (abbreviated as BMP11) as potential CC biomarkers with excellent prediction performance, with an AUC of 0.944, accuracy of 94.7 %, precision of 95.6 %, and recall of 95.6 %. For DC discrimination, eight lipids (abbreviated as BMP8) were identified, demonstrating strong efficacy, with an AUC of 0.968, accuracy of 92.2 %, precision of 88.0 %, and recall of 97.8 %.

Conclusions

This study unveiled distinct lipidomic profiles in CC and DC patients and established robust lipid-based models for CC predicting and DC discrimination.


Corresponding authors: Qishui Ou, Department of Laboratory Medicine, Gene Diagnosis Research Center, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P.R. China; Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, P.R. China; Fujian Key Laboratory of Laboratory Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P.R. China; and Fujian Clinical Research Center for Laboratory Medicine of Immunology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P.R. China, E-mail: ; and Yongjun Zeng, Department of Cardiology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, P.R. China, E-mail:
Yongbin Zeng and Li Zhang contributed equally to this work.

Award Identifier / Grant number: 82172338

Award Identifier / Grant number: 82202596

Award Identifier / Grant number: 82372316

Acknowledgments

The authors thank all the participants in this study for their cooperation in sample preparation.

  1. Research ethics: The study was approved by the Ethics Committee of the First Affiliated Hospital of Fujian Medical University (MTCA, ECFAH of FMU [2021] 047).

  2. Informed consent: Written informed consent was obtained from each patient.

  3. Author contributions: Yongbin Zeng, Ya Fu and Li Zhang: Data analysis, Manuscript writing; Zhiyi Zheng and Jingyi Su: Data collection; Yongbin Zeng, Yongjun Zeng and Qishui Ou: Project development, Manuscript editing; Kun Lin and Huanhuan Huang: Samples preparation; Tianbin Chen and Can Liu: Laboratory testing.

  4. Competing interests: The authors declare that they have no conflicts of interest regarding the publication of this article.

  5. Research funding: The study is supported by grants from National Natural Science Foundation of China (82172338, 82202596, 82372316).

  6. Data availability: The raw data can be obtained on request from the corresponding author.

References

1. Huang, DQ, Terrault, NA, Tacke, F, Gluud, LL, Arrese, M, Bugianesi, E, et al.. Global epidemiology of cirrhosis – aetiology, trends and predictions. Nat Rev Gastroenterol Hepatol 2023;20:388–98. https://doi.org/10.1038/s41575-023-00759-2.Search in Google Scholar PubMed PubMed Central

2. Schneider, ARP, Schneider, CV, Schneider, KM, Baier, V, Schaper, S, Diedrich, C, et al.. Early prediction of decompensation (EPOD) score: non-invasive determination of cirrhosis decompensation risk. Liver Int 2022;42:640–50. https://doi.org/10.1111/liv.15161.Search in Google Scholar PubMed

3. D’Amico, G, Morabito, A, D’Amico, M, Pasta, L, Malizia, G, Rebora, P, et al.. Clinical states of cirrhosis and competing risks. J Hepatol 2018;68:563–76. https://doi.org/10.1016/j.jhep.2017.10.020.Search in Google Scholar PubMed

4. D’Amico, G. The clinical course of cirrhosis. Population based studies and the need of personalized medicine. J Hepatol 2014;60:241–2. https://doi.org/10.1016/j.jhep.2013.10.023.Search in Google Scholar PubMed

5. Wishart, DS. Metabolomics for investigating physiological and pathophysiological processes. Physiol Rev 2019;99:1819–75. https://doi.org/10.1152/physrev.00035.2018.Search in Google Scholar PubMed

6. Tzeng, HT, Chyuan, IT, Chen, WY. Shaping of innate immune response by fatty acid metabolite palmitate. Cells 2019;8:1633. https://doi.org/10.3390/cells8121633.Search in Google Scholar PubMed PubMed Central

7. Armour, SM, Remsberg, JR, Damle, M, Sidoli, S, Ho, WY, Li, Z, et al.. An HDAC3-PROX1 corepressor module acts on HNF4α to control hepatic triglycerides. Nat Commun 2017;8:549. https://doi.org/10.1038/s41467-017-00772-5.Search in Google Scholar PubMed PubMed Central

8. Schuppan, D, Afdhal, NH. Liver cirrhosis. Lancet 2008;371:838–51. https://doi.org/10.1016/s0140-6736(08)60383-9.Search in Google Scholar

9. Shama, S, Jang, H, Wang, X, Zhang, Y, Shahin, NN, Motawi, TK, et al.. Phosphatidylethanolamines are associated with nonalcoholic fatty liver disease (NAFLD) in obese adults and induce liver cell metabolic perturbations and hepatic stellate cell activation. Int J Mol Sci 2023;24:1024. https://doi.org/10.3390/ijms24021034.Search in Google Scholar PubMed PubMed Central

10. Su, D, Liao, Z, Feng, B, Wang, T, Shan, B, Zeng, Q, et al.. Pulsatilla chinensis saponins cause liver injury through interfering ceramide/sphingomyelin balance that promotes lipid metabolism dysregulation and apoptosis. Phytomedicine 2020;76:153265. https://doi.org/10.1016/j.phymed.2020.153265.Search in Google Scholar PubMed

11. Wang, Y, Aoki, H, Yang, J, Peng, K, Liu, R, Li, X, et al.. The role of sphingosine 1-phosphate receptor 2 in bile-acid-induced cholangiocyte proliferation and cholestasis-induced liver injury in mice. Hepatology 2017;65:2005–18. https://doi.org/10.1002/hep.29076.Search in Google Scholar PubMed PubMed Central

12. Yuan, Z, Wang, J, Zhang, H, Chai, Y, Xu, Y, Miao, Y, et al.. Glycocholic acid aggravates liver fibrosis by promoting the up-regulation of connective tissue growth factor in hepatocytes. Cell Signal 2022;101:110508. https://doi.org/10.1016/j.cellsig.2022.110508.Search in Google Scholar PubMed

13. Hernandez-Lara, MA, Yadav, SK, Shah, SD, Okumura, M, Yokoyama, Y, Penn, RB, et al.. Regulation of airway smooth muscle cell proliferation by diacylglycerol kinase: relevance to airway remodeling in asthma. Int J Mol Sci 2022;23:11868. https://doi.org/10.3390/ijms231911868.Search in Google Scholar PubMed PubMed Central

14. Reeves, HL, Thompson, MG, Dack, CL, Burt, AD, Day, CP. The role of phosphatidic acid in platelet-derived growth factor-induced proliferation of rat hepatic stellate cells. Hepatology 2000;31:95–100. https://doi.org/10.1002/hep.510310116.Search in Google Scholar PubMed

15. Wu, X, Yao, Z, Zhao, L, Zhang, Y, Cao, M, Li, T, et al.. Phosphatidylserine on blood cells and endothelial cells contributes to the hypercoagulable state in cirrhosis. Liver Int 2016;36:1800–10. https://doi.org/10.1111/liv.13167.Search in Google Scholar PubMed

16. Serag, WM, Mohammed, BSE, Mohamed, MM, Elsayed, BE. Predicting the risk of portal vein thrombosis in patients with liver cirrhosis and hepatocellular carcinoma. Heliyon 2020;6:e04677. https://doi.org/10.1016/j.heliyon.2020.e04677.Search in Google Scholar PubMed PubMed Central

17. Han, X, Wang, J, Gu, H, Guo, H, Cai, Y, Liao, X, et al.. Predictive value of serum bile acids as metabolite biomarkers for liver cirrhosis: a systematic review and meta-analysis. Metabolomics 2022;18:43. https://doi.org/10.1007/s11306-022-01890-y.Search in Google Scholar PubMed

18. He, X, Zhou, MX, Cheng, C, Li, SS, Gao, Y, Ma, ZT, et al.. Metabolomic profiling for histologically fibrotic stage in chronic drug-induced liver injury. Front Pharmacol 2022;13:896198. https://doi.org/10.3389/fphar.2022.896198.Search in Google Scholar PubMed PubMed Central

19. Vasavan, T, Deepak, S, Jayawardane, IA, Lucchini, M, Martin, C, Geenes, V, et al.. Fetal cardiac dysfunction in intrahepatic cholestasis of pregnancy is associated with elevated serum bile acid concentrations. J Hepatol 2020;74:1087–96. https://doi.org/10.1016/j.jhep.2020.11.038.Search in Google Scholar PubMed PubMed Central

20. Miao, J, Guo, L, Cui, H, Wang, L, Zhu, B, Lei, J, et al.. Er-chen decoction alleviates high-fat diet-induced nonalcoholic fatty liver disease in rats through remodeling gut microbiota and regulating the serum metabolism. Evid Based Complement Alternat Med 2022;2022:6221340. https://doi.org/10.1155/2022/6221340.Search in Google Scholar PubMed PubMed Central

21. Claria, J, Curto, A, Moreau, R, Colsch, B, Lopez-Vicario, C, Lozano, JJ, et al.. Untargeted lipidomics uncovers lipid signatures that distinguish severe from moderate forms of acutely decompensated cirrhosis. J Hepatol 2021;75:1116–27. https://doi.org/10.1016/j.jhep.2021.06.043.Search in Google Scholar PubMed

22. Sehrawat, TS, Arab, JP, Liu, M, Amrollahi, P, Wan, M, Fan, J, et al.. Circulating extracellular vesicles carrying sphingolipid cargo for the diagnosis and dynamic risk profiling of alcoholic hepatitis. Hepatology 2020;73:571–85. https://doi.org/10.1002/hep.31256.Search in Google Scholar PubMed PubMed Central

23. Pan, X, Dutta, D, Lu, S, Bellen, HJ. Sphingolipids in neurodegenerative diseases. Front Neurosci 2023;17:1137893. https://doi.org/10.3389/fnins.2023.1137893.Search in Google Scholar PubMed PubMed Central

24. Cartier, A, Hla, T. Sphingosine 1-phosphate: lipid signaling in pathology and therapy. Science 2019;366:6463. https://doi.org/10.1126/science.aar5551.Search in Google Scholar PubMed PubMed Central

25. Yu, S, Wang, K, Li, Q, Wei, Y, Li, Y, Zhang, Q, et al.. Nonalcoholic steatohepatitis critically rewires the ischemia/reperfusion-induced dysregulation of cardiolipins and sphingolipids in mice. Hepatobiliary Surg Nutr 2023;12:3–19. https://doi.org/10.21037/hbsn-21-133.Search in Google Scholar PubMed PubMed Central

26. Höring, M, Peschel, G, Grimm, J, Krautbauer, S, Müller, M, Weigand, K, et al.. Serum ceramide species are associated with liver cirrhosis and viral genotype in patients with hepatitis C infection. Int J Mol Sci 2022;23:9806. https://doi.org/10.3390/ijms23179806.Search in Google Scholar PubMed PubMed Central


Supplementary Material

This article contains supplementary material (https://doi.org/10.1515/cclm-2023-0798).


Received: 2023-07-26
Accepted: 2023-09-23
Published Online: 2023-11-06
Published in Print: 2024-02-26

© 2023 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 9.5.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2023-0798/html
Scroll to top button