Metabolic profiles in community-acquired pneumonia: developing assessment tools for disease severity

Pu Ning, Yali Zheng, Qiongzhen Luo, Xiaohui Liu, Yu Kang, Yan Zhang, Rongbao Zhang, Yu Xu, Donghong Yang, Wen Xi, Keqiang Wang, Yusheng Chen, Shuchang An, Zhancheng Gao, Pu Ning, Yali Zheng, Qiongzhen Luo, Xiaohui Liu, Yu Kang, Yan Zhang, Rongbao Zhang, Yu Xu, Donghong Yang, Wen Xi, Keqiang Wang, Yusheng Chen, Shuchang An, Zhancheng Gao

Abstract

Background: This study aimed to determine whether community-acquired pneumonia (CAP) had a metabolic profile and whether this profile can be used for disease severity assessment.

Methods: A total of 175 individuals including 119 CAP patients and 56 controls were enrolled and divided into two cohorts. Serum samples from a discovery cohort (n = 102, including 38 non-severe CAP, 30 severe CAP, and 34 age and sex-matched controls) were determined by untargeted ultra-high-performance liquid chromatography with tandem mass spectrometry (LC-MS/MS)-based metabolomics. Selected differential metabolites between CAP patients versus controls, and between the severe CAP group versus non-severe CAP group, were confirmed by targeted mass spectrometry assays in a validation cohort (n = 73, including 32 non-severe CAP, 19 severe CAP and 22 controls). Pearson's correlation analysis was performed to assess relationships between the identified metabolites and clinical severity of CAP. The area under the curve (AUC), sensitivity and specificity of the metabolites for predicting the severity of CAP were also investigated.

Results: The metabolic signature was markedly different between CAP patients and controls. Fifteen metabolites were found to be significantly dysregulated in CAP patients, which were mainly mapped to the metabolic pathways of sphingolipid, arginine, pyruvate and inositol phosphate. The alternation trends of five metabolites among the three groups including sphinganine, p-Cresol sulfate, dehydroepiandrosterone sulfate (DHEA-S), lactate and L-arginine in the validation cohort were consistent with those in the discovery cohort. Significantly lower concentrations of sphinganine, p-Cresol sulfate and DHEA-S were observed in CAP patients than in controls (p < 0.05). Serum lactate and sphinganine levels were positively correlated with confusion, urea level, respiratory rate, blood pressure, and age > 65 years (CURB-65), pneumonia severity index (PSI) and Acute Physiology and Chronic Health Evaluation II (APACHE II) scores, while DHEA-S inversely correlated with the three scoring systems. Combining lactate, sphinganine and DHEA-S as a metabolite panel for discriminating severe CAP from non-severe CAP exhibited a better AUC of 0.911 (95% confidence interval 0.825-0.998) than CURB-65, PSI and APACHE II scores.

Conclusions: This study demonstrates that serum metabolomics approaches based on the LC-MS/MS platform can be applied as a tool to reveal metabolic changes during CAP and establish a metabolite signature related to disease severity.

Trial registration: ClinicalTrials.gov, NCT03093220 . Registered retrospectively on 28 March 2017.

Keywords: Biomarker; Community-acquired pneumonia; Liquid chromatography–mass spectrometry; Metabolomics; Severity.

Conflict of interest statement

Ethics approval and consent to participate

The study protocol was approved by the Institutional Review Board of the Peking University People’s Hospital (Beijing, China). Written informed consent was obtained from all patients or their surrogates.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
Flowchart of study population enrolment. CAP community-acquired pneumonia, ICU intensive care unit, NSCAP non-severe CAP, SCAP severe CAP
Fig. 2
Fig. 2
Multivariate statistical analysis of serum samples in discovery cohort. a PCA score plots. Five samples (four severe CAP and one non-severe CAP) are placed outside the ellipse that describes the 95% CI of Hotelling’s T-squared distribution. b OPLS-DA three-dimensional score plot discriminates all CAP subjects versus controls in discovery cohort (R2Y = 0.937, Q2 = 0.814, p < 0.0001). c OPLS-DA score plots of non-severe CAP versus severe CAP groups (R2Y = 0.757, Q2 = 0.465, p < 0.0001). d OPLS-DA score plots of non-severe CAP patients versus controls (R2Y = 0.994, Q2 = 0.955, p < 0.0001). e OPLS-DA score plots of severe CAP patients versus controls (R2Y = 0.996, Q2 = 0.854, p < 0.0001). R2Y represents goodness of fit, Q2 represents goodness of prediction, p value shows significance level of the model. CAP community-acquired pneumonia, PC principal component, QC quality control
Fig. 3
Fig. 3
Fifteen metabolites dysregulated in CAP compared to controls. a Hierarchical cluster heatmap of 15 metabolites in three groups. Row represents metabolites and column represents individual samples. Green, red and blue represent non-severe CAP (NSCAP), severe CAP (SCAP) and controls, respectively. Greater brown indicates higher relative intensity of metabolites, while light blue indicates lower intensity. b Metabolic pathway analysis of 15 metabolites changed in CAP. Node colour based on p value and node radius determined based on pathway impact values. CAP community-acquired pneumonia, DHEA-S dehydroepiandrosterone sulfate
Fig. 4
Fig. 4
Three metabolites identified that can discriminate severe CAP from non-severe CAP in validation cohort. Chemical structures of three metabolites. Box–whisker plots of concentrations of three metabolites in three groups. Horizontal line represents median; bottom and the top of box represent 25th and the 75th percentiles; whiskers represent 5% and 95% percentiles. *p < 0.01, **p < 0.001. CAP community-acquired pneumonia, DHEA-S dehydroepiandrosterone sulfate, NSCAP non-severe CAP, SCAP severe CAP
Fig. 5
Fig. 5
Correlations of five metabolites with clinical parameters and assessment performance. a Pearson’s correlation heatmap of five serum metabolites and clinical parameters. Greater intensities of brown and blue indicate higher positive or negative correlations, respectively. Resulting correlation matrix presented in Additional file 3: Table S6. b ROC curve analysis of various parameters for discrimination of severe CAP from non-severe CAP. DHEA-S dehydroepiandrosterone sulfate, APACHE II Acute Physiology and Chronic Health Evaluation II, CURB-65 confusion, urea level, respiratory rate, blood pressure, and > 65 years, PSI pneumonia severity index, ESR erythrocyte sedimentation rate, WBC white blood cell, PCT procalcitonin, NE% percentage of neutrophils, CRP C-reactive protein, AUC area under the curve

References

    1. Welte T, Torres A, Nathwani D. Clinical and economic burden of community-acquired pneumonia among adults in Europe. Thorax. 2012;67:71–79. doi: 10.1136/thx.2009.129502.
    1. Christ-Crain M, Opal SM. Clinical review: The role of biomarkers in the diagnosis and management of community-acquired pneumonia. Crit Care. 2010;14:203. doi: 10.1186/cc8155.
    1. Liu W, Peng L, Hua S. Clinical significance of dynamic monitoring of blood lactic acid, oxygenation index and C-reactive protein levels in patients with severe pneumonia. Exp Ther Med. 2015;10:1824–1828. doi: 10.3892/etm.2015.2770.
    1. Aliberti S, Brambilla AM, Chalmers JD, Cilloniz C, Ramirez J, Bignamini A, et al. Phenotyping community-acquired pneumonia according to the presence of acute respiratory failure and severe sepsis. Respir Res. 2014;15:27. doi: 10.1186/1465-9921-15-27.
    1. Biron BM, Ayala A, Lomas-Neira JL. Biomarkers for sepsis: what is and what might be? Biomark Insights. 2015;10:7–17.
    1. Scicluna BP, Klein Klouwenberg PM, van Vught LA, Wiewel MA, Ong DS, Zwinderman AH, et al. A molecular biomarker to diagnose community-acquired pneumonia on intensive care unit admission. Am J Respir Crit Care Med. 2015;192:826–835. doi: 10.1164/rccm.201502-0355OC.
    1. Severino P, Silva E, Baggio-Zappia GL, Brunialti MK, Nucci LA, Rigato O, Jr, et al. Patterns of gene expression in peripheral blood mononuclear cells and outcomes from patients with sepsis secondary to community acquired pneumonia. PLoS One. 2014;9:e91886. doi: 10.1371/journal.pone.0091886.
    1. Lin WC, Lu SL, Lin CF, Chen CW, Chao L, Chao J, et al. Plasma kallistatin levels in patients with severe community-acquired pneumonia. Crit Care. 2013;17:R27. doi: 10.1186/cc12507.
    1. Schuetz P, Wolbers M, Christ-Crain M, Thomann R, Falconnier C, Widmer I, et al. Prohormones for prediction of adverse medical outcome in community-acquired pneumonia and lower respiratory tract infections. Crit Care. 2010;14:R106. doi: 10.1186/cc9055.
    1. Mauri T, Coppadoro A, Bombino M, Bellani G, Zambelli V, Fornari C, et al. Alveolar pentraxin 3 as an early marker of microbiologically confirmed pneumonia: a threshold-finding prospective observational study. Crit Care. 2014;18:562. doi: 10.1186/s13054-014-0562-5.
    1. Serkova NJ, Standiford TJ, Stringer KA. The emerging field of quantitative blood metabolomics for biomarker discovery in critical illnesses. Am J Respir Crit Care Med. 2011;184:647–655. doi: 10.1164/rccm.201103-0474CI.
    1. Slupsky CM, Rankin KN, Fu H, Chang D, Rowe BH, Charles PG, et al. Pneumococcal pneumonia: potential for diagnosis through a urinary metabolic profile. J Proteome Res. 2009;8:5550–5558. doi: 10.1021/pr9006427.
    1. Laiakis EC, Morris GA, Fornace AJ, Howie SR. Metabolomic analysis in severe childhood pneumonia in the Gamebia, West Africa: findings from a pilot study. PLoS One. 2010;5:e12655. doi: 10.1371/journal.pone.0012655.
    1. Banoei MM, Vogel HJ, Weljie AM, Kumar A, Yende S, Angus DC, et al. Plasma metabolomics for the diagnosis and prognosis of H1N1 influenza pneumonia. Crit Care. 2017;21:97. doi: 10.1186/s13054-017-1672-7.
    1. Niederman MS, Mandell LA, Anzueto A, Bass JB, Broughton WA, Campbell GD, et al. Guidelines for the management of adults with community-acquired pneumonia. Diagnosis, assessment of severity, antimicrobial therapy, and prevention. Am J Respir Crit Care Med. 2001;163:1730–1754. doi: 10.1164/ajrccm.163.7.at1010.
    1. Mandell LA, Wunderink RG, Anzueto A, Bartlett JG, Campbell GD, Dean NC, et al. Infectious Diseases Society of America/American Thoracic Society Consensus Guidelines on the management of community-acquired pneumonia in adults. Clin Infect Dis. 2007;44(Suppl 2):S27–S72. doi: 10.1086/511159.
    1. Xia J, Wishart DS. Using MetaboAnalyst 3.0 for comprehensive metabolomics data analysis. Curr Protoc Bioinformatics. 2016;55:14.10.1–14.10.91. doi: 10.1002/cpbi.11.
    1. Tan B, Qiu Y, Zou X, Chen T, Xie G, Cheng Y, et al. Metabonomics identifies serum metabolite markers of colorectal cancer. J Proteome Res. 2013;12:3000–3009. doi: 10.1021/pr400337b.
    1. Luies L, Mienie J, Motshwane C, Ronacher K, Walzl G, Loots DT. Urinary metabolite markers characterizing tuberculosis treatment failure. Metabolomics. 2017;13:124. doi: 10.1007/s11306-017-1261-4.
    1. Wiklund S, Johansson E, Sjöström L, Mellerowicz EJ, Edlund U, Shockcor JP, et al. Visualization of GC/TOF-MS-based metabolomics data for identification of biochemically interesting compounds using OPLS class models. Anal Chem. 2008;80:115–122. doi: 10.1021/ac0713510.
    1. Wheelock CE, Goss VM, Balgoma D, Nicholas B, Brandsma J, Skipp PJ, et al. Application of ‘omics technologies to biomarker discovery in inflammatory lung diseases. Eur Respir J. 2013;42:802–825. doi: 10.1183/09031936.00078812.
    1. Hanada K, Nishijima M. Selection of mammalian cell mutants in sphingolipid biosynthesis. Methods Enzymol. 2000;312:304–317. doi: 10.1016/S0076-6879(00)12917-9.
    1. Ohanian J, Ohanian V. Sphingolipids in mammalian cell signalling. Cell Mol Life Sci. 2001;58:2053–2068. doi: 10.1007/PL00000836.
    1. Sharma L, Prakash H. Sphingolipids are dual specific drug targets for the management of pulmonary infections: perspective. Front Immunol. 2017;8:378.
    1. Nishimura M, Saida T, Kuroki S, Kawabata T, Obayashi H, Saida K, et al. Post-infectious encephalitis with anti-galactocerebroside antibody subsequent to Mycoplasma pneumoniae infection. J Neurol Sci. 1996;140:91–95. doi: 10.1016/0022-510X(96)00106-2.
    1. Wolf K, Hackstadt T. Sphingomyelin trafficking in Chlamydia pneumoniae-infected cells. Cell Microbiol. 2001;3:145–152. doi: 10.1046/j.1462-5822.2001.00098.x.
    1. Dias TR, Alves MG, Almeida SP, Silva J, Barros A, Sousa M, et al. Dehydroepiandrosterone and 7-oxo-dehydroepiandrosterone in male reproductive health: implications of differential regulation of human Sertoli cells metabolic profile. J Steroid Biochem Mol Biol. 2015;154:1–11. doi: 10.1016/j.jsbmb.2015.06.004.
    1. Oberbeck R, Dahlweid M, Koch R, van Griensven M, Emmendörfer A, Tscherne H, et al. Dehydroepiandrosterone decreases mortality rate and improves cellular immune function during polymicrobial sepsis. Crit Care Med. 2001;29:380–384. doi: 10.1097/00003246-200102000-00029.
    1. Suzuki T, Suzuki N, Daynes RA, Engleman EG. Dehydroepiandrosterone enhances IL2 production and cytotoxic effector function of human T cells. Clin Immunol Immunopathol. 1991;61:202–211. doi: 10.1016/S0090-1229(05)80024-8.
    1. Bozza VV, D'Attilio L, Mahuad CV, Giri AA, Del Rey A, Besedovsky H, et al. Altered cortisol/DHEA ratio in tuberculosis patients and its relationship with abnormalities in the mycobacterial-driven cytokine production by peripheral blood mononuclear cells. Scand J Immunol. 2007;66:97–103. doi: 10.1111/j.1365-3083.2007.01952.x.
    1. Radford DJ, Wang K, McNelis JC, Taylor AE, Hechenberger G, Hofmann J, et al. Dehydroepiandrosterone sulfate directly activates protein kinase C-beta to increase human neutrophil superoxide generation. Mol Endocrinol. 2010;24:813–821. doi: 10.1210/me.2009-0390.
    1. Monahan LG, Hajduk IV, Blaber SP, Charles IG, Harry EJ. Coordinating bacterial cell division with nutrient availability: a role for glycolysis. MBio. 2014;5:e00935–e00914. doi: 10.1128/mBio.00935-14.
    1. Javed A, Guirgis FW, Sterling SA, Puskarich MA, Bowman J, Robinson T, et al. Clinical predictors of early death from sepsis. J Crit Care. 2017;42:30–34. doi: 10.1016/j.jcrc.2017.06.024.
    1. Shapiro NI, Howell MD, Talmor D, Nathanson LA, Lisbon A, Wolfe RE, et al. Serum lactate as a predictor of mortality in emergency department patients with infection. Ann Emerg Med. 2005;45:524–528. doi: 10.1016/j.annemergmed.2004.12.006.
    1. Mikkelsen ME, Miltiades AN, Gaieski DF, Goyal M, Fuchs BD, Shah CV, et al. Serum lactate is associated with mortality in severe sepsis independent of organ failure and shock. Crit Care Med. 2009;37:1670–1677. doi: 10.1097/CCM.0b013e31819fcf68.
    1. Frenzen FS, Kutschan U, Meiswinkel N, Schulte-Hubbert B, Ewig S, Kolditz M. Admission lactate predicts poor prognosis independently of the CRB/CURB-65 scores in community-acquired pneumonia. Clin Microbiol Infect. 2018;24:306.e1–306.e6. doi: 10.1016/j.cmi.2017.07.007.

Source: PubMed

3
Abonnieren