Biomarkers of inflammation, immunosuppression and stress with active disease are revealed by metabolomic profiling of tuberculosis patients

January Weiner 3rd, Shreemanta K Parida, Jeroen Maertzdorf, Gillian F Black, Dirk Repsilber, Anna Telaar, Robert P Mohney, Cordelia Arndt-Sullivan, Christian A Ganoza, Kellen C Faé, Gerhard Walzl, Stefan H E Kaufmann, January Weiner 3rd, Shreemanta K Parida, Jeroen Maertzdorf, Gillian F Black, Dirk Repsilber, Anna Telaar, Robert P Mohney, Cordelia Arndt-Sullivan, Christian A Ganoza, Kellen C Faé, Gerhard Walzl, Stefan H E Kaufmann

Abstract

Although tuberculosis (TB) causes more deaths than any other pathogen, most infected individuals harbor the pathogen without signs of disease. We explored the metabolome of >400 small molecules in serum of uninfected individuals, latently infected healthy individuals and patients with active TB. We identified changes in amino acid, lipid and nucleotide metabolism pathways, providing evidence for anti-inflammatory metabolomic changes in TB. Metabolic profiles indicate increased activity of indoleamine 2,3 dioxygenase 1 (IDO1), decreased phospholipase activity, increased abundance of adenosine metabolism products, as well as indicators of fibrotic lesions in active disease as compared to latent infection. Consistent with our predictions, we experimentally demonstrate TB-induced IDO1 activity. Furthermore, we demonstrate a link between metabolic profiles and cytokine signaling. Finally, we show that 20 metabolites are sufficient for robust discrimination of TB patients from healthy individuals. Our results provide specific insights into the biology of TB and pave the way for the rational development of metabolic biomarkers for TB.

Conflict of interest statement

Competing Interests: RM is an employee of Metabolon, Inc. This does not alter the authors’ adherence to all the PLoS ONE policies on sharing data and materials.

Figures

Figure 1. Examples of metabolite patterns in…
Figure 1. Examples of metabolite patterns in tuberculosis patients (TBactive), healthy uninfected (TST–) and latently infected (TST+) individuals.
(A) Changes in relative abundance of three exemplary small metabolites: fibrinopeptide A, inosine and kynurenine. Red line indicates sample mean, blue line indicates sample median. Stars indicate significant differences between profiles (result from t-test corrected for multiple testing; *, p–; green circles, TST+; red triangles, TBactive. Spearman correlation coefficients (rho) and corresponding p-values are given.
Figure 2. Heat map showing fold changes…
Figure 2. Heat map showing fold changes of small metabolic compounds in the three study groups, TB patients, healthy uninfected and latently infected individuals.
Fold changes are relative to the average abundance in the TST– group. Red indicates relative abundance higher than average in the TST– group; blue indicates relative abundance lower than average in the TST– group. Horizontal axis: samples belonging to the three study groups; vertical axis: top 50 compounds selected by variable importance in RF analysis, including compounds that could not be identified, but were strong predictors of sample status. Color bars above the heat map denote study groups: grey, TST–; green, TST+; red, TBactive. See also Figure S2.
Figure 3. Network showing functional relationships between…
Figure 3. Network showing functional relationships between the small metabolic compounds in TB patients, healthy uninfected and latently infected individuals.
Nodes correspond to metabolites; edges correspond to statistically significant correlation between residual small metabolite profiles corrected for study classes. Colors correspond to differences between the TST+ and classes (see Figure S3 for additional comparisons). Color intensity indicates significance of difference with darker colors corresponding to more significant differences. Metabolites with adjusted p value >0.05 are not colored. Line thickness corresponds to the absolute Spearman correlation coefficients corrected for groups (see Methods). See also Figure S3.
Figure 4. Abundance of cytokines and their…
Figure 4. Abundance of cytokines and their correlation with selected metabolites in TB patients, healthy uninfected and latently infected individuals.
(A) Strip charts showing abundances of eight cytokines that differed significantly between the study groups. Significance thresholds for a two-tailed t-test corrected for multiple testing: *, p–); green circles, latently infected individuals (TST+); brown triangles, active TB patients (TBactive). Spearman correlation coefficient (rho) and p-values are given.
Figure 5. Demonstration of IDO1 expression and…
Figure 5. Demonstration of IDO1 expression and kynurenine production in response to M. tuberculosis infection and regulation of M. tuberculosis-specific T-cell responses by kynurenines.
(A) Immunohistochemistry staining of formalin-fixed, paraffin-embedded tissue of a murine pulmonary TB lesion stained with anti-IDO polyclonal antibody; staining representative of lesions from five animals. Bar is equal to 200 nm. Human monocyte-derived dendritic cells (DCs) (B) and macrophages (C) were infected with M. tuberculosis H37Rv or stimulated with irradiated and heat-killed M. tuberculosis H37Rv for 24 h and indoleamine 2,3 dioxygenase 1 (IDO1) gene expression was measured by qPCR. Mean and standard deviation (SD) of fold-change IDO1 gene expression of one donor representative of four. Line indicates minimal significant fold change threshold equal to 1.5. DCs (D) and macrophages (E) were infected with M. tuberculosis H37Rv and cell culture supernatants were collected at indicated times for measurement of kynurenines by HPLC; kynurenine levels from uninfected controls were subtracted. Means and SD of four donors are depicted (C and D). Star indicates significance (p<0.05) in Friedman test.(F) Human monocyte-derived DCs were pulsed with purified protein derivative (PPD) and mannosylated lipoarabinomannan (ManLAM) and co-cultured for 4 days with autologous CFSE-labeled purified T cells (DC:T cell ratio 1∶20) in the presence or absence of 1-methyl-DL-tryptophan (1-MT-DL) and 3-OH-kynurenine (Kyn). Cell proliferation was assessed by CFSE dilution using flowcytometry. Figure representative of three independent experiments (ANOVA F = 4.1 for CD+CD4+ cells and F = 3.3 for CD3+CD8+ cells, p<0.05).

References

    1. Illig T, Gieger C, Zhai G, Römisch-Margl W, Wang-Sattler R, et al. A genome-wide perspective of genetic variation in human metabolism. Nat Genet. 2009;42:137–141.
    1. Kaddurah-Daouk R, Kristal BS, Weinshilboum RM. Metabolomics: a global biochemical approach to drug response and disease. Pharmacol Toxicol. 2008;48:653–683.
    1. Han X, Holtzman M, McKeel DW, Jr, Kelley J, Morris JC. Substantial sulfatide deficiency and ceramide elevation in very early Alzheimer’s disease: potential role in disease pathogenesis. J Neurochem 82: 809–818. 0997 [pii] 2002.
    1. Bogdanov M, Matson WR, Wang L, Matson T, Saunders-Pullman R, et al. Metabolomic profiling to develop blood biomarkers for Parkinson’s disease. Brain. 2008;131:389–396.
    1. Sabatine MS, Liu E, Morrow DA, Heller E, McCarroll R, et al. Metabolomic identification of novel biomarkers of myocardial ischemia. Circulation. 2005;112:3868–3875.
    1. Brindle JT, Nicholson JK, Schofield PM, Grainger DJ, Holmes E. Application of chemometrics to 1 H NMR spectroscopic data to investigate a relationship between human serum metabolic profiles and hypertension. Analyst. 2003;128:32–36.
    1. Yang J, Xu G, Zheng Y, Kong H, Pang T, et al. Diagnosis of liver cancer using HPLC-based metabonomics avoiding false-positive result from hepatitis and hepatocirrhosis diseases. J Chromatogr B. 2004;813:59–65.
    1. Wang C, Kong H, Guan Y, Yang J, Gu J, et al. Plasma phospholipid metabolic profiling and biomarkers of type 2 diabetes mellitus based on high-performance liquid chromatography/electrospray mass spectrometry and multivariate statistical analysis. Anal Chem. 2005;77:4108–4116.
    1. Yuan K, Kong H, Guan Y, Yang J, Xu G. A GC-based metabonomics investigation of type 2 diabetes by organic acids metabolic profile. J Chromatogr B. 2007;850:236–240.
    1. Salek RM, Maguire ML, Bentley E, Rubtsov DV, Hough T, et al. A metabolomic comparison of urinary changes in type 2 diabetes in mouse, rat, and human. Physiological genomics. 2007;29:99–108.
    1. Suhre K, Wallaschofski H, Raffler J, Friedrich N, Haring R, et al. A genome-wide association study of metabolic traits in human urine. Nat Genet. 2011;43:565–569.
    1. Laiakis EC, Morris GAJ, Fornace AJ, Howie SRC. Metabolomic analysis in severe childhood pneumonia in the Gambia, West Africa: findings from a pilot study. PloS one. 2010;5:e12655.
    1. Wang Y, Utzinger J, Saric J, Li JV, Burckhardt J, et al. Global metabolic responses of mice to Trypanosoma brucei brucei infection. Proc Natl Acad Sci USA. 2008;105:6127–6132.
    1. Slupsky CM, Rankin KN, Fu H, Chang D, Rowe BH, et al. Pneumococcal pneumonia: potential for diagnosis through a urinary metabolic profile. J Proteome Res. 2009;8:5550–5558.
    1. Somashekar BS, Amin AG, Rithner CD, Troudt JL, Basaraba R, et al. Metabolic profiling of lung granuloma in Mycobacterium tuberculosis infected guinea pigs: ex vivo 1 H magic angle spinning NMR studies. J Proteome Res. 2011;10:4186–4195.
    1. Shin JH, Yang JY, Jeon BY, Yoon YJ, Cho SN, et al. 1H-NMR-based metabolomic profiling in mice infected with Mycobacterium tuberculosis. J Proteome Res. 2011;10:2238–2247.
    1. Lawn SD, Zumla AI. Tuberculosis. Lancet. 2011;378:57–72.
    1. Philips JA, Ernst JD. Tuberculosis Pathogenesis and Immunity. Ann Rev Pathol Mech Dis. 2012;7:353–384.
    1. Lonnroth K, Castro KG, Chakaya JM, Chauhan LS, Floyd K, et al. Tuberculosis control and elimination 2010–50: cure, care, and social development. Lancet. 2010;375:1814–1829.
    1. Jacobsen M, Repsilber D, Gutschmidt A, Neher A, Feldmann K, et al. Candidate biomarkers for discrimination between infection and disease caused by Mycobacterium tuberculosis. J Mol Med. 2007;85:613–621.
    1. Berry MPR, Graham CM, McNab FW, Xu Z, Bloch SAA, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–977.
    1. Maertzdorf J, Repsilber D, Parida SK, Stanley K, Roberts T, et al. Human gene expression profiles of susceptibility and resistance in tuberculosis. Genes Immun. 2011;12:15–22.
    1. Breiman L. Random forests. Mach Learn. 2001;45:5–32.
    1. Lê Cao K-A, González I, Déjean S. integrOmics: an R package to unravel relationships between two omics datasets. Bioinformatics. 2009;25:2855.
    1. Mellor AL, Munn DH. IDO expression by dendritic cells: tolerance and tryptophan catabolism. Nature Rev Immunol. 2004;4:762–774.
    1. Munn DH, Zhou M, Attwood JT, Bondarev I, Conway SJ, et al. Prevention of allogeneic fetal rejection by tryptophan catabolism. Science. 1998;281:1191.
    1. Munn DH, Sharma MD, Hou D, Baban B, Lee JR, et al. Expression of indoleamine 2, 3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest. 2004;114:280–290.
    1. Katz JB, Muller AJ, Prendergast GC. Indoleamine 2, 3-dioxygenase in T-cell tolerance and tumoral immune escape. Immunol Rev. 2008;222:206–221.
    1. Popov A, Abdullah Z, Wickenhauser C, Saric T, Driesen J, et al. Indoleamine 2, 3-dioxygenase-expressing dendritic cells form suppurative granulomas following Listeria monocytogenes infection. J Clin Invest. 2006;116:3160.
    1. Boasso A, Herbeuval JP, Hardy AW, Anderson SA, Dolan MJ, et al. HIV inhibits CD4+ T-cell proliferation by inducing indoleamine 2, 3-dioxygenase in plasmacytoid dendritic cells. Blood. 2007;109:3351.
    1. Romani L, Fallarino F, De Luca A, Montagnoli C, D’Angelo C, et al. Defective tryptophan catabolism underlies inflammation in mouse chronic granulomatous disease. Nature. 2008;451:211–215.
    1. Favre D, Mold J, Hunt PW, Kanwar B, Loke P, et al. Tryptophan catabolism by indoleamine 2, 3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Science Trans Med. 2010;2:32ra36.
    1. Makala LHC, Baban B, Lemos H, El-Awady AR, Chandler PR, et al. Leishmania major attenuates host immunity by stimulating local indoleamine 2, 3-dioxygenase expression. J Infect Dis. 2011;203:715.
    1. Platten M, Ho PP, Youssef S, Fontoura P, Garren H, et al. Treatment of autoimmune neuroinflammation with a synthetic tryptophan metabolite. Science. 2005;310:850.
    1. Sharma MD, Hou DY, Liu Y, Koni PA, Metz R, et al. Indoleamine 2, 3-dioxygenase controls conversion of Foxp3+ Tregs to TH17-like cells in tumor-draining lymph nodes. Blood. 2009;113:6102.
    1. Sharma MD, Hou DY, Baban B, Koni PA, He Y, et al. Reprogrammed Foxp3+ regulatory T cells provide essential help to support cross-presentation and CD8+ T cell priming in naive mice. Immunity. 2010;33:942–954.
    1. Müller A, Heseler K, Schmidt SK, Spekker K, MacKenzie CR, et al. The missing link between indoleamine 2, 3-dioxygenase mediated antibacterial and immunoregulatory effects. J Cell Mol Med. 2009;13:1125–1135.
    1. Grohmann U, Volpi C, Fallarino F, Bozza S, Bianchi R, et al. Reverse signaling through GITR ligand enables dexamethasone to activate IDO in allergy. Nature Med. 2007;13:579–586.
    1. Suzuki Y, Suda T, Asada K, Miwa S, Suzuki M, et al. Serum indoleamine 2, 3-dioxygenase activity predicts prognosis of pulmonary tuberculosis. Clinical and Vaccine Immunology. 2012.
    1. Mehta D, Gupta S, Gaur SN, Gangal SV, Agrawal KP. Increased leukocyte phospholipase A2 activity and plasma lysophosphatidylcholine levels in asthma and rhinitis and their relationship to airway sensitivity to histamine. Am Rev Resp Dis. 1990;142:157–161.
    1. Okita M, Gaudette DC, Mills GB, Holub BJ. Elevated levels and altered fatty acid composition of plasma lysophosphatidlycholine(lysoPC) in ovarian cancer patients. Int J Cancer. 1997;71:31–34.
    1. Taylor L, Arends J, Hodina A, Unger C, Massing U. Plasma lyso-phosphatidylcholine concentration is decreased in cancer patients with weight loss and activated inflammatory status. Lipids Health Dis. 2007;6:17.
    1. Drobnik W, Liebisch G, Audebert FX, Frohlich D, Gluck T, et al. Plasma ceramide and lysophosphatidylcholine inversely correlate with mortality in sepsis patients. J Lipid Res. 2003;44:754–761.
    1. Duan L, Gan H, Arm J, Remold HG. Cytosolic Phospholipase A2 Participates with TNF-{{alpha}} in the Induction of Apoptosis of Human Macrophages Infected with Mycobacterium tuberculosis H37Ra. J Immunol. 2001;166:7469–7476.
    1. Lewis GD, Wei RU, Liu E, Yang E, Shi X, et al. Metabolite profiling of blood from individuals undergoing planned myocardial infarction reveals early markers of myocardial injury. J Clin Invest. 2008;118:3503.
    1. Bell MJ, Kochanek PM, Carcillo JA, Mi Z, Schiding JK, et al. Interstitial adenosine, inosine, and hypoxanthine are increased after experimental traumatic brain injury in the rat. J Neurotraum. 1998;15:163–170.
    1. Jin X, Shepherd RK, Duling BR, Linden J. Inosine binds to A3 adenosine receptors and stimulates mast cell degranulation. J Clin Invest. 1997;100:2849–2857.
    1. Gomez G, Sitkovsky MV. Differential requirement for A2a and A3 adenosine receptors for the protective effect of inosine in vivo. Blood. 2003;102:4472–4478.
    1. Den Boon S, Van Lill SWP, Borgdorff MW, Verver S, Bateman ED, et al. Association between smoking and tuberculosis infection: a population survey in a high tuberculosis incidence area. Thorax. 2005;60:555–557.
    1. Dever JT, Elfarra AA. In Vivo Metabolism of l-Methionine in Mice: Evidence for Stereoselective Formation of Methionine-d-Sulfoxide and Quantitation of Other Major Metabolites. Drug Metab Dispos. 2006;34:2036–2043.
    1. Avila MA, Carretero MV, Rodriguez EN, Mato JM. Regulation by hypoxia of methionine adenosyltransferase activity and gene expression in rat hepatocytes. Gastroenterology. 1998;114:364–371.
    1. Kono H, Enomoto N, Connor HD, Wheeler MD, Bradford BU, et al. Medium-chain triglycerides inhibit free radical formation and TNF-alpha production in rats given enteral ethanol. Am J Physiol Gastrointest Liver Physiol. 2000;278:G467–G476.
    1. Harrison RA, Farries TC, Northrop FD, Lachmann PJ, Davis AE. Structure of C3f, a small peptide specifically released during inactivation of the third component of complement. Complement. 1988;5:27–32.
    1. Sakamoto M, Fujisawa Y, Nishioka K. Physiologic role of the complement system in host defense, disease, and malnutrition. Nutrition. 1998;14:391–398.
    1. Szeplaki G, Prohaszka Z, Duba J, Rugonfalvi-Kiss S, Karadi I, et al. 2004. Association of high serum concentration of the third component of complement (C3) with pre-existing severe coronary artery disease and new vascular events in women. Atherosclerosis 177: 383–389. S0021–9150(04)00394–6 [pii];10.1016/j.atherosclerosis.2004.07.022 [doi]
    1. Chan TYK. Vitamin D Deficiency and Susceptibility to Tuberculosis. Calcified Tissue Int. 2000;66:476–478.
    1. Liu PT, Stenger S, Li H, Wenzel L, Tan BH, et al. Toll-like receptor triggering of a vitamin D-mediated human antimicrobial response. Science. 2006;311:1770.
    1. Wilkinson RJ, Llewelyn M, Toossi Z, Patel P, Pasvol G, et al. Influence of vitamin D deficiency and vitamin D receptor polymorphisms on tuberculosis among Gujarati Asians in west London: a case-control study. Lancet. 2000;355:618–621.
    1. Ustianowski A, Shaffer R, Collin S, Wilkinson RJ, Davidson RN. Prevalence and associations of vitamin D deficiency in foreign-born persons with tuberculosis in London. J Infection. 2005;50:432–437.
    1. Cohen G, Rudnicki M, Hoorl WH. Uremic toxins modulate the spontaneous apoptotic cell death and essential functions of neutrophils. Kidney Int. 2001;59:S48–S52.
    1. Hutchings A, van Staden J. Plants used for stress-related ailments in traditional Zulu, Xhosa and Sotho medicine. Part 1: Plants used for headaches. J Ethnopharmacol 43: 89–124. 0378–8741(94)90008–6 [pii] 1994.
    1. Chang WK, Yang KD, Shaio MF. Lymphocyte proliferation modulated by glutamine: involved in the endogenous redox reaction. Clin Exp Immunol. 1999;117:482–488.
    1. Parry-Billings M, Calder PC, Newsholme EA, Evans J. Does glutamine contribute to immunosuppression after major burns? Lancet. 1990;336:523–525.
    1. Kinscherf R, Hack V, Fischbach T, Friedmann B, Weiss C, et al. Low plasma glutamine in combination with high glutamate levels indicate risk for loss of body cell mass in healthy individuals: the effect of N-acetyl-cysteine. J Mol Med. 1996;74:393–400.
    1. Huettmann A, Duehrsen U, Stypmann J, Noppeney R, Nueckel H, et al. Granulocyte colony-stimulating factor-induced blood stem cell mobilisation in patients with chronic heart failure. Basic Res Cardiol. 2006;101:78–86.
    1. To LB, Haylock DN, Simmons PJ, Juttner CA. The biology and clinical uses of blood stem cells. Blood. 1997;89:2233.
    1. Schwenk A, Macallan DC. Tuberculosis, malnutrition and wasting. Curr Opin Clin Nutr Metab Care. 2000;3:285.
    1. Evans AM, DeHaven CD, Barrett T, Mitchell M, Milgram E. Integrated, nontargeted ultrahigh performance liquid chromatography/electrospray ionization tandem mass spectrometry platform for the identification and relative quantification of the small-molecule complement of biological systems. Anal Chem. 2009;81:6656–6667.
    1. Sha W, da Costa KA, Fischer LM, Milburn MV, Lawton KA, et al. Metabolomic profiling can predict which humans will develop liver dysfunction when deprived of dietary choline. FASEB J. 2010;24:2962–2975.
    1. DeHaven C, Evans A, Dai H, Lawton K. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. Journal of cheminformatics. 2010;2:9.
    1. Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, et al. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol. 2003;4:1206–1212.
    1. Smyth GK. Limma: linear models for microarray data. In: Bioinformatics and Computational Biology Solutions Using R and Bioconductor. New York: Springer. 2005. pp. 397–420.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Stat Soc B Met. 1995;57:289–300.
    1. Liaw A, Wiener M. Classification and Regression by randomForest. R news. 2002;2:18–22.
    1. Suzuki R, Shimodaira H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics. 2006;22:1540–1542.

Source: PubMed

3
Abonneren