Human Serum Metabolites Associate With Severity and Patient Outcomes in Traumatic Brain Injury

Matej Orešič, Jussi P Posti, Maja H Kamstrup-Nielsen, Riikka S K Takala, Hester F Lingsma, Ismo Mattila, Sirkku Jäntti, Ari J Katila, Keri L H Carpenter, Henna Ala-Seppälä, Anna Kyllönen, Henna-Riikka Maanpää, Jussi Tallus, Jonathan P Coles, Iiro Heino, Janek Frantzén, Peter J Hutchinson, David K Menon, Olli Tenovuo, Tuulia Hyötyläinen, Matej Orešič, Jussi P Posti, Maja H Kamstrup-Nielsen, Riikka S K Takala, Hester F Lingsma, Ismo Mattila, Sirkku Jäntti, Ari J Katila, Keri L H Carpenter, Henna Ala-Seppälä, Anna Kyllönen, Henna-Riikka Maanpää, Jussi Tallus, Jonathan P Coles, Iiro Heino, Janek Frantzén, Peter J Hutchinson, David K Menon, Olli Tenovuo, Tuulia Hyötyläinen

Abstract

Traumatic brain injury (TBI) is a major cause of death and disability worldwide, especially in children and young adults. TBI is an example of a medical condition where there are still major lacks in diagnostics and outcome prediction. Here we apply comprehensive metabolic profiling of serum samples from TBI patients and controls in two independent cohorts. The discovery study included 144 TBI patients, with the samples taken at the time of hospitalization. The patients were diagnosed as severe (sTBI; n=22), moderate (moTBI; n=14) or mild TBI (mTBI; n=108) according to Glasgow Coma Scale. The control group (n=28) comprised of acute orthopedic non-brain injuries. The validation study included sTBI (n=23), moTBI (n=7), mTBI (n=37) patients and controls (n=27). We show that two medium-chain fatty acids (decanoic and octanoic acids) and sugar derivatives including 2,3-bisphosphoglyceric acid are strongly associated with severity of TBI, and most of them are also detected at high concentrations in brain microdialysates of TBI patients. Based on metabolite concentrations from TBI patients at the time of hospitalization, an algorithm was developed that accurately predicted the patient outcomes (AUC=0.84 in validation cohort). Addition of the metabolites to the established clinical model (CRASH), comprising clinical and computed tomography data, significantly improved prediction of patient outcomes. The identified 'TBI metabotype' in serum, that may be indicative of disrupted blood-brain barrier, of protective physiological response and altered metabolism due to head trauma, offers a new avenue for the development of diagnostic and prognostic markers of broad spectrum of TBIs.

Keywords: Biomarkers; Mass spectrometry; Metabolomics; Traumatic brain injury.

Copyright © 2016. Published by Elsevier B.V.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Overview of the workflow to study metabolome in traumatic brain injury (TBI). (a) Serum metabolomics was performed in a series of samples from TBI patients and controls from the city of Turku, Finland. Significant metabolites associated with severity of TBI were identified. (b) The results from the Turku series were compared with metabolomics data from TBI patients and controls from the city of Cambridge, UK. (c) Brain microdialysate (BMD) samples were also analyzed from the severe TBI patients in Cambridge, for possible associations of BMD metabolite concentrations with the changes observed in serum. (d) In order to assess if metabolites can serve as predictor of outcomes in TBI patients, metabolites from the Turku dataset were screened for associations with patient outcomes. Predictive models were developed, which were independently tested in the Cambridge dataset.
Fig. 2
Fig. 2
Serum metabolome is associated with severity of TBI. (a) Principal Component Analysis (PCA) scores for the first two principal components (out of seven), using the dataset comprising 98 metabolites with FDR q t-test two sided unequal variance). (d) Scatter plot of log2 scaled mean metabolite levels in sTBI vs. mTBI patients, with significant metabolites in both groups marked with full circles (t-test two sided unequal variance). Regression lines in panels c and d are drawn based on significant metabolites in both groups. In panels b-d, the selected unidentified metabolites are listed in italic and annotated according to their structural class (Castillo et al., 2011). Abbreviations: 2,3-BPG, 2,3-bisphosphoglycerate; α-KG, alpha-ketoglutarate; DA, decanoic acid; IPA, indole-3-propionic acid; moTBI, moderate TBI; mTBI, mild TBI; OA, octanoic acid; PC, principal component; sTBI, severe TBI.
Fig. 3
Fig. 3
Serum metabolites found associated with TBI display similar associations in an independent cohort and are found enriched in brain microdialysates. (a) Scatter plot of log2 scaled mean metabolite levels in sTBI patients from the cities of Cambridge vs. Turku, with significant metabolites in both groups marked with full circles. Regression line is drawn based on significant metabolites in both groups. (b) Comparison of mean serum metabolite level changes (sTBI vs. Controls; Turku) and mean brain microdialysate (BMD) metabolite levels (normalized to mean serum metabolite levels in Controls from Turku), shown as a scatter plot of all metabolites detected both in serum and BMD. The metabolites that are found significantly associated with TBI in serum (dark blue circles) display a close association with BMD levels, while the non-associated metabolites (light blue circles) do not.
Fig. 4
Fig. 4
Serum metabolite levels at the time of hospitalization predict the patient outcomes in TBI. Performance of the model to predict patient outcomes in TBI, shown as ROC curves for the training (Turku) and validation (Cambridge) data. Abbreviation: AUC, area under the ROC curve.

References

    1. Anderson V.A., Catroppa C., Haritou F., Morse S., Pentland L., Rosenfeld J., Stargatt R. Predictors of acute child and family outcome following traumatic brain injury in children. Pediatr. Neurosurg. 2001;34:138–148.
    1. Berger R.P., Dulani T., Adelson P.D., Leventhal J.M., Richichi R., Kochanek P.M. Identification of inflicted traumatic brain injury in well-appearing infants using serum and cerebrospinal markers: a possible screening tool. Pediatrics. 2006;117:325–332.
    1. Brain Trauma Foundation, American Association of Neurological Surgeons, Congress of Neurological Surgeons, Joint Section on Neurotrauma, Critical Care Aans Cns, Carney N.A., Ghajar J. Guidelines for the management of severe traumatic brain injury. Introduction. J. Neurotrauma. 2007;24(Suppl. 1):S1–S2.
    1. Castillo S., Mattila I., Miettinen J., Oresic M., Hyotylainen T. Data analysis tool for comprehensive two-dimensional gas chromatography/time-of-flight mass spectrometry. Anal. Chem. 2011;83:3058–3067.
    1. Chyan Y.J., Poeggeler B., Omar R.A., Chain D.G., Frangione B., Ghiso J., Pappolla M.A. Potent neuroprotective properties against the Alzheimer beta-amyloid by an endogenous melatonin-related indole structure, indole-3-propionic acid. J. Biol. Chem. 1999;274:21937–21942.
    1. Clausen F., Hillered L., Gustafsson J. Cerebral glucose metabolism after traumatic brain injury in the rat studied by 13C-glucose and microdialysis. Acta Neurochir. 2011;153:653–658.
    1. Coles J.P., Fryer T.D., Smielewski P., Chatfield D.A., Steiner L.A., Johnston A.J., Downey S.P., Williams G.B., Aigbirhio F., Hutchinson P.J. Incidence and mechanisms of cerebral ischemia in early clinical head injury. J. Cereb. Blood Flow Metab. 2004;24:202–211.
    1. Crash Trial Collaborators M.R.C., Perel P., Arango M., Clayton T., Edwards P., Komolafe E., Poccock S., Roberts I., Shakur H., Steyerberg E. Predicting outcome after traumatic brain injury: practical prognostic models based on large cohort of international patients. BMJ. 2008;336:425–429.
    1. Ebert D., Haller R.G., Walton M.E. Energy contribution of octanoate to intact rat brain metabolism measured by 13C nuclear magnetic resonance spectroscopy. J. Neurosci. 2003;23:5928–5935.
    1. Glenn T.C., Kelly D.F., Boscardin W.J., McArthur D.L., Vespa P., Oertel M., Hovda D.A., Bergsneider M., Hillered L., Martin N.A. Energy dysfunction as a predictor of outcome after moderate or severe head injury: indices of oxygen, glucose, and lactate metabolism. J. Cereb. Blood Flow Metab. 2003;23:1239–1250.
    1. Green D.R., Reed J.C. Mitochondria and apoptosis. Science. 1998;281:1309–1312.
    1. Harrell F.E. second ed. Springer; New York: 2015. Regression Modeling Strategies: With Applications to Linear Models, Logistic and Ordinal Regression, and Survival Analysis.
    1. Hartonen M., Mattila I., Ruskeepää A.-L., Orešič M., Hyötyläinen T. Characterization of cerebrospinal fluid by comprehensive two-dimensional gas chromatography coupled to time-of-flight mass spectrometry. J. Chromatogr. A. 2013
    1. Hovda D.A., Becker D.P., Katayama Y. Secondary injury and acidosis. J. Neurotrauma. 1992;9(Suppl. 1):S47–S60.
    1. Hsia C.C. Respiratory function of hemoglobin. N. Engl. J. Med. 1998;338:239–247.
    1. Hwang I.K., Yoo K.Y., Li H., Park O.K., Lee C.H., Choi J.H., Jeong Y.G., Lee Y.L., Kim Y.M., Kwon Y.G. Indole-3-propionic acid attenuates neuronal damage and oxidative stress in the ischemic hippocampus. J. Neurosci. Res. 2009;87:2126–2137.
    1. Ingebrigtsen T., Romner B. Biochemical serum markers of traumatic brain injury. J. Trauma. 2002;52:798–808.
    1. Jeter C.B., Hergenroeder G.W., Ward N.H., III, Moore A.N., Dash P.K. Human mild traumatic brain injury decreases circulating branched-chain amino acids and their metabolite levels. J. Neurotrauma. 2013;30:671–679.
    1. Karbownik M., Reiter R.J., Garcia J.J., Cabrera J., Burkhardt S., Osuna C., Lewinski A. Indole-3-propionic acid, a melatonin-related molecule, protects hepatic microsomal membranes from iron-induced oxidative damage: relevance to cancer reduction. J. Cell. Biochem. 2001;81:507–513.
    1. Koh S.X., Lee J.K. S100B as a marker for brain damage and blood-brain barrier disruption following exercise. Sports Med. 2014;44:369–385.
    1. Kopka J., Schauer N., Krueger S., Birkemeyer C., Usadel B., Bergmuller E., Dormann P., Weckwerth W., Gibon Y., Stitt M. GMD@CSB.DB: the Golm Metabolome Database. Bioinformatics. 2005;21:1635–1638.
    1. Lakshmanan R., Loo J.A., Drake T., Leblanc J., Ytterberg A.J., McArthur D.L., Etchepare M., Vespa P.M. Metabolic crisis after traumatic brain injury is associated with a novel microdialysis proteome. Neurocrit. Care. 2010;12:324–336.
    1. Maas A.I., Stocchetti N., Bullock R. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 2008;7:728–741.
    1. Maas A.I., Menon D.K., Steyerberg E.W., Citerio G., Lecky F., Manley G.T., Hill S., Legrand V., Sorgner A., Participants C.-T. Collaborative european neurotrauma effectiveness research in traumatic brain injury (CENTER-TBI): a prospective longitudinal observational study. Neurosurgery. 2015;76:67–80.
    1. Menon D.K., Coles J.P., Gupta A.K., Fryer T.D., Smielewski P., Chatfield D.A., Aigbirhio F., Skepper J.N., Minhas P.S., Hutchinson P.J. Diffusion limited oxygen delivery following head injury. Crit. Care Med. 2004;32:1384–1390.
    1. Mondello S., Papa L., Buki A., Bullock M.R., Czeiter E., Tortella F.C., Wang K.K., Hayes R.L. Neuronal and glial markers are differently associated with computed tomography findings and outcome in patients with severe traumatic brain injury: a case control study. Crit. Care. 2011;15:R156.
    1. Oldendorf W.H. Carrier-mediated blood-brain barrier transport of short-chain monocarboxylic organic acids. Am. J. Phys. 1973;224:1450–1453.
    1. Paik M.J., Li W.Y., Ahn Y.H., Lee P.H., Choi S., Kim K.R., Kim Y.M., Bang O.Y., Lee G. The free fatty acid metabolome in cerebral ischemia following human mesenchymal stem cell transplantation in rats. Clin. Chim. Acta. 2009;402:25–30.
    1. Papa L., Akinyi L., Liu M.C., Pineda J.A., Tepas J.J., III, Oli M.W., Zheng W., Robinson G., Robicsek S.A., Gabrielli A. Ubiquitin C-terminal hydrolase is a novel biomarker in humans for severe traumatic brain injury. Crit. Care Med. 2010;38:138–144.
    1. Pelinka L.E., Kroepfl A., Leixnering M., Buchinger W., Raabe A., Redl H. GFAP versus S100B in serum after traumatic brain injury: relationship to brain damage and outcome. J. Neurotrauma. 2004;21:1553–1561.
    1. Reis de Assis D., Maria Rde C., Borba Rosa R., Schuck P.F., Ribeiro C.A., da Costa Ferreira G., Dutra-Filho C.S., Terezinha de Souza Wyse A., Duval Wannmacher C.M., Santos Perry M.L. Inhibition of energy metabolism in cerebral cortex of young rats by the medium-chain fatty acids accumulating in MCAD deficiency. Brain Res. 2004;1030:141–151.
    1. Saatman K.E., Duhaime A.C., Bullock R., Maas A.I., Valadka A., Manley G.T. Classification of traumatic brain injury for targeted therapies. J. Neurotrauma. 2008;25:719–738.
    1. Savola O., Pyhtinen J., Leino T.K., Siitonen S., Niemela O., Hillbom M. Effects of head and extracranial injuries on serum protein S100B levels in trauma patients. J. Trauma. 2004;56:1229–1234. (discussion 1234)
    1. Saw M.M., Chamberlain J., Barr M., Morgan M.P., Burnett J.R., Ho K.M. Differential disruption of blood-brain barrier in severe traumatic brain injury. Neurocrit. Care. 2014;20:209–216.
    1. Schuck P.F., Ferreira G.C., Moura A.P., Busanello E.N., Tonin A.M., Dutra-Filho C.S., Wajner M. Medium-chain fatty acids accumulating in MCAD deficiency elicit lipid and protein oxidative damage and decrease non-enzymatic antioxidant defenses in rat brain. Neurochem. Int. 2009;54:519–525.
    1. Schuck P.F., Ferreira Gda C., Tonin A.M., Viegas C.M., Busanello E.N., Moura A.P., Zanatta A., Klamt F., Wajner M. Evidence that the major metabolites accumulating in medium-chain acyl-CoA dehydrogenase deficiency disturb mitochondrial energy homeostasis in rat brain. Brain Res. 2009;1296:117–126.
    1. Spector R. Fatty acid transport through the blood-brain barrier. J. Neurochem. 1988;50:639–643.
    1. Storey J.D. A direct approach to false discovery rates. J. R. Stat. Soc. Ser. B Stat Methodol. 2002;64:479–498.
    1. Takala R.S., Posti J.P., Runtti H., Newcombe V.F., Outtrim J., Katila A.J., Frantzen J., Ala-Seppala H., Kyllonen A., Maanpaa H.R. Glial fibrillary acidic protein and ubiquitin C-terminal hydrolase-L1 as outcome predictors in traumatic brain injury. World Neurosurg. 2016;87:8–20.
    1. Timofeev I., Carpenter K.L., Nortje J., Al-Rawi P.G., O'Connell M.T., Czosnyka M., Smielewski P., Pickard J.D., Menon D.K., Kirkpatrick P.J. Cerebral extracellular chemistry and outcome following traumatic brain injury: a microdialysis study of 223 patients. Brain. 2011;134:484–494.
    1. Torabian S., Kashani-Sabet M. Biomarkers for melanoma. Curr. Opin. Oncol. 2005;17:167–171.
    1. Vos P.E., Lamers K.J., Hendriks J.C., van Haaren M., Beems T., Zimmerman C., van Geel W., de Reus H., Biert J., Verbeek M.M. Glial and neuronal proteins in serum predict outcome after severe traumatic brain injury. Neurology. 2004;62:1303–1310.
    1. Wikoff W.R., Anfora A.T., Liu J., Schultz P.G., Lesley S.A., Peters E.C., Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. U. S. A. 2009;106:3698–3703.
    1. Wilson J.T., Pettigrew L.E., Teasdale G.M. Structured interviews for the Glasgow outcome scale and the extended Glasgow outcome scale: guidelines for their use. J. Neurotrauma. 1998;15:573–585.
    1. Wolf A., Weir P., Segar P., Stone J., Shield J. Impaired fatty acid oxidation in propofol infusion syndrome. Lancet. 2001;357:606–607.

Source: PubMed

3
Subscribe