A systems-level "misunderstanding": the plasma metabolome in Huntington's disease

Herminia D Rosas, Gheorghe Doros, Swati Bhasin, Beena Thomas, Sona Gevorkian, Keith Malarick, Wayne Matson, Steven M Hersch, Herminia D Rosas, Gheorghe Doros, Swati Bhasin, Beena Thomas, Sona Gevorkian, Keith Malarick, Wayne Matson, Steven M Hersch

Abstract

Objective: Huntington's disease (HD) is a rare neurodegenerative disease caused by the expansion of an N-terminal repeat in the huntingtin protein. The protein is expressed in all cells in the body; hence, peripheral tissues, such as blood, may recapitulate processes in the brain. The plasma metabolome may provide a window into active processes that influence brain health and a unique opportunity to noninvasively identify processes that may contribute to neurodegeneration. Alterations in metabolic pathways in brain have been shown to profoundly impact HD. Therefore, identification and quantification of critical metabolomic perturbations could provide novel biomarkers for disease onset and disease progression.

Methods: We analyzed the plasma metabolomic profiles from 52 premanifest (PHD), 102 early symptomatic HD, and 140 healthy controls (NC) using liquid chromatography coupled with a highly sensitive electrochemical detection platform.

Results: Alterations in tryptophan, tyrosine, purine, and antioxidant pathways were identified, including many related to energetic and oxidative stress and derived from the gut microbiome. Multivariate statistical modeling demonstrated mutually distinct metabolomic profiles, suggesting that the processes that determine onset were likely distinct from those that determine progression. Gut microbiome-derived metabolites particularly differentiated the PHD metabolome, while the symptomatic HD metabolome was increasingly influenced by metabolites that may reflect mutant huntingtin toxicity and neurodegeneration.

Interpretation: Understanding the complex changes in the delicate balance of the metabolome and the gut microbiome in HD, and how they relate to disease onset, progression, and phenotypic variability in HD are critical questions for future research.

Figures

Figure 1
Figure 1
Receiver operator curves. (A) NC versus PHD. (B) NC versus HD. (C) PHD versus HD. In each case, the AUC demonstrated excellent sensitivity and specificity, demonstrating clear separation between groups and suggesting specific effects on the HD plasma metabolome. AUC, area under the curve.
Figure 2
Figure 2
One out testing scores using PLSDA models for NC versus PHD, NC versus HD and PHD versus HD, using the 29 chosen compounds from the tyrosine, tryptophan, purine pathways and markers of oxidative progression (from Table1). The CCR are summarized in Table3. PLSDA, partial least squares discriminant analysis; HD, Huntington’s disease; CCR, correct classification rates.
Figure 3
Figure 3
Heat map demonstrating altered correlations in the cross-metabolite correlations. Red demonstrates a positive correlation, blue a negative correlation, between pairs of metabolites.
Figure 4
Figure 4
Network graph representing Pearson correlations between metabolites for NC, PHD, and HD groups (r = ±0.7 to ±0.2) for each pathway. Red lines represent negative correlations while blue lines represent positive correlations. These results demonstrate altered relationships amongst metabolites across disease groups, suggesting unique alterations in the feedback control of key enzymatic processes that change with HD progression.
Figure 5
Figure 5
Summation of one-out scoring. The graph is presented with 0.5 subtracted from all values to center around 0 and scaled by a factor of 100 for visualization. From the left, the first two columns score NC versus HD with NC as category A. The third and fourth columns score NC versus PHD with NC as category A. The fifth and sixth columns score HD versus PHD with HD as category A.
Figure 6
Figure 6
PLSDA modeling of effects of medications on metabolomics profiling. (A) There was no difference in the metabolomic profiles of PHD and HD individuals taking SSRI’s (Red) and those who were not on SSRI’s (Green). (B) There was no difference in the metabolomic profiles of PHD and HD individuals taking Neuroleptics (Green) and those not on neuroleptics (Red). This suggests that the HD metabotype is sufficiently robust to be insignificantly affected by these medications.

References

    1. Beal MF, Matson WR, Swartz KJ, et al. Kynurenine pathway measurements in Huntington’s disease striatum: evidence for reduced formation of kynurenic acid. J Neurochem. 1990;55:1327–1339.
    1. Stoy N, Mackay GM, Forrest CM, et al. Tryptophan metabolism and oxidative stress in patients with Huntington’s disease. J Neurochem. 2005;93:611–623.
    1. Pearson SJ, Reynolds GP. Increased brain concentrations of a neurotoxin, 3-hydroxykynurenine, in Huntington’s disease. Neurosci Lett. 1992;144:199–201.
    1. Kristal BS, Shurubor YI, Kaddurah-Daouk R, Matson WR. Metabolomics in the study of aging and caloric restriction. Methods Mol Biol. 2007;371:393–409.
    1. Kristal BS, Shurubor YI, Kaddurah-Daouk R, Matson WR. High-performance liquid chromatography separations coupled with coulometric electrode array detectors: a unique approach to metabolomics. Methods Mol Biol. 2007;358:159–174.
    1. Kristal BS, Vigneau-Callahan KE, Matson WR. Simultaneous analysis of the majority of low-molecular-weight, redox-active compounds from mitochondria. Anal Biochem. 1998;263:18–25.
    1. Kristal BS, Vigneau-Callahan KE, Moskowitz AJ, Matson WR. Purine catabolism: links to mitochondrial respiration and antioxidant defenses? Arch Biochem Biophys. 1999;370:22–33.
    1. Schiavo S, Ebbel E, Sharma S, et al. Metabolite identification using a nanoelectrospray LC-EC-array-MS integrated system. Anal Chem. 2008;80:5912–5923.
    1. Shi H, Paolucci U, Vigneau-Callahan KE, et al. Development of biomarkers based on diet-dependent metabolic serotypes: practical issues in development of expert system-based classification models in metabolomic studies. OMICS. 2004;8:197–208.
    1. Shurubor YI, Matson WR, Willett WC, et al. Biological variability dominates and influences analytical variance in HPLC-ECD studies of the human plasma metabolome. BMC Clin Pathol. 2007;7:9.
    1. Paolucci U, Vigneau-Callahan KE, Shi H, et al. Development of biomarkers based on diet-dependent metabolic serotypes: concerns and approaches for cohort and gender issues in serum metabolome studies. OMICS. 2004;8:209–220.
    1. Rozen S, Cudkowicz ME, Bogdanov M, et al. Metabolomic analysis and signatures in motor neuron disease. Metabolomics. 2005;1:101–108.
    1. Bogdanov M, Matson WR, Wang L, et al. Metabolomic profiling to develop blood biomarkers for Parkinson’s disease. Brain. 2008;131:389–396.
    1. Johansen KK, Wang L, Aasly JO, et al. Metabolomic profiling in LRRK2-related Parkinson’s disease. PLoS One. 2009;4:e7551.
    1. Kaddurah-Daouk R, Boyle SH, Matson W, et al. Pretreatment metabotype as a predictor of response to sertraline or placebo in depressed outpatients: a proof of concept. Transl Psychiat. 2011;1:e26.
    1. Harrell FE. Regression modeling strategies: with applications to linear models, logistic regression, and survival analysis. New York, NY: Springer; 2001. p. xxii. Springer series in statistics. 568 p.
    1. Steyerberg EW. SpringerLink (online service) In: Gail M, Samet JM, Tsiatis A, Wong W, editors. Statistics for biology and health. New York, NY: Springer; 2009. p. xxviii. 497 p.
    1. Efron B. How biased is the apparent error rate of a prediction rule? J Am Stat Assoc. 1986;81:461–470.
    1. Wikoff WR, Anfora AT, Liu J, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci USA. 2009;106:3698–3703.
    1. Vaarala O, Atkinson MA, Neu J. The “perfect storm” for type 1 diabetes: the complex interplay between intestinal microbiota, gut permeability, and mucosal immunity. Diabetes. 2008;57:2555–2562.
    1. King C, Sarvetnick N. The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions. PLoS One. 2011;6:e17049.
    1. Soulet D, Cicchetti F. The role of immunity in Huntington’s disease. Mol Psychiatry. 2011;16:889–902.
    1. De Ponti F. Drug development for the irritable bowel syndrome: current challenges and future perspectives. Front Pharmacol. 2013;4:7.
    1. Politis M, Su P, Piccini P. Imaging of microglia in patients with neurodegenerative disorders. Front Pharmacol. 2012;3:96.
    1. Bjorkqvist M, Wild EJ, Thiele J, et al. A novel pathogenic pathway of immune activation detectable before clinical onset in Huntington’s disease. J Exp Med. 2008;205:1869–1877.
    1. Nicholson JK, Holmes E, Kinross J, et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262–1267. published online EpubJun 8.
    1. Resta SC. Effects of probiotics and commensals on intestinal epithelial physiology: implications for nutrient handling. J Physiol. 2009;587:4169–4174.
    1. Pauley RJ, Fredricks WW, Smith OH. Effect of tryptophan analogs on derepression of the Escherichia coli tryptophan operon by indole-3-propionic acid. J Bacteriol. 1978;136:219–226.
    1. Chyan YJ, Poeggeler B, Omar RA, et al. 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. Karbownik M, Reiter RJ, Garcia JJ, et al. 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. Bendheim PE, Poeggeler B, Neria E, et al. Development of indole-3-propionic acid (OXIGON) for Alzheimer’s disease. J Mol Neurosci. 2002;19:213–217.
    1. Coyle JT, Schwarcz R. Lesion of striatal neurones with kainic acid provides a model for Huntington’s chorea. Nature. 1976;263:244–246.
    1. Thevandavakkam MA, Schwarcz R, Muchowski PJ, Giorgini F. Targeting kynurenine 3-monooxygenase (KMO): implications for therapy in Huntington’s disease. CNS Neurol Disord Drug Targets. 2010;9:791–800.
    1. Zwilling D, Huang SY, Sathyasaikumar KV, et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. 2011;145:863–874.
    1. Beal MF, Matson WR, Storey E, et al. Kynurenic acid concentrations are reduced in Huntington’s disease cerebral cortex. J Neurol Sci. 1992;108:80–87.
    1. Zhou H, Wang J, Jiang J, et al. N-acetyl-serotonin offers neuroprotection through inhibiting mitochondrial death pathways and autophagic activation in experimental models of ischemic injury. J Neurosci. 2014;34:2967–2978.
    1. Cardinali DP, Pagano ES, Scacchi Bernasconi PA, et al. Melatonin and mitochondrial dysfunction in the central nervous system. Horm Behav. 2013;63:322–330.
    1. Aziz NA, Pijl H, Frolich M, et al. Delayed onset of the diurnal melatonin rise in patients with Huntington’s disease. J Neurol. 2009;256:1961–1965.
    1. Christofides J, Bridel M, Egerton M, et al. Blood 5-hydroxytryptamine, 5-hydroxyindoleacetic acid and melatonin levels in patients with either Huntington’s disease or chronic brain injury. J Neurochem. 2006;97:1078–1088.
    1. Mohamed Mel S, Ismail W, Heider J, Fuchs G. Aerobic metabolism of phenylacetic acids in Azoarcus evansii. Arch Microbiol. 2002;178:180–192.
    1. Mu W, Yang Y, Jia J, et al. Production of 4-hydroxyphenyllactic acid by Lactobacillus sp. SK007 fermentation. J Biosci Bioeng. 2010;109:369–371.
    1. Petersen M, Abdullah Y, Benner J, et al. Evolution of rosmarinic acid biosynthesis. Phytochemistry. 2009;70:1663–1679. doi: ; published online Epub Oct-Nov.
    1. Beloborodova N, Bairamov I, Olenin A, et al. Effect of phenolic acids of microbial origin on production of reactive oxygen species in mitochondria and neutrophils. J Biomed Sci. 2012;19:89.
    1. Kayden HJ, Traber MG. Absorption, lipoprotein transport, and regulation of plasma concentrations of vitamin E in humans. J Lipid Res. 1993;34:343–358.
    1. Hughes PE, Tove SB. Occurrence of alpha-tocopherolquinone and alpha-tocopherolquinol in microorganisms. J Bacteriol. 1982;151:1397–1402.
    1. Cani PD, Delzenne NM. The gut microbiome as therapeutic target. Pharmacol Ther. 2011;130:202–212.
    1. Sternberg DE, Heninger GR, Roth RH. Plasma homovanillic acid as an index of brain dopamine metabolism: enhancement with debrisoquin. Life Sci. 1983;32:2447–2452.
    1. Amin F, Davidson M, Davis KL. Homovanillic acid measurement in clinical research: a review of methodology. Schizophr Bull. 1992;18:123–148.
    1. Russo S, Kema IP, Bosker F, et al. Tryptophan as an evolutionarily conserved signal to brain serotonin: molecular evidence and psychiatric implications. World J Biol Psychiatry. 2009;10:258–268.
    1. Russo S, Kema IP, Haagsma EB, et al. Irritability rather than depression during interferon treatment is linked to increased tryptophan catabolism. Psychosom Med. 2005;67:773–777.
    1. Kurlan R, Goldblatt D, Zaczek R, et al. Cerebrospinal fluid homovanillic acid and parkinsonism in Huntington’s disease. Ann Neurol. 1988;24:282–284.
    1. Ziegler MG, Kennedy B, Holland OB, et al. The effects of dopamine agonists on human cardiovascular and sympathetic nervous systems. Int J Clin Pharmacol Ther Toxicol. 1985;23:175–179.
    1. Norman TR, Chiu E, French MA. Platelet monoamine oxidase activity in patients with Huntington’s disease. Clin Exp Pharmacol Physiol. 1987;14:547–550.
    1. Ascherio A, LeWitt PA, Xu K, et al. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch Neurol. 2009;66:1460–1468.
    1. Schwarzschild MA, Schwid SR, Marek K, et al. Serum urate as a predictor of clinical and radiographic progression in Parkinson disease. Arch Neurol. 2008;65:716–723.
    1. Parkinson Study Group S.-P. D. I. Schwarzschild MA, Ascherio A, et al. Inosine to increase serum and cerebrospinal fluid urate in Parkinson disease: a randomized clinical trial. JAMA Neurol. 2014;71:141–150.
    1. Chen X, Wu G, Schwarzschild MA. Urate in Parkinson’s disease: more than a biomarker? Curr Neurol Neurosci Rep. 2012;12:367–375. doi: ; published online Epub Aug.
    1. Thomas B, Matson S, Chopra V, et al. A novel method for detecting 7-methyl guanine reveals aberrant methylation levels in Huntington disease. Anal Biochem. 2013;436:112–120.
    1. Fox JH, Connor T, Stiles M, et al. Cysteine oxidation within N-terminal mutant huntingtin promotes oligomerization and delays clearance of soluble protein. J Biol Chem. 2011;286:18320–18330.
    1. Matson W U. S. Patents. 2001. Method of diagnosing or categorizing disorders from biochemical profiles.

Source: PubMed

3
Prenumerera