Identification of urinary metabolites that correlate with clinical improvements in children with autism treated with sulforaphane from broccoli

Stephen Bent, Brittany Lawton, Tracy Warren, Felicia Widjaja, Katherine Dang, Jed W Fahey, Brian Cornblatt, Jason M Kinchen, Kevin Delucchi, Robert L Hendren, Stephen Bent, Brittany Lawton, Tracy Warren, Felicia Widjaja, Katherine Dang, Jed W Fahey, Brian Cornblatt, Jason M Kinchen, Kevin Delucchi, Robert L Hendren

Abstract

Background: Children with autism spectrum disorder (ASD) have urinary metabolites suggesting impairments in several pathways, including oxidative stress, inflammation, mitochondrial dysfunction, and gut microbiome alterations. Sulforaphane, a supplement with indirect antioxidant effects that are derived from broccoli sprouts and seeds, was recently shown to lead to improvements in behavior and social responsiveness in children with ASD. We conducted the current open-label study to determine if we could identify changes in urinary metabolites that were associated with clinical improvements with the goal of identifying a potential mechanism of action.

Methods: Children and young adults enrolled in a school for children with ASD and related neurodevelopmental disorders were recruited to participate in a 12-week, open-label study of sulforaphane. Fasting urinary metabolites and measures of behavior (Aberrant Behavior Checklist-ABC) and social responsiveness (Social Responsiveness Scale-SRS) were measured at baseline and at the end of the study. Pearson's correlation coefficient was calculated for the pre- to post-intervention change in each of the two clinical scales (ABS and SRS) versus the change in each metabolite.

Results: Fifteen children completed the 12-week study. Mean scores on both symptom measures showed improvements (decreases) over the study period, but only the change in the SRS was significant. The ABC improved - 7.1 points (95% CI - 17.4 to 3.2), and the SRS improved - 9.7 points (95% CI - 18.7 to - 0.8). We identified 77 urinary metabolites that were correlated with changes in symptoms, and they clustered into pathways of oxidative stress, amino acid/gut microbiome, neurotransmitters, hormones, and sphingomyelin metabolism.

Conclusions: Urinary metabolomics analysis is a useful tool to identify pathways that may be involved in the mechanism of action of treatments targeting abnormal physiology in ASD.

Trial registration: This study was prospectively registered at clinicaltrials.gov (NCT02654743) on January 11, 2016.

Keywords: Antioxidant; Autism; Biomarker; Metabolomics.

Conflict of interest statement

The study was approved by the Committee on Human Research at the University of California, San Francisco (UCSF) on November 5, 2015. Informed consent was obtained from the parent/caregiver of all study participants.RLH reports research grants from Curemark, BioMarin, Roche, Shire, Sunovion, Vitamin D Council and Advisory Boards for Curemark, BioMarin, Neuren, and Janssen. None of these grants or companies are directly involved in the subject of the current study. JWF was a co-author on the first randomized controlled trial examining the efficacy of sulforaphane and manufactured the product used in that study, which found a beneficial effect. BC is the Medical Director and Director of Consumer Product Support for Nutramax Laboratories Consumer Care, Inc., which supplied the sulforaphane supplement and matching placebo for this study at no cost. JMK is a senior scientist for Metabolon, which performed the metabolomic analyses for this study under a paid contract. All other authors declare that they have no conflict of interest.Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Change in mean outcome scores over time. a Change in mean aberrant behavior (ABC). b Change in mean social responsiveness (SRS). Mean scores were adjusted for sex and age of subjects. Decreasing score indicates clinical improvement

References

    1. Rossignol DA, Frye RE. A review of research trends in physiological abnormalities in autism spectrum disorders: immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Mol Psychiatry. 2012;17(4):389–401. doi: 10.1038/mp.2011.165.
    1. Ashwood P, Wills S, Van de Water J. The immune response in autism: a new frontier for autism research. J Leukoc Biol. 2006;80(1):1–15. doi: 10.1189/jlb.1205707.
    1. James SJ, Cutler P, Melnyk S, Jernigan S, Janak L, Gaylor DW, Neubrander JA. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr. 2004;80(6):1611–1617. doi: 10.1093/ajcn/80.6.1611.
    1. Rossignol DA, Bradstreet JJ. Evidence of mitochondrial dysfunction in autism and implications for treatment. Am J Biochem Biotechnol. 2008;4(2):208–217. doi: 10.3844/ajbbsp.2008.208.217.
    1. Liu H, Talalay P, Fahey JW. Biomarker-guided strategy for treatment of autism spectrum disorder (ASD) CNS Neurol Disord Drug Targets. 2016;15(5):602–613. doi: 10.2174/1871527315666160413120414.
    1. National Research Council. Educating Children with Autism. Committee on Educational Interventions for Children with Autism. Catherine Lord and James P. McGee, eds. Division of Behavioral and Social Sciences and Education. Washington, DC: National Academy Press; 2001.
    1. Dieme B, Mavel S, Blasco H, Tripi G, Bonnet-Brilhault F, Malvy J, Bocca C, Andres CR, Nadal-Desbarats L, Emond P. Metabolomics study of urine in autism spectrum disorders using a multiplatform analytical methodology. J Proteome Res. 2015;14(12):5273–5282. doi: 10.1021/acs.jproteome.5b00699.
    1. Emond P, Mavel S, Aidoud N, Nadal-Desbarats L, Montigny F, Bonnet-Brilhault F, Barthelemy C, Merten M, Sarda P, Laumonnier F, et al. GC-MS-based urine metabolic profiling of autism spectrum disorders. Anal Bioanal Chem. 2013;405(15):5291–5300. doi: 10.1007/s00216-013-6934-x.
    1. Gevi F, Zolla L, Gabriele S, Persico AM. Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism. Mol Autism. 2016;7:47. doi: 10.1186/s13229-016-0109-5.
    1. Kuwabara H, Yamasue H, Koike S, Inoue H, Kawakubo Y, Kuroda M, Takano Y, Iwashiro N, Natsubori T, Aoki Y, et al. Altered metabolites in the plasma of autism spectrum disorder: a capillary electrophoresis time-of-flight mass spectroscopy study. PLoS One. 2013;8(9):e73814. doi: 10.1371/journal.pone.0073814.
    1. Mavel S, Nadal-Desbarats L, Blasco H, Bonnet-Brilhault F, Barthelemy C, Montigny F, Sarda P, Laumonnier F, Vourc’h P, Andres CR, et al. 1H-13C NMR-based urine metabolic profiling in autism spectrum disorders. Talanta. 2013;114:95–102. doi: 10.1016/j.talanta.2013.03.064.
    1. Ming X, Stein TP, Barnes V, Rhodes N, Guo L. Metabolic perturbance in autism spectrum disorders: a metabolomics study. J Proteome Res. 2012;11(12):5856–5862. doi: 10.1021/pr300910n.
    1. Nadal-Desbarats L, Aidoud N, Emond P, Blasco H, Filipiak I, Sarda P, Bonnet-Brilhault F, Mavel S, Andres CR. Combined 1H-NMR and 1H-13C HSQC-NMR to improve urinary screening in autism spectrum disorders. Analyst. 2014;139(13):3460–3468. doi: 10.1039/C4AN00552J.
    1. Noto A, Fanos V, Barberini L, Grapov D, Fattuoni C, Zaffanello M, Casanova A, Fenu G, De Giacomo A, De Angelis M, et al. The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J Matern Fetal Neonatal Med. 2014;27(Suppl 2):46–52. doi: 10.3109/14767058.2014.954784.
    1. West PR, Amaral DG, Bais P, Smith AM, Egnash LA, Ross ME, Palmer JA, Fontaine BR, Conard KR, Corbett BA, et al. Metabolomics as a tool for discovery of biomarkers of autism spectrum disorder in the blood plasma of children. PLoS One. 2014;9(11):e112445. doi: 10.1371/journal.pone.0112445.
    1. Yap IK, Angley M, Veselkov KA, Holmes E, Lindon JC, Nicholson JK. Urinary metabolic phenotyping differentiates children with autism from their unaffected siblings and age-matched controls. J Proteome Res. 2010;9(6):2996–3004. doi: 10.1021/pr901188e.
    1. Hardan AY, Fung LK, Libove RA, Obukhanych TV, Nair S, Herzenberg LA, Frazier TW, Tirouvanziam R. A randomized controlled pilot trial of oral N-acetylcysteine in children with autism. Biol Psychiatry. 2012;71(11):956–961. doi: 10.1016/j.biopsych.2012.01.014.
    1. Hendren RL, James SJ, Widjaja F, Lawton B, Rosenblatt A, Bent S. Randomized, placebo-controlled trial of methyl B12 for children with autism. J Child Adolesc Psychopharmacol. 2016;26(9):774–783. doi: 10.1089/cap.2015.0159.
    1. Singh K, Connors SL, Macklin EA, Smith KD, Fahey JW, Talalay P, Zimmerman AW. Sulforaphane treatment of autism spectrum disorder (ASD) Proc Natl Acad Sci U S A. 2014;111(43):15550–15555. doi: 10.1073/pnas.1416940111.
    1. Fahey JW, Talalay P. Antioxidant functions of sulforaphane: a potent inducer of phase II detoxication enzymes. Food Chem Toxicol. 1999;37(9–10):973–979. doi: 10.1016/S0278-6915(99)00082-4.
    1. Fahey JW, Holtzclaw WD, Wehage SL, Wade KL, Stephenson KK, Talalay P. Sulforaphane bioavailability from glucoraphanin-rich broccoli: control by active endogenous myrosinase. PLoS One. 2015;10(11):e0140963. doi: 10.1371/journal.pone.0140963.
    1. Long T, Hicks M, Yu HC, Biggs WH, Kirkness EF, Menni C, Zierer J, Small KS, Mangino M, Messier H, et al. Whole-genome sequencing identifies common-to-rare variants associated with human blood metabolites. Nat Genet. 2017;49(4):568–578. doi: 10.1038/ng.3809.
    1. Dehaven CD, Evans AM, Dai H, Lawton KA. Organization of GC/MS and LC/MS metabolomics data into chemical libraries. J Cheminform. 2010;2(1):9. doi: 10.1186/1758-2946-2-9.
    1. Chetwynd AJ, Abdul-Sada A, Holt SG, Hill EM. Use of a pre-analysis osmolality normalisation method to correct for variable urine concentrations and for improved metabolomic analyses. J Chromatogr A. 2016;1431:103–110. doi: 10.1016/j.chroma.2015.12.056.
    1. Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology. 1990;1(1):43–46. doi: 10.1097/00001648-199001000-00010.
    1. Mukaka MM. Statistics corner: a guide to appropriate use of correlation coefficient in medical research. Malawi Med J. 2012;24:69–71.
    1. McCracken JT, McGough J, Shah B, Cronin P, Hong D, Aman MG, Arnold LE, Lindsay R, Nash P, Hollway J, et al. Risperidone in children with autism and serious behavioral problems. N Engl J Med. 2002;347(5):314–321. doi: 10.1056/NEJMoa013171.
    1. Juge N, Mithen RF, Traka M. Molecular basis for chemoprevention by sulforaphane: a comprehensive review. Cell Mol Life Sci. 2007;64(9):1105–1127. doi: 10.1007/s00018-007-6484-5.
    1. Melnyk S, Fuchs GJ, Schulz E, Lopez M, Kahler SG, Fussell JJ, Bellando J, Pavliv O, Rose S, Seidel L, et al. Metabolic imbalance associated with methylation dysregulation and oxidative damage in children with autism. J Autism Dev Disord. 2012;42(3):367–377. doi: 10.1007/s10803-011-1260-7.
    1. Lussu M, Noto A, Masili A, Rinaldi AC, Dessi A, De Angelis M, De Giacomo A, Fanos V, Atzori L, Francavilla R. The urinary 1 H-NMR metabolomics profile of an Italian autistic children population and their unaffected siblings. Autism Res. 2017;10(6):1058–66.
    1. Ogawa S, Lee YA, Yamaguchi Y, Shibata Y, Goto Y. Associations of acute and chronic stress hormones with cognitive functions in autism spectrum disorder. Neuroscience. 2017;343:229–239. doi: 10.1016/j.neuroscience.2016.12.003.
    1. Putnam SK, Lopata C, Thomeer ML, Voker MA, Rodgers JD. Salivary cortisol levels and diurnal patterns in children with autism spectrum disorder. J Dev Physic Disabil. 2015;27:453–465. doi: 10.1007/s10882-015-9428-2.
    1. Martinez-Cerdeno V. Dendrite and spine modifications in autism and related neurodevelopmental disorders in patients and animal models. Dev Neurobiol. 2017;77(4):393–404. doi: 10.1002/dneu.22417.
    1. Berkecz R, Tomosi F, Kormoczi T, Szegedi V, Horvath J, Janaky T. Comprehensive phospholipid and sphingomyelin profiling of different brain regions in mouse model of anxiety disorder using online two-dimensional (HILIC/RP)-LC/MS method. J Pharm Biomed Anal. 2017;149:308–317. doi: 10.1016/j.jpba.2017.10.043.
    1. Blasco H, Veyrat-Durebex C, Bocca C, Patin F, Vourc’h P, Kouassi Nzoughet J, Lenaers G, Andres CR, Simard G, Corcia P, et al. Lipidomics reveals cerebrospinal-fluid signatures of ALS. Sci Rep. 2017;7(1):17652. doi: 10.1038/s41598-017-17389-9.
    1. Dinoff A, Saleem M, Herrmann N, Mielke MM, Oh PI, Venkata SLV, Haughey NJ, Lanctot KL. Plasma sphingolipids and depressive symptoms in coronary artery disease. Brain and behavior. 2017;7(11):e00836. doi: 10.1002/brb3.836.
    1. Kosicek M, Zetterberg H, Andreasen N, Peter-Katalinic J, Hecimovic S. Elevated cerebrospinal fluid sphingomyelin levels in prodromal Alzheimer’s disease. Neurosci Lett. 2012;516(2):302–305. doi: 10.1016/j.neulet.2012.04.019.

Source: PubMed

3
S'abonner