Skip to main content
Top
Published in: Molecular Autism 1/2016

Open Access 01-12-2016 | Research

Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism

Authors: Federica Gevi, Lello Zolla, Stefano Gabriele, Antonio M. Persico

Published in: Molecular Autism | Issue 1/2016

Login to get access

Abstract

Background

Autism spectrum disorder (ASD) is still diagnosed through behavioral observation, due to a lack of laboratory biomarkers, which could greatly aid clinicians in providing earlier and more reliable diagnoses. Metabolomics on human biofluids provides a sensitive tool to identify metabolite profiles potentially usable as biomarkers for ASD. Initial metabolomic studies, analyzing urines and plasma of ASD and control individuals, suggested that autistic patients may share some metabolic abnormalities, despite several inconsistencies stemming from differences in technology, ethnicity, age range, and definition of “control” status.

Methods

ASD-specific urinary metabolomic patterns were explored at an early age in 30 ASD children and 30 matched controls (age range 2–7, M:F = 22:8) using hydrophilic interaction chromatography (HILIC)-UHPLC and mass spectrometry, a highly sensitive, accurate, and unbiased approach. Metabolites were then subjected to multivariate statistical analysis and grouped by metabolic pathway.

Results

Urinary metabolites displaying the largest differences between young ASD and control children belonged to the tryptophan and purine metabolic pathways. Also, vitamin B6, riboflavin, phenylalanine-tyrosine-tryptophan biosynthesis, pantothenate and CoA, and pyrimidine metabolism differed significantly. ASD children preferentially transform tryptophan into xanthurenic acid and quinolinic acid (two catabolites of the kynurenine pathway), at the expense of kynurenic acid and especially of melatonin. Also, the gut microbiome contributes to altered tryptophan metabolism, yielding increased levels of indolyl 3-acetic acid and indolyl lactate.

Conclusions

The metabolic pathways most distinctive of young Italian autistic children largely overlap with those found in rodent models of ASD following maternal immune activation or genetic manipulations. These results are consistent with the proposal of a purine-driven cell danger response, accompanied by overproduction of epileptogenic and excitotoxic quinolinic acid, large reductions in melatonin synthesis, and gut dysbiosis. These metabolic abnormalities could underlie several comorbidities frequently associated to ASD, such as seizures, sleep disorders, and gastrointestinal symptoms, and could contribute to autism severity. Their diagnostic sensitivity, disease-specificity, and interethnic variability will merit further investigation.
Appendix
Available only for authorised users
Literature
1.
go back to reference American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington, DC; 2013. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Washington, DC; 2013.
2.
3.
go back to reference Developmental Disabilities Monitoring Network Surveillance Year 2010 Principal Investigators, Centers for Disease Control and Prevention (CDC). Prevalence of autism spectrum disorder among children aged 8 years—autism and developmental disabilities monitoring network, 11 sites, United States, 2010. MMWR Surveill Summ. 2014;63:1–21. Developmental Disabilities Monitoring Network Surveillance Year 2010 Principal Investigators, Centers for Disease Control and Prevention (CDC). Prevalence of autism spectrum disorder among children aged 8 years—autism and developmental disabilities monitoring network, 11 sites, United States, 2010. MMWR Surveill Summ. 2014;63:1–21.
4.
go back to reference Rutter M. Incidence of autism spectrum disorders: changes over time and their meaning. Acta Paediatr. 2005;94:2–15.CrossRefPubMed Rutter M. Incidence of autism spectrum disorders: changes over time and their meaning. Acta Paediatr. 2005;94:2–15.CrossRefPubMed
5.
go back to reference Frans EM, Sandin S, Reichenberg A, Långström N, Lichtenstein P, McGrath JJ, Hultman CM. Autism risk across generations: a population-based study of advancing grandpaternal and paternal age. JAMA Psychiat. 2013;70:516–21.CrossRef Frans EM, Sandin S, Reichenberg A, Långström N, Lichtenstein P, McGrath JJ, Hultman CM. Autism risk across generations: a population-based study of advancing grandpaternal and paternal age. JAMA Psychiat. 2013;70:516–21.CrossRef
6.
go back to reference Persico AM, Merelli S. Environmental factors and autism spectrum disorder. In: Leboyer M, Chaste P, editors. Autism spectrum disorders: phenotypes, mechanisms and treatments. vol. 180. Basilea: Karger; 2015. p. 113–34. Persico AM, Merelli S. Environmental factors and autism spectrum disorder. In: Leboyer M, Chaste P, editors. Autism spectrum disorders: phenotypes, mechanisms and treatments. vol. 180. Basilea: Karger; 2015. p. 113–34.
9.
go back to reference Ruggeri B, Sarkans U, Schumann G, Persico AM. Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology (Berl). 2014;231:1201–16.CrossRef Ruggeri B, Sarkans U, Schumann G, Persico AM. Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology (Berl). 2014;231:1201–16.CrossRef
10.
go back to reference 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;9:2996–3004.CrossRefPubMed 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;9:2996–3004.CrossRefPubMed
11.
go back to reference Ming X, Stein TP, Barnes V, Rhodes N, Guo L. Metabolic perturbance in autism spectrum disorders: a metabolomics study. J Proteome Res. 2012;11:5856–62.PubMed Ming X, Stein TP, Barnes V, Rhodes N, Guo L. Metabolic perturbance in autism spectrum disorders: a metabolomics study. J Proteome Res. 2012;11:5856–62.PubMed
12.
go back to reference Emond P, Mavel S, Aidoud N, Nadal-Desbarats L, Montigny F, Bonnet-Brilhault F, Barthélémy C, Merten M, Sarda P, Laumonnier F, Vourc’h P, Blasco H, Andres CR. GC-MS-based urine metabolic profiling of autism spectrum disorders. Anal Bioanal Chem. 2013;15:5291–300.CrossRef Emond P, Mavel S, Aidoud N, Nadal-Desbarats L, Montigny F, Bonnet-Brilhault F, Barthélémy C, Merten M, Sarda P, Laumonnier F, Vourc’h P, Blasco H, Andres CR. GC-MS-based urine metabolic profiling of autism spectrum disorders. Anal Bioanal Chem. 2013;15:5291–300.CrossRef
13.
go back to reference Mavel S, Nadal-Desbarats L, Blasco H, Bonnet-Brilhault F, Barthélémy C, Montigny F, Sarda P, Laumonnier F, Vourc’h P, Andres CR, Emond P. 1H-13C NMR-based urine metabolic profiling in autism spectrum disorders. Talanta. 2013;114:95–102.CrossRefPubMed Mavel S, Nadal-Desbarats L, Blasco H, Bonnet-Brilhault F, Barthélémy C, Montigny F, Sarda P, Laumonnier F, Vourc’h P, Andres CR, Emond P. 1H-13C NMR-based urine metabolic profiling in autism spectrum disorders. Talanta. 2013;114:95–102.CrossRefPubMed
14.
go back to reference Nadal-Desbarats L, Aïdoud 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;139:3460–8.CrossRefPubMed Nadal-Desbarats L, Aïdoud 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;139:3460–8.CrossRefPubMed
15.
go back to reference Noto A, Fanos V, Barberini L, Grapov D, Fattuoni C, Zaffanello M, Casanova A, Fenu G, De Giacomo A, De Angelis M, Moretti C, Papoff P, Ditonno R, Francavilla R. The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J Matern Fetal Neonatal Med. 2014;2:46–52.CrossRef Noto A, Fanos V, Barberini L, Grapov D, Fattuoni C, Zaffanello M, Casanova A, Fenu G, De Giacomo A, De Angelis M, Moretti C, Papoff P, Ditonno R, Francavilla R. The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J Matern Fetal Neonatal Med. 2014;2:46–52.CrossRef
16.
go back to reference Diémé 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;14:5273–82.CrossRefPubMed Diémé 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;14:5273–82.CrossRefPubMed
17.
go back to reference West PR, Amaral DG, Bais P, Smith AM, Egnash LA, Ross ME, Palmer JA, Fontaine BR, Conard KR, Corbett BA, Cezar GG, Donley EL, Burrier RE. Metabolomics as a tool for discovery of biomarkers of autism spectrum disorder in the blood plasma of children. PLoS One. 2014;9:e112445.CrossRefPubMedPubMedCentral West PR, Amaral DG, Bais P, Smith AM, Egnash LA, Ross ME, Palmer JA, Fontaine BR, Conard KR, Corbett BA, Cezar GG, Donley EL, Burrier RE. Metabolomics as a tool for discovery of biomarkers of autism spectrum disorder in the blood plasma of children. PLoS One. 2014;9:e112445.CrossRefPubMedPubMedCentral
18.
go back to reference Nicholson G, Rantalainen M, Li JV, Maher AD, Malmodin D, Ahmadi KR, Faber JH, Barrett A, Min JL, Rayner NW, Toft H, Krestyaninova M, Viksna J, Neogi SG, Dumas ME, Sarkans U, MolPAGE Consortium, Donnelly P, Illig T, Adamski J, Suhre K, Allen M, Zondervan KT, Spector TD, Nicholson JK, Lindon JC, Baunsgaard D, Holmes E, McCarthy MI, Holmes CC. A genome-wide metabolic QTL analysis in Europeans identifies functional effects of two loci shaped by recent positive selection. PLoS Genet. 2011;7:1002270.CrossRef Nicholson G, Rantalainen M, Li JV, Maher AD, Malmodin D, Ahmadi KR, Faber JH, Barrett A, Min JL, Rayner NW, Toft H, Krestyaninova M, Viksna J, Neogi SG, Dumas ME, Sarkans U, MolPAGE Consortium, Donnelly P, Illig T, Adamski J, Suhre K, Allen M, Zondervan KT, Spector TD, Nicholson JK, Lindon JC, Baunsgaard D, Holmes E, McCarthy MI, Holmes CC. A genome-wide metabolic QTL analysis in Europeans identifies functional effects of two loci shaped by recent positive selection. PLoS Genet. 2011;7:1002270.CrossRef
19.
go back to reference Gebregiworgis T, Powers R. Application of NMR metabolomics to search for human disease biomarkers. Comb Chem High Throughput Screen. 2012;15:595–610.CrossRefPubMed Gebregiworgis T, Powers R. Application of NMR metabolomics to search for human disease biomarkers. Comb Chem High Throughput Screen. 2012;15:595–610.CrossRefPubMed
20.
go back to reference Altieri L, Neri C, Sacco R, Curatolo P, Benvenuto A, Muratori F, Santocchi E, Bravaccio C, Lenti C, Saccani M, Rigardetto R, Gandione M, Urbani A, Persico AM. Urinary p-cresol is elevated in small children with autism spectrum disorder. Biomarkers. 2011;16:252–60.CrossRefPubMed Altieri L, Neri C, Sacco R, Curatolo P, Benvenuto A, Muratori F, Santocchi E, Bravaccio C, Lenti C, Saccani M, Rigardetto R, Gandione M, Urbani A, Persico AM. Urinary p-cresol is elevated in small children with autism spectrum disorder. Biomarkers. 2011;16:252–60.CrossRefPubMed
21.
go back to reference Gabriele S, Sacco R, Cerullo S, Neri C, Urbani A, Tripi G, Malvy J, Barthelemy C, Bonnet-Brihault F, Persico AM. Urinary p-cresol is elevated in young French children with autism spectrum disorder: a replication study. Biomarkers. 2014;19:463–70.CrossRefPubMed Gabriele S, Sacco R, Cerullo S, Neri C, Urbani A, Tripi G, Malvy J, Barthelemy C, Bonnet-Brihault F, Persico AM. Urinary p-cresol is elevated in young French children with autism spectrum disorder: a replication study. Biomarkers. 2014;19:463–70.CrossRefPubMed
22.
go back to reference Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, Chugani HT. Developmental changes in brain serotonin synthesis capacity in autistic and nonautistic children. Ann Neurol. 1999;45:287–95.CrossRefPubMed Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, Chugani HT. Developmental changes in brain serotonin synthesis capacity in autistic and nonautistic children. Ann Neurol. 1999;45:287–95.CrossRefPubMed
23.
go back to reference Chandana SR, Behen ME, Juhász C, Muzik O, Rothermel RD, Mangner TJ, Chakraborty PK, Chugani HT, Chugani DC. Significance of abnormalities in developmental trajectory and asymmetry of cortical serotonin synthesis in autism. Int J Dev Neurosci. 2005;23:171–82.CrossRefPubMed Chandana SR, Behen ME, Juhász C, Muzik O, Rothermel RD, Mangner TJ, Chakraborty PK, Chugani HT, Chugani DC. Significance of abnormalities in developmental trajectory and asymmetry of cortical serotonin synthesis in autism. Int J Dev Neurosci. 2005;23:171–82.CrossRefPubMed
24.
go back to reference Courchesne E, Pierce K, Schumann CM, Redcay E, Buckwalter JA, Kennedy DP, Morgan J. Mapping early brain development in autism. Neuron. 2007;56:399–413.CrossRefPubMed Courchesne E, Pierce K, Schumann CM, Redcay E, Buckwalter JA, Kennedy DP, Morgan J. Mapping early brain development in autism. Neuron. 2007;56:399–413.CrossRefPubMed
25.
go back to reference Piven J, Palmer P, Jacobi D, Childress D, Arndt S. Broader autism phenotype: evidence from a family history study of multiple-incidence autism families. Am J Psychiatry. 1997;154:185–90.CrossRefPubMed Piven J, Palmer P, Jacobi D, Childress D, Arndt S. Broader autism phenotype: evidence from a family history study of multiple-incidence autism families. Am J Psychiatry. 1997;154:185–90.CrossRefPubMed
26.
27.
go back to reference Alpert AJ. Electrostatic repulsion hydrophilic interaction chromatography for isocratic separation of charged solutes and selective isolation of phosphopeptides. Anal Chem. 2008;80:62–76.CrossRefPubMed Alpert AJ. Electrostatic repulsion hydrophilic interaction chromatography for isocratic separation of charged solutes and selective isolation of phosphopeptides. Anal Chem. 2008;80:62–76.CrossRefPubMed
28.
go back to reference D’Alessandro A, Gevi F, Zolla L. A robust high resolution reversed-phase HPLC strategy to investigate various metabolic species in different biological models. Mol Biosyst. 2011;7:1024–32.CrossRefPubMed D’Alessandro A, Gevi F, Zolla L. A robust high resolution reversed-phase HPLC strategy to investigate various metabolic species in different biological models. Mol Biosyst. 2011;7:1024–32.CrossRefPubMed
29.
go back to reference Gevi F, D’Alessandro A, Rinalducci S, Zolla L. Alterations of red blood cell metabolome during cold liquid storage of erythrocyte concentrates in CPD–SAGM. J Proteomics. 2012;76 Spec No:168–80.CrossRefPubMed Gevi F, D’Alessandro A, Rinalducci S, Zolla L. Alterations of red blood cell metabolome during cold liquid storage of erythrocyte concentrates in CPD–SAGM. J Proteomics. 2012;76 Spec No:168–80.CrossRefPubMed
30.
go back to reference Whiteley P, Waring R, Williams L, Klovrza L, Nolan F, Smith S, Farrow M, Dodou K, Lough WJ, Shattock P. Spot urinary creatinine excretion in pervasive developmental disorders. Pediatr Int. 2006;48:292–7.CrossRefPubMed Whiteley P, Waring R, Williams L, Klovrza L, Nolan F, Smith S, Farrow M, Dodou K, Lough WJ, Shattock P. Spot urinary creatinine excretion in pervasive developmental disorders. Pediatr Int. 2006;48:292–7.CrossRefPubMed
31.
go back to reference Melamud E, Vastag L, Rabinowitz JD. Metabolomic analysis and visualization engine for LC-MS data. Anal Chem. 2010;82:9818–26.CrossRefPubMed Melamud E, Vastag L, Rabinowitz JD. Metabolomic analysis and visualization engine for LC-MS data. Anal Chem. 2010;82:9818–26.CrossRefPubMed
32.
go back to reference Xia J, Mandal R, Sinelnikov I, Broadhurst D, Wishart DS. MetaboAnalyst 2.0—a comprehensive server for metabolomic data analysis. Nucleic Acids Res. 2012;40:W127–33.CrossRefPubMedPubMedCentral Xia J, Mandal R, Sinelnikov I, Broadhurst D, Wishart DS. MetaboAnalyst 2.0—a comprehensive server for metabolomic data analysis. Nucleic Acids Res. 2012;40:W127–33.CrossRefPubMedPubMedCentral
34.
go back to reference Ellis DI, Goodacre R. Metabolic fingerprinting in disease diagnosis: biomedical applications of infrared and Raman spectroscopy. Analyst. 2006;131:875–85.CrossRefPubMed Ellis DI, Goodacre R. Metabolic fingerprinting in disease diagnosis: biomedical applications of infrared and Raman spectroscopy. Analyst. 2006;131:875–85.CrossRefPubMed
35.
go back to reference Naviaux JC, Wang L, Li K, Bright AT, Alaynick WA, Williams KR, Powell SB, Naviaux RK. Antipurinergic therapy corrects the autism-like features in the fragile X (Fmr1 knockout) mouse model. Mol Autism. 2015;6:1.CrossRefPubMedPubMedCentral Naviaux JC, Wang L, Li K, Bright AT, Alaynick WA, Williams KR, Powell SB, Naviaux RK. Antipurinergic therapy corrects the autism-like features in the fragile X (Fmr1 knockout) mouse model. Mol Autism. 2015;6:1.CrossRefPubMedPubMedCentral
36.
go back to reference Naviaux JC, Schuchbauer MA, Li K, Wang L, Risbrough VB, Powell SB, Naviaux RK. Reversal of autism-like behaviors and metabolism in adult mice with single-dose antipurinergic therapy. Transl Psychiatry. 2014;4:e400.CrossRefPubMedPubMedCentral Naviaux JC, Schuchbauer MA, Li K, Wang L, Risbrough VB, Powell SB, Naviaux RK. Reversal of autism-like behaviors and metabolism in adult mice with single-dose antipurinergic therapy. Transl Psychiatry. 2014;4:e400.CrossRefPubMedPubMedCentral
37.
go back to reference Naviaux RK, Zolkipli Z, Wang L, Nakayama T, Naviaux JC, Le TP, Schuchbauer MA, Rogac M, Tang Q, Dugan LL, Powell SB. Antipurinergic therapy corrects the autism-like features in the poly(IC) mouse model. PLoS One. 2013;8:e57380.CrossRefPubMedPubMedCentral Naviaux RK, Zolkipli Z, Wang L, Nakayama T, Naviaux JC, Le TP, Schuchbauer MA, Rogac M, Tang Q, Dugan LL, Powell SB. Antipurinergic therapy corrects the autism-like features in the poly(IC) mouse model. PLoS One. 2013;8:e57380.CrossRefPubMedPubMedCentral
38.
go back to reference Böhmer C, Bröer A, Munzinger M, Kowalczuk S, Rasko JE, Lang F, Bröer S. Characterization of mouse amino acid transporter B0AT1 (slc6a19). Biochem J. 2005;389(Pt 3):745–51.CrossRefPubMedPubMedCentral Böhmer C, Bröer A, Munzinger M, Kowalczuk S, Rasko JE, Lang F, Bröer S. Characterization of mouse amino acid transporter B0AT1 (slc6a19). Biochem J. 2005;389(Pt 3):745–51.CrossRefPubMedPubMedCentral
39.
go back to reference Singer D, Camargo SM, Ramadan T, Schäfer M, Mariotta L, Herzog B, Huggel K, Wolfer D, Werner S, Penninger JM, Verrey F. Defective intestinal amino acid absorption in Ace2 null mice. Am J Physiol Gastrointest Liver Physiol. 2012;303:G686–95.CrossRefPubMed Singer D, Camargo SM, Ramadan T, Schäfer M, Mariotta L, Herzog B, Huggel K, Wolfer D, Werner S, Penninger JM, Verrey F. Defective intestinal amino acid absorption in Ace2 null mice. Am J Physiol Gastrointest Liver Physiol. 2012;303:G686–95.CrossRefPubMed
40.
go back to reference Keszthelyi D, Troost FJ, Masclee AA. Understanding the role of tryptophan and serotonin metabolism in gastrointestinal function. Neurogastroenterol Motil. 2009;21:1239–49.CrossRefPubMed Keszthelyi D, Troost FJ, Masclee AA. Understanding the role of tryptophan and serotonin metabolism in gastrointestinal function. Neurogastroenterol Motil. 2009;21:1239–49.CrossRefPubMed
41.
go back to reference Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol. 2015;40:134–41.CrossRefPubMed Maddison DC, Giorgini F. The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol. 2015;40:134–41.CrossRefPubMed
42.
go back to reference Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2004;57:67–8.CrossRef Vargas DL, Nascimbene C, Krishnan C, Zimmerman AW, Pardo CA. Neuroglial activation and neuroinflammation in the brain of patients with autism. Ann Neurol. 2004;57:67–8.CrossRef
43.
go back to reference Suzuki K, Sugihara G, Ouchi Y, Nakamura K, Futatsubashi M, Takebayashi K, Yoshihara Y, Omata K, Matsumoto K, Tsuchiya KJ, Iwata Y, Tsujii M, Sugiyama T, Mori N. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiat. 2013;70:49–58.CrossRef Suzuki K, Sugihara G, Ouchi Y, Nakamura K, Futatsubashi M, Takebayashi K, Yoshihara Y, Omata K, Matsumoto K, Tsuchiya KJ, Iwata Y, Tsujii M, Sugiyama T, Mori N. Microglial activation in young adults with autism spectrum disorder. JAMA Psychiat. 2013;70:49–58.CrossRef
44.
go back to reference Gupta S, Ellis SE, Ashar FN, Moes A, Bader JS, Zhan J, West AB, Arking DE. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat Commun. 2014;5:5748.CrossRefPubMedPubMedCentral Gupta S, Ellis SE, Ashar FN, Moes A, Bader JS, Zhan J, West AB, Arking DE. Transcriptome analysis reveals dysregulation of innate immune response genes and neuronal activity-dependent genes in autism. Nat Commun. 2014;5:5748.CrossRefPubMedPubMedCentral
45.
go back to reference Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, Courchesne E, Everall IP. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010;68:368–76.CrossRefPubMed Morgan JT, Chana G, Pardo CA, Achim C, Semendeferi K, Buckwalter J, Courchesne E, Everall IP. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry. 2010;68:368–76.CrossRefPubMed
46.
go back to reference Guillemin G, Smith DG, Smythe GA, Armati PJ, Brew GJ. Expression of the kynurenine pathway enzymes in human microglia and macrophages. Adv Exp Med Biol. 2003;527:105–12.CrossRefPubMed Guillemin G, Smith DG, Smythe GA, Armati PJ, Brew GJ. Expression of the kynurenine pathway enzymes in human microglia and macrophages. Adv Exp Med Biol. 2003;527:105–12.CrossRefPubMed
48.
go back to reference Kohane IS, McMurry A, Weber G, MacFadden D, Rappaport L, Kunkel L, Bickel J, Wattanasin N, Spence S, Murphy S, Churchill S. The co-morbidity burden of children and young adults with autism spectrum disorders. PLoS One. 2012;7:e33224.CrossRefPubMedPubMedCentral Kohane IS, McMurry A, Weber G, MacFadden D, Rappaport L, Kunkel L, Bickel J, Wattanasin N, Spence S, Murphy S, Churchill S. The co-morbidity burden of children and young adults with autism spectrum disorders. PLoS One. 2012;7:e33224.CrossRefPubMedPubMedCentral
49.
go back to reference Nir I, Meir D, Zilber N, Knobler H, Hadjez J, Lerner Y. Brief report: circadian melatonin, thyroid-stimulating hormone, prolactin, and cortisol levels in serum of young adults with autism. J Autism Dev Disord. 1995;25:641–54.CrossRefPubMed Nir I, Meir D, Zilber N, Knobler H, Hadjez J, Lerner Y. Brief report: circadian melatonin, thyroid-stimulating hormone, prolactin, and cortisol levels in serum of young adults with autism. J Autism Dev Disord. 1995;25:641–54.CrossRefPubMed
50.
go back to reference Kulman G, Lissoni P, Rovelli F, Roselli MG, Brivio F, Sequeri P. Evidence of pineal endocrine hypofunction in autistic children. Neuroendocrinol Lett. 2000;21:31–4.PubMed Kulman G, Lissoni P, Rovelli F, Roselli MG, Brivio F, Sequeri P. Evidence of pineal endocrine hypofunction in autistic children. Neuroendocrinol Lett. 2000;21:31–4.PubMed
51.
go back to reference Tordjman S, Anderson GM, Pichard N, Charbuy H, Touitou Y. Nocturnal excretion of 6-sulphatoxymelatonin in children and adolescents with autistic disorder. Biol Psychiatry. 2005;57:134–8.CrossRefPubMed Tordjman S, Anderson GM, Pichard N, Charbuy H, Touitou Y. Nocturnal excretion of 6-sulphatoxymelatonin in children and adolescents with autistic disorder. Biol Psychiatry. 2005;57:134–8.CrossRefPubMed
52.
go back to reference Tordjman S, Anderson GM, Bellissant E, Botbol M, Charbuy H, Camus F, Graignic R, Kermarrec S, Fougerou C, Cohen D, Touitou Y. Day and nighttime excretion of 6-sulphatoxymelatonin in adolescents and young adults with autistic disorder. Psychoneuroendocrinology. 2012;37:1990–7.CrossRefPubMed Tordjman S, Anderson GM, Bellissant E, Botbol M, Charbuy H, Camus F, Graignic R, Kermarrec S, Fougerou C, Cohen D, Touitou Y. Day and nighttime excretion of 6-sulphatoxymelatonin in adolescents and young adults with autistic disorder. Psychoneuroendocrinology. 2012;37:1990–7.CrossRefPubMed
53.
go back to reference Melke J, Goubran Botros H, Chaste P, Betancur C, Nygren G, Anckarsäter H, Rastam M, Ståhlberg O, Gillberg IC, Delorme R, Chabane N, Mouren-Simeoni MC, Fauchereau F, Durand CM, Chevalier F, Drouot X, Collet C, Launay JM, Leboyer M, Gillberg C, Bourgeron T. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13:90–8.CrossRefPubMed Melke J, Goubran Botros H, Chaste P, Betancur C, Nygren G, Anckarsäter H, Rastam M, Ståhlberg O, Gillberg IC, Delorme R, Chabane N, Mouren-Simeoni MC, Fauchereau F, Durand CM, Chevalier F, Drouot X, Collet C, Launay JM, Leboyer M, Gillberg C, Bourgeron T. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008;13:90–8.CrossRefPubMed
54.
go back to reference Pagan C, Delorme R, Callebert J, Goubran-Botros H, Amsellem F, Drouot X, Boudebesse C, Le Dudal K, Ngo-Nguyen N, Laouamri H, Gillberg C, Leboyer M, Bourgeron T, Launay JM. The serotonin-N-acetylserotonin–melatonin pathway as a biomarker for autism spectrum disorders. Transl Psychiatry. 2014;4:e479.CrossRefPubMedPubMedCentral Pagan C, Delorme R, Callebert J, Goubran-Botros H, Amsellem F, Drouot X, Boudebesse C, Le Dudal K, Ngo-Nguyen N, Laouamri H, Gillberg C, Leboyer M, Bourgeron T, Launay JM. The serotonin-N-acetylserotonin–melatonin pathway as a biomarker for autism spectrum disorders. Transl Psychiatry. 2014;4:e479.CrossRefPubMedPubMedCentral
55.
go back to reference Vitte PA, Harthe C, Lestage P, Claustrat B, Bobillier P. Plasma, cerebrospinal fluid, and brain distribution of 14C-melatonin in rat: a biochemical and autoradiographic study. J Pineal Res. 1988;5:437–53.CrossRefPubMed Vitte PA, Harthe C, Lestage P, Claustrat B, Bobillier P. Plasma, cerebrospinal fluid, and brain distribution of 14C-melatonin in rat: a biochemical and autoradiographic study. J Pineal Res. 1988;5:437–53.CrossRefPubMed
56.
go back to reference Rossignol DA, Frye RE. Melatonin in autism spectrum disorders: a systematic review and meta-analysis. Dev Med Child Neurol. 2011;53:783–92.CrossRefPubMed Rossignol DA, Frye RE. Melatonin in autism spectrum disorders: a systematic review and meta-analysis. Dev Med Child Neurol. 2011;53:783–92.CrossRefPubMed
57.
go back to reference Bull G, Shattock P, Whiteley P, Anderson R, Groundwater P, Lough JW, Lees G. Indolyl-3- acryloylglycine (IAG) is a putative diagnostic urinary marker for autism spectrum disorders. Med Sci Monit. 2003;9:CR422–5.PubMed Bull G, Shattock P, Whiteley P, Anderson R, Groundwater P, Lough JW, Lees G. Indolyl-3- acryloylglycine (IAG) is a putative diagnostic urinary marker for autism spectrum disorders. Med Sci Monit. 2003;9:CR422–5.PubMed
58.
go back to reference Wang L, Angley MT, Gerber JP, Young RL, Abarno DV, McKinnon RA, Sorich MJ. Is urinary indolyl-3-acryloylglycine a biomarker for autism with gastrointestinal symptoms? Biomarkers. 2009;14:596–603.CrossRefPubMed Wang L, Angley MT, Gerber JP, Young RL, Abarno DV, McKinnon RA, Sorich MJ. Is urinary indolyl-3-acryloylglycine a biomarker for autism with gastrointestinal symptoms? Biomarkers. 2009;14:596–603.CrossRefPubMed
59.
go back to reference Wright B, Brzozowski AM, Calvert E, Farnworth H, Goodall DM, Holbrook I, Imrie G, Jordan J, Kelly A, Miles J, Smith R, Town J. Is the presence of urinary indolyl-3-acryloylglycine associated with autism spectrum disorder? Dev Med Child Neurol. 2005;47:190–2.CrossRefPubMed Wright B, Brzozowski AM, Calvert E, Farnworth H, Goodall DM, Holbrook I, Imrie G, Jordan J, Kelly A, Miles J, Smith R, Town J. Is the presence of urinary indolyl-3-acryloylglycine associated with autism spectrum disorder? Dev Med Child Neurol. 2005;47:190–2.CrossRefPubMed
60.
go back to reference Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Zhang M, Wei H, Chen Y, Lu H, Zuo J, Su M, Qiu Y, Jia W, Xiao C, Smith LM, Yang S, Holmes E, Tang H, Zhao G, Nicholson JK, Li L, Zhao L. Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A. 2008;105:2117–22.CrossRefPubMedPubMedCentral Li M, Wang B, Zhang M, Rantalainen M, Wang S, Zhou H, Zhang Y, Shen J, Pang X, Zhang M, Wei H, Chen Y, Lu H, Zuo J, Su M, Qiu Y, Jia W, Xiao C, Smith LM, Yang S, Holmes E, Tang H, Zhao G, Nicholson JK, Li L, Zhao L. Symbiotic gut microbes modulate human metabolic phenotypes. Proc Natl Acad Sci U S A. 2008;105:2117–22.CrossRefPubMedPubMedCentral
61.
go back to reference Thomas RH, Meeking MM, Mepham JR, Tichenoff L, Possmayer F, Liu S, MacFabe DF. The enteric bacterial metabolite propionic acid alters brain and plasma phospholipid molecular species: further development of a rodent model of autism spectrum disorders. J Neuroinflammation. 2012;9:153.CrossRefPubMedPubMedCentral Thomas RH, Meeking MM, Mepham JR, Tichenoff L, Possmayer F, Liu S, MacFabe DF. The enteric bacterial metabolite propionic acid alters brain and plasma phospholipid molecular species: further development of a rodent model of autism spectrum disorders. J Neuroinflammation. 2012;9:153.CrossRefPubMedPubMedCentral
62.
go back to reference Watanabe K, Watanabe T, Nakayama M. Cerebro-renal interactions: impact of uremic toxins on cognitive function. Neurotoxicology. 2014;44:184–93.CrossRefPubMed Watanabe K, Watanabe T, Nakayama M. Cerebro-renal interactions: impact of uremic toxins on cognitive function. Neurotoxicology. 2014;44:184–93.CrossRefPubMed
63.
go back to reference Ohtsuki S, Asaba H, Takanaga H, Deguchi T, Hosoya K, Otagiri M, Terasaki T. Role of blood-brain barrier organic anion transporter 3 (OAT3) in the efflux of indoxyl sulfate, a uremic toxin: its involvement in neurotransmitter metabolite clearance from the brain. J Neurochem. 2002;83:57–66.CrossRefPubMed Ohtsuki S, Asaba H, Takanaga H, Deguchi T, Hosoya K, Otagiri M, Terasaki T. Role of blood-brain barrier organic anion transporter 3 (OAT3) in the efflux of indoxyl sulfate, a uremic toxin: its involvement in neurotransmitter metabolite clearance from the brain. J Neurochem. 2002;83:57–66.CrossRefPubMed
64.
go back to reference Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Codelli JA, Chow J, Reisman SE, Petrosino JF, Patterson PH, Mazmanian SK. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–63.CrossRefPubMedPubMedCentral Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, Codelli JA, Chow J, Reisman SE, Petrosino JF, Patterson PH, Mazmanian SK. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–63.CrossRefPubMedPubMedCentral
65.
66.
go back to reference Nyhan WL, James JA, Teberg AJ, Sweetman L, Nelson LG. A new disorder of purine metabolism with behavioral manifestations. J Pediatr. 1969;74:20–7.CrossRefPubMed Nyhan WL, James JA, Teberg AJ, Sweetman L, Nelson LG. A new disorder of purine metabolism with behavioral manifestations. J Pediatr. 1969;74:20–7.CrossRefPubMed
67.
go back to reference Tada K, Yokoyama Y, Nakagawa H, Yoshida T, Arakawa T. Vitamin B6 dependent xanthurenic aciduria. Tohoku J Exp Med. 1967;93:115–24.CrossRefPubMed Tada K, Yokoyama Y, Nakagawa H, Yoshida T, Arakawa T. Vitamin B6 dependent xanthurenic aciduria. Tohoku J Exp Med. 1967;93:115–24.CrossRefPubMed
68.
go back to reference Martineau J, Barthelemy C, Garreau B, Lelord G. Vitamin B6, magnesium, and combined B6-Mg: therapeutic effects in childhood autism. Biol Psychiatry. 1985;20:467–78.CrossRefPubMed Martineau J, Barthelemy C, Garreau B, Lelord G. Vitamin B6, magnesium, and combined B6-Mg: therapeutic effects in childhood autism. Biol Psychiatry. 1985;20:467–78.CrossRefPubMed
69.
go back to reference Nye C, Brice A. Combined vitamin B6-magnesium treatment in autism spectrum disorder. Cochrane Database Syst Rev. 2005;4:CD003497. Nye C, Brice A. Combined vitamin B6-magnesium treatment in autism spectrum disorder. Cochrane Database Syst Rev. 2005;4:CD003497.
Metadata
Title
Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism
Authors
Federica Gevi
Lello Zolla
Stefano Gabriele
Antonio M. Persico
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Molecular Autism / Issue 1/2016
Electronic ISSN: 2040-2392
DOI
https://doi.org/10.1186/s13229-016-0109-5

Other articles of this Issue 1/2016

Molecular Autism 1/2016 Go to the issue