Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Oct 1.
Published in final edited form as: Metab Brain Dis. 2021 Aug 2;36(7):1641–1671. doi: 10.1007/s11011-021-00788-3

Profiles of urine and blood metabolomics in autism spectrum disorders

Narueporn Likhitweerawong 1, Chanisa Thonusin 2,3,4, Nonglak Boonchooduang 1, Orawan Louthrenoo 1, Intawat Nookaew 5, Nipon Chattipakorn 2,3,4, Siriporn C Chattipakorn 2,4,6
PMCID: PMC8502415  NIHMSID: NIHMS1742027  PMID: 34338974

Abstract

Early diagnosis and treatment for autism spectrum disorder (ASD) pose challenges. The current diagnostic approach for ASD is mainly clinical assessment of patient behaviors. Biomarkers-based identification of ASD would be useful for pediatricians. Currently, there is no specific treatment for ASD, and evidence for the efficacy of alternative treatments remains inconclusive. The prevalence of ASD is increasing, and it is becoming more urgent to find the pathogenesis of such disorder. Metabolomic studies have been used to deeply investigate the alteration of metabolic pathways, including those associated with ASD. Metabolomics is a promising tool for identifying potential biomarkers and possible pathogenesis of ASD. This review comprehensively summarizes and discusses the abnormal metabolic pathways in ASD children, as indicated by evidence from metabolomic studies in urine and blood. In addition, the targeted interventions that could correct the metabolomic profiles relating to the improvement of autistic behaviors in affected animals and humans have been included. The results revealed that the possible underlying pathophysiology of ASD were alterations of amino acids, reactive oxidative stress, neurotransmitters, and microbiota-gut-brain axis. The potential common pathways shared by animal and human studies related to the improvement of ASD symptoms after pharmacological interventions were mammalian-microbial co-metabolite, purine metabolism, and fatty acid oxidation. The content of this review may contribute to novel biomarkers for the early diagnosis of ASD and possible therapeutic paradigms.

Keywords: Metabolome, Metabolite, Autism, Diagnostic marker, Therapeutic marker, Intervention

Introduction

There are concerns about the increasing prevalence of ASD in recent decades (Hansen et al. 2015). The underlying pathogenesis of ASD remains unclear, and there is no definitive treatment for ASD (Emberti Gialloreti and Curatolo 2018). Several hypotheses have proposed that many factors might cause ASD, and the most well-known are genetic factors. There might also be interactions between genetic and environmental factors or epigenetics (Chaste and Leboyer 2012). In addition, some prenatal factors have been shown to increase the risk of ASD, including prenatal infection and exposure to specific drugs or toxins (Dietert et al. 2011). Interestingly, advanced technology-based genetic testing has revealed an abundance of candidate genes relating to ASD (da Silva Montenegro et al. 2020). Although ASD phenotypes can be seen in some single-gene disorders and metabolic diseases, only a few identifiable causes of ASD can be detected by genetic or metabolic testing. Therefore, the majority of cases are idiopathic ASD. With the lack of gold-standard laboratory tests for ASD, early diagnosis and initial intervention of such disorder are challenging (Huerta and Lord 2012).

Due to the complexity of ASD, the target approach for diagnosis is shifting to omics studies, including metabolomics. Due to its ability to measure metabolic profiles influenced by genetic and environmental factors, a metabolomic study may reveal the cause of abnormal metabolic pathways and identify possible biomarkers for diagnosis in various diseases, including ASD (Glinton and Elsea 2019).

An accumulation of evidence has indicated that the metabolomic profiles in biological samples were different between autistic and typically developing children (Anwar et al. 2018; Bitar et al. 2018; Chen et al. 2019; Cozzolino et al. 2014; Delaye et al. 2018; Diémé et al. 2015; Emond et al. 2013; Gevi et al. 2016; Kelly et al. 2019; Kuwabara et al. 2013; Liu et al. 2019; Lussu et al. 2017; Lv et al. 2018; Mavel et al. 2013; Ming et al. 2012; Nadal-Desbarats et al. 2014; Noto et al. 2014; Orozco et al. 2019; Rangel-Huerta et al. 2019; Smith et al. 2019; Wang et al. 2016; West et al. 2014; Xiong et al. 2019, 2016; Yap et al. 2010). The affected pathways may be responsible for the mechanisms underlying ASD. Although there are several metabolomic studies in ASD, a few literatures have focused directly on perturbed metabolic pathways. This review will comprehensively discuss and summarize the abnormal metabolic pathways in ASD patients from the previous metabolomic studies. Alterations of metabolomic profiles after receiving the pharmacological interventions from animal and human studies are also reviewed.

Abnormal metabolism associated with ASD from metabolomic studies in urine samples

The abnormal metabolisms associated with ASD from metabolomic studies in urine samples are listed in Table 1. These abnormalities reflected disturbances of various metabolic pathways. In the case of the pathways of oxidative damage, the oxidative stress markers were found to be positively correlated with the degrees of ASD severity (Damodaran and Arumugam 2011). Previous studies demonstrated that lower levels of antioxidants such as carnosine and anserine occurred in children with ASD compared to those of typical children (Liu et al. 2019; Ming et al. 2012; Yui et al. 2017). An increase in the levels of oxidative stress via nitric oxide production could be indicated by an excessive amount of arginine (Delwing et al. 2008), as demonstrated by the higher levels of N-acetylarginine in ASD compared to that of typical children (Diémé et al. 2015; Olesova et al. 2020). The proposed mechanism of oxidative stress might be implicated in the pathophysiology of ASD.

Table 1.

Abnormal metabolism associated with autism spectrum disorder from metabolomic studies in urine samples

Ref and Subjects Age (years) Method Categories of Metabo-
lite/ Metabolic pathway
involvement
Changes in metabolites compared to typical children
Interpretation
Increase Decrease
Gevi et al. 2020
40 ASD compared with 40 typical children 3-8 Untargeted approach using UHPLC-MS Neurotransmitter Glutamate GABA There are abnormalities of neurotransmitters and metabolites related to microbiota-brain-gut axis in the urine of children with ASD.
Pyridoxal phosphate
Neurotransmitter/microbiota-brain-gut axis 4-Cresol Noradrenaline
Ascorbate Adrenaline
Homovanillic acid MHPG
Vanillylmandelic acid
Liang, Ke et al. 2020
40 ASD compared with 40 typical children 3-12 Untargeted approach using UHPLC-QTOF/MS Nicotinate and nicotinamide metabolism Nicotinamide There are abnormalities of nicotinamide, phosphorylcholine, amino acid, acetyl-CoA synthase, lysine, nucleosides, neurotransmitter, naphthalene metabolites, sialic acid, purine, and lipid metabolism in urine of children with ASD.
Phosphorylcholine metabolism Phosphorylcholine
Amino acid metabolism Glycylglutamate
Alanylthreonine
Threonylaspartate
Histidylproline
Lysylproline
Valylmethionine
Methionylglutamine
Prolylserine
Acetyl-CoA synthase Acetylcarnitine
Tryptophan kynurenine pathway D-neopterin
7,8-Dihydroneopterin
Anthranilic acid
Catabolism of lysine 5-Aminopentanoic acid
Modified nucleosides 1-Methyladenosine
3’-O-Methylinosine
Neurotransmitter N-Acetylaspartylglutamate
Naphthalene metabolites 1-Naphthol
Sialic acid pathway N-Acetylneuraminic acid
Purine metabolism Deoxyinosine
Lipid metabolism Behenic acid
Other Bethanechol cation
S-Methyl-5’-thioadenosine
Liang, Xiao et al. 2020
22 ASD compared with 22 non-ASD siblings 3-9 Untargeted approach using 1H-NMR Cysteine metabolism Taurine There are abnormalities of cysteine, methionine, oxidative stress, sulfur, and tryptophan-serotonin-melatonin metabolic pathway in urine of children with ASD.
Methionine cycle Taurine
Reactive oxidative stress Taurine
Sulfur metabolism Taurine
Tryptophan-serotonin-melatonin pathway Tryptophan
Serotonin
Melatonin
Others Creatine Butyrate
Citrate
Lactate
Pantothenate
Trigonelline
Mussap et al. 2020
31 ASD compared with 26 typical children 2-11 Untargeted approach using GC-MS Oxidative stress Cystine 7-Methylxanthine
Uric acid
There are differences of metabolite levels resulting from oxidative stress, mitochondrial dysfunction, sugar metabolism, gut dysbiosis, and diet in urine of children with ASD compared to those of typically developing children.
Mitochondrial dysfunction Lactic acid
Sugar metabolism Scylloinositol
Gut dysbiosis Quinic acid
Hippuric acid
Tryptophan
Indole-3-acetic acid
Allyl thioacetic acid
Leucine
Diet 1-Methylhistidine
Others Aminomalonic acid
Olesova et al. 2020
24 ASD compared with 13 typical children 6-10 Targeted approach using UHPLC-QqQ-MS/MS Reactive oxidative stress Methylguanidine
N-acetylarginine
There are abnormalities of reactive oxidative stress and gut bacteria metabolism in the urine of children with ASD.
Gut bacteria metabolism Indoxyl sulphate
Indole-3-acetic acid
Liu et al. 2019
57 ASD compared with 81 typical children 2-12 Targeted approach using LC-MS/MS Ornithine (Urea) cycle
Ornithine (Urea) cycle
Ornithine-to-Citrulline ratio Proline There are abnormalities of ornithine (urea) cycle, methionine, lysine, reactive oxidative stress, tryptophan-serotonin metabolism in urine of children with ASD.
Arginine-to-Ornithine Citrulline
Arginine

4-Hydroxyproline
Aspartic acid
Methionine cycle Methionine sulfoxide Homocysteine
5-Aminovaleric acid
a-Aminoadipic acid
Ethanolamine
Lysine metabolism Lysine
Reactive oxidative stress Anserine
Carnosine
Tryptophan-Serotonin metabolism 5-Hydroxytryptamine
Others 2-Aminoisobutyric acid
Chen et al. 2019
156 ASD compared with 64 typical children NA Untargeted approach using GC/MS Phenylalanine metabolism Phenylactic acid There are abnormalities of phenylalanine, TCA cycle, bone metabolism, mammalian-microbial co-metabolism in urine of ASD.
TCA cycle Aconitic acid
Carboxycitric acid
Bone metabolism Phosphoric acid
Mammalian-microbial co-metabolism 3-Oxoglutaric acid
Carboxycitric acid
Others 3-Hydroxy-3-methylglutaric Fumaric
Creatinine N-Acetylcysteine
Oxalic Malonic
Pyruvic Tricarballylic
4-cresol Glycolic
2-hydroxybutyric Malic
Tartaric
3-Hydroxyglutaric
2-Oxoglutaric
Xiong et al. 2019
51 ASD compared with 51 typical children 3-7 Untargeted approach using HPLC-QTOF-MS Adenosine-Adenine pathway Adenine There are abnormalities of adenosine-adenine pathway, methylation regulation of retinoic acid-RORA pathway, and arginine metabolism, predominantly in female patient with ASD.
Methylation regulation of retinoic acid-RORA pathway 2-Methylguanosine
7alpha-Hydroxytestololactone
Arginine metabolism Creatinine Creatine
Bitar et al. 2018
40 ASD compared with 40 typical children NA Untargeted approach using 1H-NMR and LC-MS Glycine, serine and threonine metabolism Phosphoserine Threonine

Creatine

Serine
There are abnormalities in amino acids, nicotinic acid, TCA cycle, purine, vitamin, and fatty acid metabolism in urine of ASD.
Phenylalanine metabolism N-acetylphenylalanine

Tyrosine

Hydroxybenzoic acid
Glutamate, Arginine and Proline metabolism Glutamic acid Creatine

Hydroxyproline
Histidine metabolism Glutamic acid Urocanic acid
Cysteine and Methionine metabolism Phosphoserine Cysteic acid

Serine
Propanoate metabolism 2-hydroxybutyric acid
Nicotinate and nicotinamide metabolism Nicotinamide ribotide

Trigonelline
TCA cycle Citric acid
Purine metabolism 5-amino-imidazole-4-carboxamide Guanine
Vitamin B6 metabolism Riboflavin
Fatty acid oxidation N-amidino aspartic acid

Acetylcarnitine
Others Glycerol-3-phosphate

Cholic acid
Methyl acetoacetic acid
Lussu et al. 2017
21 ASD compared with 21 non-ASD siblings 4-16 Untargeted approach using 1H-NMR Neurotransmitter Glycine Glutamate There are abnormalities of neurotransmitter, tryptophan-serotonin, mammalian-microbial co-metabolism, oxidative stress metabolism in urine of ASD.
Tryptophan-serotonin metabolism Tryptophan
Mammalian-microbial co-metabolism Hippurate
Oxidative stress Taurine

Lactate
Others Creatine

D-threitol
Valine

Betaine

Creatinine
Xiong et al. 2016
62 ASD compared with 62 typical children 1.5-7 Untargeted approach using GC/MS Mammalian-microbial co-metabolism 3-(3-hydroxyphenyl)-3-Hydroxypropionic acid
3-Hydroxyphenylacetic acid
3-Hydroxyhippuric acid
There are abnormalities in mammalian-microbial co-metabolism in urine of ASD.
Gevi et al. 2016
30 ASD compared with 30 typical children 2-7 Untargeted approach using LC-MS Purine metabolism Inosine

Hypoxanthine

Xanthosine
Adenosine diphosphate There are abnormalities of nucleic acid (purine, pyrimidine), amino acid (tryptophan), vitamins (B1, B2, B6, and pantothenate), mammalian-microbial co-metabolism, glutathione, pentose phosphate pathway, benzoate degradation, and carbohydrate metabolism in urine of ASD.
Pyrimidine metabolism Uridine
Tryptophan metabolism Xanthurenic acid

Quinolinic acid

Tryptophan
Kynurenine
Mammalian-microbial co-metabolism Indolyl 3-acetic acid

Indolyl lactate
Disaccharide metabolism Trehalose/sucrose Cellobiose
Other amino acid metabolism Phenylalanine

Histidine
Methionine
Glutathione metabolism Pyroglutamic acid
Pentose Phosphate Pathway 6-phospho-D-gluconic acid

Ribose
Benzoate degradation p-cresol p-hydroxybenzoate
Vit B1 metabolism Thiamine
Vit B2 metabolism Riboflavin
Vit B6 metabolism 4-pyriodic acid
Pantothenate and CoA metabolism Glucose-6-phosphate
Glycolysis Glucose-6-phosphate
Diémé et al. 2015
30 ASD compared with 32 typical children NA Untargeted approach using 1H-NMR, 1H-13C-NMR and LC-HRMS Reactive oxidative stress N-acetylarginine Methylguanidine

Guanidinosuccinic acid
There are abnormalities in reactive oxidative stress, tyrosine, pyrimidine, and mammalian-microbial co-metabolism in urine of ASD.
Tyrosine metabolism Dihydroxy-1H-indole glucuronide I
Pyrimidine metabolism Dihydrouracil
Mammalian-microbial co-metabolism Indoxyl

Indoxyl sulfate

Alpha-N-Phenylacetyl-L-glutamine

p-cresol sulfate
Desaminotyrosine
Others N-Acetylasparagine

Valine

Glucuronic acid
Cozzolino et al. 2014
24 ASD compared with 21 typical children 6.9 +/−2.1 Untargeted approach using Solid-phase microextraction with GC/MS Leucine metabolism 3-Methylbutanal (under acid conditions) There are abnormalities in leucine, fatty acid, and degradation of carbohydrate metabolism in urine of ASD.
Peroxidation of fatty acid 3-Methylbutanal (under acid conditions)

2-Methylbutanal (under acid conditions)
Dietary degradation of carbohydrate 2-Methylmercaptofuran (under acid conditions)

2-Pentylfuran (under acid conditions)
Others 3-Methylcyclopentanone (under acid conditions) 2-Heptanone (under alkaline conditions)
Hexane (under acid conditions) Ethanol (under acid conditions)
2,3-Dimethylpyrazine (under alkaline conditions) Dimethyl trisulfide (under acid conditions)
2-Methylpyrazine (under alkaline conditions) Methoxy-phenyloxime (under alkaline conditions)
Isoxazole (under alkaline conditions) 3-Ethylpyridine (under alkaline conditions)

Acetophenone (under alkaline conditions)
Noto et al. 2014
21 ASD compared with 21 non-ASD siblings 4-16 Untargeted approach using GC/MS Mammalian-microbial co-metabolism 3-(3-Hydroxyphenyl)-3-hydroxypropanoic acid

Glycolic acid

4-Hydroxyhippuric acid
There are abnormalities of TCA cycle, Tryptophan, Glutathione, oxidative cleavage of N-acetylglucosamine, Tyrosine, pentose phosphate pathway, and mammalian-microbial co-metabolism in urine of ASD.
TCA cycle Aconitic acid
Tryptophan metabolism Tryptophan
Glutathione metabolism Pyroglutamic acid
Oxidative cleavage of N-acetylglucosamine Erythronic acid
Tyrosine pathway Phenylalanine

Tyrosine

4-Hydroxyphenylacetic acid

Homovanillic acid
Pentose Phosphate Pathway Ribose

Arabinofuranose

Threitol

Polyols arabitol

Xylitol
From dietary sources Fructose

1,2,3-Butanetriol

Propylene glycol
Mavel et al. 2013
30 ASD compared with 28 typical children 6-14 Targeted approach using 1H-NMR Organic acid Succinic acid There are abnormalities of organic acid and amino acid metabolism in urine of ASD.
Amino acid Glycine Creatine
β-alanine
Taurine
3-methylhistidine
Emond et al. 2013
26 ASD compared with 24 typical children 6-14 Untargeted approach using GC/MS Mammalian-microbial co-metabolism 1H-indole-3-acetate

Hippurate

3-hydroxyhippurate
There are abnormalities of organic acid, fatty acid, phenols, and mammalian-microbial co-metabolism in urine of ASD.
Organic acid Succinate

Glycolate
Fatty acid Palmitate

Stearate

3-methyladipate
Phenols p-Hydroxy mandelate

3-hydroxyphenylacetate
Ming et al. 2012
48 ASD compared with 53 typical children 6-14 Untargeted approach combined using UPLC-MS/MS and GC/MS Histidine catabolism Trans-Urocanate There are abnormalities of amino acid (Histidine, Lysine, Tryptophan, Leucine), reactive oxidative stress, and mammalian-microbial co-metabolism in urine of ASD.
Lysine and tryptophan catabolism Glutaroylcarnitine
Leucine catabolism 3-Methylglutaroylcarnitine
Mammalian-microbial co-metabolism 2-(4-Hydroxyphenyl) propionate 3-(3-Hydroxyphenyl) propionate
Taurocholenate sulfate 5-Amino-valerate
Reactive oxidative stress Carnosine

Urate
Amino acid and gamma-glutaryl amino acids Glycine

Serine

Threonine

Alanine

β-alanine

Histidine

Taurine

N-acetylglycine

Gamma-glutamylleucine

Gamma-glutamyltyrosine

Gamma-glutamylthreonine
Yap et al. 2010
39 ASD compared with 28 non-ASD siblings 34 typical children 3-9 Untargeted approach using 1H-NMR Nicotinic acid metabolism N-methyl-2-pyridone-5-carboxamide

N-methyl nicotinic acid

N-methyl nicotinamide
There are abnormalities of nicotinic acid, amino acid, mammalian microbial co-metabolism in urine of ASD
Amino acid metabolism Taurine

N-acetyl glycoprotein fragment

Succinate
Glutamate
Mammalian microbial co-metabolites Dimethylamine Hippurate

Phenylacetylglutamine
Nadal-Desbarats et al. 2014
30 ASD compared with 28 typical children 6-14 Targeted approach using combined 1H-NMR and 2D 1H-13C HSQC NMR TCA cycle Succinate There are abnormalities of TCA cycle and glutamate metabolism in urine of ASD.
Glutamate metabolism Glutamate
Others 3-methylhistidine

ASD autism spectrum disorder, TCA tricarboxylic acid cycle, UPLC-MS/MS ultra-performance liquid chromatography—tandem mass spectrometer, UHPLC-MS ultra-high-performance liquid chromatography-mass spectrometry, UHPLC-QTOF/MS ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry, UHPLC-QqQ-MS/MS ultra-high-performance liquid chromatography-tandem triple quadrupole-mass spectrometry, LC–MS liquid chromatography–mass spectrometry, LC–MS/MS liquid chromatography–tandem mass spectrometry, LC-HRMS liquid chromatography–high resolution mass spectrometry, GC–MS gas chromatography–mass spectrometry, 1H-NMR hydrogen-1 nuclear magnetic resonance, 1H-13C-NMR hydrogen-1 and carbon-13 nuclear magnetic resonance, 2D 1H-13C HSQC NMR two-dimensional hydrogen-1 and carbon-13 heteronuclear single quantum correlation nuclear magnetic resonance; MS/MS: tandem mass spectrometry, HPLC-QTOF-MS high-performance liquid chromatography-quadrupole-time of flight mass spectrometry, GABA gamma-aminobutyric acid, RORA related orphan receptor alpha, NA data not available

Mammalian-microbial co-metabolism was also found to be associated with ASD symptoms. The comparison of the urine metabolite levels between children with autism and typical development showed the reduced levels of carboxycitric acid and 3-oxoglutaric acid (Chen et al. 2019), while the levels of hippurate (Kałużna-Czaplińska 2011; Lussu et al. 2017), 3-(3-hydroxyphenyl)-3-hydroxypropionic acid (HPHPA) (Noto et al. 2014; Xiong et al. 2016), 3-hydroxyphenylacetic acid (3HPA) (Xiong et al. 2016), 3-hydroxyhippuric acid (3HHA) (Xiong et al. 2016), indole-3-acetic acid (Gevi et al. 2016; Olesova et al. 2020), indolyl lactate (Gevi et al. 2016), and glycolic acid (Noto et al. 2014) were increased in children with ASD compared to children with typical development. However, these results were inconsistent with some studies (Emond et al. 2013; Yap et al. 2010), in which the decreased levels of hippurate was shown. These contrary results could be as a result of unrestricted food among participants prior to the sample collection and heterogeneity of ASD.

Abnormalities of various pathways associated with amino acids, including tryptophan, arginine, phenylalanine, tyrosine, leucine, histidine, and methionine were also observed in cases of ASD. Tryptophan has been the most discussed amino acid associated with the development of ASD. Liang et al. (2020b) showed evidence supporting the association between tryptophan-serotonin-melatonin pathways and autism. In fact, they found increased tryptophan and serotonin levels while decreased melatonin in the urine of children with ASD (Liang et al. 2020b). This finding was in line with the dysfunction of melatonin that leads to sleep–wake rhythm disturbance in patients with ASD (Wu et al. 2020). Another study by Liang et al. (2020a, b) was found an increase in 7,8-dihydroneopterin and neopterin in the urine of children with ASD. The increase in these two metabolites, which are the markers of reactive oxygen species, were proposed to be related to the disruption of tryptophan kynurenine pathway and pathogenesis of autism (Liang et al. 2020a, b). Prior studies observed a higher concentration of 5-hydroxytryptamine (Liu et al. 2019), a downstream metabolite of tryptophan, and tryptophan (Gevi et al. 2016; Lussu et al. 2017; Noto et al. 2014) in the urine samples of children with ASD compared to that of typical children. Additionally, the disturbance of tryptophan metabolism was revealed by the alteration of multiple tryptophan intermediates, including xanthurenic acid, quinolinic acid, and kynurenine in the urine samples of ASD cases (Gevi et al. 2016). Abnormalities in arginine metabolism were also exhibited, as indicated by the increased levels of creatinine while decreased levels of creatine in the urine of female children with ASD (Xiong et al. 2019). The abnormalities regarding arginine pathway-related enzymes and receptors could be explained by creatine is one of the byproducts of this pathway. Abnormalities in phenylalanine metabolism were shown as the higher excretion of phenylactic acid and phenylalanine in the urine of children with ASD compared to that of typical children (Chen et al. 2019; Gevi et al. 2016; Noto et al. 2014). Disturbance of tyrosine metabolism was also proposed in some studies, as alterations of tyrosine intermediates in children with ASD were found (Diémé et al. 2015; Noto et al. 2014). Leucine metabolism was also assumed to be dysfunctional, as indicated by the increased levels of 3-methylbutanal, a leucine intermediate, in children with ASD (Cozzolino et al. 2014). Histidine catabolic dysfunction in cases of ASD was also revealed in a study by Ming et al. (2012). Indeed, they found the increased levels of a histidine intermediate–urocanate in children with ASD compared to the controls (Ming et al. 2012). This was supported by the discovery of urocanase deficiency, which leads to excessive excretion of urocanic acid and manifestations of developmental delay (Espinós et al. 2009). Abnormalities in the methionine cycle were found as the levels of methionine sulfoxide increased and homocysteine decreased in the urine of children with ASD compared to those of typically developing children (Liu et al. 2019). Those two metabolites act as methyl group donors; therefore, these findings supported the association between abnormal methylation and the development of ASD (Behnia et al. 2015; Hannon et al. 2018; Ladd-Acosta et al. 2014).

There are evidence links between abnormalities of amino acid metabolism and the imbalance of neurotransmitters. Decreased lysine and ethanolamine levels were exhibited in urine from children with ASD and were thought to be associated with the lower levels of the excitatory neurotransmitters glutamate and acetylcholine (Liu et al. 2019). On the other hand, increased levels of 2-aminoisobutyric acid which is assumed to play a similar role as an inhibitory neurotransmitter, gamma aminobutyric acid (GABA), were observed in the urine of children with ASD compared to typically developing children (Liu et al. 2019). Decrease in excitatory neurotransmitter, glutamate, but increases in the inhibitory neurotransmitter, glycine, have also been demonstrated in several studies (Lussu et al. 2017; Nadal-Desbarats et al. 2014; Xiong et al. 2016). In contrast with other prior studies, an increase in urine glutamate were discovered in children with ASD (Bitar et al. 2018; Gevi et al. 2020). A reduction of urine GABA in children with ASD was explained by the possible link of the microbiota-gut-brain axis. The increased 4-cresol, a consequence of gut dysbiosis, inhibits dopamine β-hydroxylase that is responsible for converting dopamine to noradrenaline. Therefore, the accumulation of dopamine results in the reduction of GABA (Gevi et al. 2020). These inconsistent results could be due to the different ethnicities and spectrometric techniques among the studies.

Regarding the urea cycle pathway, there was a positive association between ASD and blood ammonia (Cohen 2002; Wang et al. 2012). The proposed mechanism responsible for the elevation of ammonia was an increase in the conversion of proline to 4-hydroxyproline by the enzyme pyrroline-5-carboxylate reductase (Mitsubuchi et al. 2008). In addition, the dysfunction of the enzymes arginine transcarbamylase and ornithine transcarbamylase led to the malfunction of the urea cycle pathway, as Liu et al. (2019) showed by demonstrating an increased metabolic ratio of ornithine-to-citrulline and arginine-to-ornithine in the urine of children with ASD.

It has been speculated that abnormalities in the TCA cycle was one of the potential mechanisms to be related to ASD. Previous studies had revealed a decrease in aconitic acid, carboxycitric acid, and citric acid in the urine samples of children with ASD compared with those of typical children (Bitar et al. 2018; Chen et al. 2019). On the other hand, another study reported increased cis-aconitic acid levels in children with ASD (Noto et al. 2014). Based on these findings, decreased levels of such metabolites were believed to be associated with aconitase deficiency (Noto et al. 2014). The inconsistent findings of aconitic levels were possibly due to the difference in ethnicity.

Abnormalities in purine metabolism, such as the adenosine-adenine pathway, in cases of ASD have been reported. These abnormalities were evidenced by increased levels of aminoimidazole (Bitar et al. 2018), inosine, hypoxanthine, and xanthosine (Gevi et al. 2016), but decreased levels of guanine (Bitar et al. 2018) and dihydrouracil (Diémé et al. 2015) in children with ASD compared to typically developing children.

There is some evidence to show alteration of both nicotinate and nicotinamide metabolism in cases of ASD. Indeed, previous studies demonstrated increased levels of nicotinamide (Bitar et al. 2018; Liang et al. 2020a, b; Yap et al. 2010) in urine compared to that of typically developing children. Excessive excretion of such metabolites was thought to cause damage to neurons and had been previously implicated in Parkinson’s disease (Willets et al. 1993; Williams et al. 1993). An increased nicotinic acid level has also been related to ASD (Melke et al. 2008). This alteration could be due to diminished conversion of tryptophan to serotonin by reduced acetylserotonin methyltransferase enzyme activity (Melke et al. 2008).

With regard to the abnormalities of carbohydrate metabolism, a study proposed that the lower activity of intestinal carbohydrate degradation enzymes might lead to the lower levels of furans in cases of ASD (Cozzolino et al. 2014). There was the first-time reporting of a significant reduction of urine scylloinositol in children with ASD (Mussap et al. 2020). A similar metabolome pattern of scylloinositol was also found in the brain of rats with ASD-like features (Toczylowska et al. 2020). These two studies might support the evidence of the association between inositol metabolism and autism. There are also abnormalities in the pentose phosphate pathway, as indicated by increased levels of pentose phosphate pathway-related metabolites such as ribose and arabinofuranose in children with ASD (Noto et al. 2014).

The alteration of lipid peroxidation could explain increased aldehydes such as 3-methylbutanal and 2-methylbutanal in ASD patients, because such aldehydes can derive from free radical-mediated lipid peroxidation (Cozzolino et al. 2014). A decrease in acetylcarnitine in urine samples of children with ASD (Bitar et al. 2018) supports the alteration of fatty acid oxidation. Also, the decreased level of acetylcarnitine could lead to impaired energy transportation. Thus, some ASD behaviors can be improved by L-carnitine supplementation (Frye 2015).

The distinguishable set of metabolites by metabolomics study in urine mentioned above could explain the underlying abnormal metabolic pathways in patients with ASD including the antioxidants or oxidative stress pathway, mammalian-microbial co-metabolism pathway, metabolism of some amino acids such as tryptophan and phenylalanine. Interestingly, the metabolite of such metabolic pathways might be possible biomarkers for ASD diagnosis that needed to be further investigated.

Abnormal metabolism associated with ASD from metabolomic studies in blood samples

The abnormal metabolic pathways associated with ASD from metabolomic studies in blood samples are listed in Table 2. The abnormalities pertinent to various amino acids, including tryptophan, tyrosine, and branched-chain amino acid pathways in blood samples have been reported as being associated with ASD. Abnormal tryptophan metabolism was indicated by decreased levels of N-formylanthranilic acid, a downstream metabolite of the tryptophan-derived kynurenine pathway, but increased serotonin levels in children with poor communication skills compared to those with typical skills (Kelly et al. 2019). Perturbation in tyrosine metabolism was also found to be correlated with ASD. A previous study revealed that blood levels of N-formylphenylalanine, which is a tyrosine intermediate, of children with a low communication score were lower than that of children with a typical score (Kelly et al. 2019). Furthermore, alterations in the metabolism of branched-chain amino acids (BCAA) were found in patients with ASD, as indicated by higher blood levels of alpha-keto derivatives from BCAA catabolism (Rangel-Huerta et al. 2019). The increase in ratios of specific amino acids such as glutamine, glycine, and ornithine with BCAA was reported in children with ASD (Smith et al. 2019). These abnormalities were assumed to be caused by a deficiency in the branched-chain ketoacid dehydrogenase kinase enzyme (Smith et al. 2019). The dysfunction of this enzyme eventually leads to a lower concentration of BCAA in plasma, resulting in neurological impairment (Novarino et al. 2012).

Table 2.

Abnormal metabolism associated with autism spectrum disorder from metabolomic studies in blood samples

Ref and Subjects Age (years) Method Categories of Metabo-
lites/ Involved metabolic
pathways
Changes in metabolites compared to typical children
Interpretation
Increase Decrease
Kelly et al. 2019
403 children (365 categorized to “on schedule” and 38 categorized to “requiring further monitoring/evaluation” from ASQ) 3 Targeted approach using UPLC-MS/MS Tyrosine metabolism N-formylphenylalanine There are abnormalities of amino acid (tyrosine, tryptophan, arginine, proline, methionine, cysteine, and taurine), lipid (phospholipid, sphingolipid, and fatty acid), urea cycle, and xenobiotics metabolism in children with poor ASQ communication skills, relative to children with typical communication development.
Phospholipid metabolism trimethylamine N-oxide
Food Component/Plant Cinnamoylglycine
Erythritol
Pyrraline
Endocannabinoid Oleoyl ethanolamide
Palmitoyl ethanolamide
Linoleoyl ethanolamide
Tryptophan metabolism Serotonin 5-hydroxyindoleacetate
N-formylanthranilic acid
Sphingolipid metabolism Sphingomyelin
Fatty acid metabolism Docosahexaenoylcarnitine
Urea cycle Prolylhydroxyproline
Arginine and Proline metabolism Prolylhydroxyproline
Methionine, cysteine, and Taurine metabolism Alpha-ketobutyrate
Smith et al. 2019
516 ASD compared with 164 typical children 1.5-4 Targeted approach using LC-MS BCAA metabolism Glutamine-to-isoleucine ratio
Glutamine-to-leucine ratio
Glutamine-to-valine ratio
Glycine-to-isoleucine ratio
Glycine-to-leucine ratio
Glycine-to-valine ratio
Ornithine-to-isoleucine
Ornithine-to-leucine ratio
Ornithine-to-valine ratio
There are abnormalities of BCAA metabolism in plasma of ASD.
Rangel-Huerta et al. 2019
30 ASD compared with 30 typical children 2-6 Untargeted approach using LC-MS Neurotransmitter Glutamate There are abnormalities of neurotransmitter, reactive oxidative stress, BCAA, NAD, phospholipid metabolism in plasma of ASD.
Reactive oxidative stress Arginine
N-acetylarginine
Homoarginine
BCAA metabolism 3-methyl-2-oxobutyrate
3-methyl-2-oxovalerate
4-methyl-2-oxopentanoate
Isovalerylcarnitine
Isobutyrylcarnitine
NAD+ metabolism 1-methylnicotinamide
N-methyl-2-pyridone-5-carboxamide
Nicotinamide
Phospholipid metabolism 1-palmitoyl-glycerol-phosphatidyl-etholamine
1-stearoyl-glycerol-phosphatidyl-etholamine
Others n-alpha-acetylornithine
Tryptophan
Kynurenine
5-bromotryptophan
3-indoxyl sulfate
Indole lactate
6-hydroxyindole sulfate
Orozco et al. 2019
167 ASD 51 i-DD 31 DS 193 typical children 2-5 Untargeted approach using 1H-NMR One carbon metabolism Glycine
Serine
There are abnormalities of one carbon metabolism, TCA cycle, urea cycle in plasma of ASD.
TCA cycle Cis-aconitate
Urea cycle Ornithine
Lv et al. 2018
60 ASD compared with 30 typical children 2-5 Targeted approach using MS/MS Fatty acid metabolism Free carnitine
Glutaryl carnitine
Octyl carnitine
Twenty-four carbonyl carnitine
Carnosyl carnitine
There are abnormalities of fatty acid metabolism (free carnitine, short and long chain acyl carnitines) in plasma of ASD.
Anwar et al. 2018
38 ASD compared with 31 typical children 5-12 Targeted approach using LC-MS Protein glycation Nε-carboxymethyllysine

Nω-carboxymethyllysine
3-deoxyglucoosone-derived hydroimidazolone There are abnormalities of protein glycation and protein oxidation metabolism in plasma of ASD.
Protein oxidation Dityrosine
Wang et al. 2016
173 ASD compared with 163 typical children 3-6 Untargeted approach using UPLC/Q-TOF MS/MS Fatty acid metabolism Docosahexaenoic acid
Docosapentaenoic acid
There are abnormalities of fatty acid, sphingomyelin, lysophosphatide, and beta oxidation in plasma of ASD.
Sphingomyelin metabolism Sphingosine-1-phosphate
Lysophosphatide metabolism LPA
LysoPE
Fatty acid beta oxidation Decanoylcarnitine 9,10-Epoxyoctadecenoic acid
Others Pregnanetriol Adrenic acid
Uric acid
West et al. 2014
52 ASD compared with 30 typical children 4-6 Targeted approach using GC-MS and untargeted approach using LC-HRMS TCA cycle Succinate Citrate There are abnormalities of TCA cycle, fatty acid, oxidative phosphorylation, mitochondrial dysfunction, and gut microbiome metabolism in plasma of ASD.
Fatty acid metabolism and oxidative stress Methylhexa-decanoic acid
Hepta-decanoic acid
Oxidative phosphorylation Aspartate
Glutamate
Mitochondrial energy production DHEA-S Isoleucine
Gut microbiome 4-hydroxyphenyllactate
Others Glutaric acid
3-aminoisobutyric acid
Creatinine
Homocitrulline
Kuwabara et al. 2013
25 ASD compared with 28 typical children 25-40 male Untargeted approach using CE-TOF-MS Oxidative stress Arginine
Taurine
5-Oxoproline There are abnormalities of oxidative stress and mitochondrial metabolism in plasma of ASD.
Mitochondrial dysfunction Lactic acid
Delaye et al. 2018
22 ASD 29 ID 30 typical children > 18 Targeted approach using amino acid chromatography Neurotransmitter Glutamate
Serine
There are abnormalities of neurotransmitter metabolism in plasma of ASD.
Others Proline
Ornithine

ASD autism spectrum disorder, i-DD idiopathic-developmental delay, DS Down syndrome, ID intellectual disability, ASQ Ages and Stages Questionnaire, UPLC-MS/MS ultra-performance liquid chromatography-tandem mass spectrometer, LC–MS liquid chromatography–mass spectrometry, GC–MS gas chromatography-mass spectrometry, 1H-NMR hydrogen-1 nuclear magnetic resonance, MS/MS tandem mass spectrometry, UPLC/Q-TOF–MS ultra-high performance liquid chromatography-quadrupole time-of-flight mass spectrometry, LC-HRMS liquid chromatography-high resolution mass spectrometry, CE-TOF–MS capillary-electrophoresis-time-of-flight-mass-spectrometer, NAD nicotinamide adenine dinucleotide, LPA lysophosphatidic acid, LysoPE lysophosphatidylethanolamine, BCAA branched-chain amino acids, TCA tricarboxylic acid cycle, DHEA-S dehydroepiandrosterone sulfate

Alteration in the endocannabinoid metabolic pathway may be related to ASD. Endocannabinoids are neuromodulators that play a critical role in regulating emotional, behavioral, and social interaction. Kelly et al. (2019) found increased levels in specific endocannabinoids in children predicted to be at risk for ASD.

Disturbed neurotransmitter homeostasis has been proposed as possible pathogenesis of ASD. The decrease of glutamate level in the striatum was related to the symptom severity in autistic adult patients (Horder et al. 2018). Also, the perturbation of the malate-aspartate shuttle activity presented by the mitochondrial system might affect glutamate function (Monné et al. 2019). This was supported by the perturbation of glutamate-aspartate transporter activity in the gray matter of children with ASD (Palmieri et al. 2010). Additionally, two previous studies revealed decreased concentrations of glutamate in blood samples of children with ASD compared to age-matched controls (Delaye et al. 2018; Rangel-Huerta et al. 2019). On the other hand, some studies demonstrated increased blood levels of glutamate in cases of ASD (Cai et al. 2016; Hassan et al. 2013; Naushad et al. 2013; Tirouvanziam et al. 2012). These different findings might be due to the variation in the age range of the samples, study methodology and sample collection techniques, or the possible heterogeneity nature of the subjects.

ASD symptoms have been associated with pathways of oxidative damage (Chauhan and Chauhan 2006). This was suggested by the abnormalities in the nitric oxide pathway, as indicated by increased levels of arginine and its downstream metabolites in blood samples of children with ASD, especially those with developmental regression (Kuwabara et al. 2013; Rangel-Huerta et al. 2019). Moreover, an increase in blood taurine levels in ASD cases was observed (Kuwabara et al. 2013). Taurine is thought to be a neuroprotective compound against oxidative stress reactions (Niu et al. 2018), and therefore an increased concentration of taurine might reflect the compensation of oxidative stress.

Nicotinamide adenine dinucleotide (NAD) metabolism is also related to ASD, as indicated by the lower levels of nicotinamide in the plasma of children with ASD than that of typical children (Rangel-Huerta et al. 2019). NAD synthesis is important in several metabolic pathways, such as the TCA cycle, glycolysis, and fatty acid oxidation. The assumption is that decreased levels of nicotinamide might be due to the less preferential transformation of tryptophan to the melatonin pathway, leading to an accumulation of oxidative stress (Mussap et al. 2016).

Lipid metabolism complications have been assumed to be a causative mechanism of ASD. Metabolomic profiles in the blood revealed decreased levels of lysolipids (Rangel-Huerta et al. 2019), free carnitine (Lv et al. 2018), short- and long-chain acylcarnitines (Lv et al. 2018), and docosahexaenoic acid (DHA) (Wang et al. 2016) in children with ASD in comparison to age-matched controls. Lower levels of polyunsaturated fatty acids that play a vital role in the structural and functional integrity of neurons were found in children with ASD (Mazahery et al. 2017; Parletta et al. 2016). The addition of DHA as a supplement could potentially improve behavior in children with ASD, adding weight to this finding (Amminger et al. 2007; Yui et al. 2012). In addition, abnormal white matter development has been proposed as being related to abnormal sphingomyelin metabolism in ASD, as indicated by increased levels of sphingosine-1-phosphate in the blood of children with ASD (Wang et al. 2016).

The link of Urea cycle defects to ASD was supported by the evidence of higher ornithine levels in blood samples of children with ASD (Orozco et al. 2019), which might result from ornithine transcarbamylase (OTC) dysfunction. The evidence of OTC deficiency related to ASD-like symptoms was presented in a case report (Görker and Tüzün 2005).

Disruption of the TCA cycle has been reported as being associated with ASD. This was evidenced by the increased blood levels of cis-aconitate (Orozco et al. 2019) and succinic acid (West et al. 2014), but decreased blood levels of citric acid (West et al. 2014) in children with ASD compared to children with typical development.

Proteotoxic stress induced by protein glycation and oxidation may be linked to the pathogenesis of ASD. The elevation of advanced glycation end products (AGEs) and dityrosine residue were revealed in the plasma of children with ASD (Anwar et al. 2018). It is proposed that the accumulation of AGEs is associated with perturbation of the lipid peroxidation process, and the increased levels of markers of this process, such as hexanoyl-lysine adduct, urinary 8-isoprostane-F2a, and plasma malondialdehyde, have also been related to ASD symptoms (Chauhan and Chauhan 2006; Ghezzo et al. 2013; Ming et al. 2005). The increased level of dityrosine was a result of increased dual oxidase (DUOX) activity, which was related to gut mucosal dysfunction (Bae et al. 2010; Chang et al. 2013).

The disruption of oxidative phosphorylation in mitochondria could also be related to ASD. The higher levels of aspartate and glutamate in blood samples of children with ASD may be linked the aspartate/glutamate transporter gene mutation, SLC25A12 (West et al. 2014). SLC25A12 plays a crucial role in the malate/aspartate shuttle producing cellular energy via oxidative phosphorylation (West et al. 2014). The disruption of the respiratory chain reaction was also indicated by the increased levels of dehydroepiandrosterone (DHEAS) in children with ASD (West et al. 2014). DHEAS can inhibit the enzymes related to the mitochondrial respiratory chain process, thus interfering with energy production (Safiulina et al. 2006).

Alteration in mammalian-microbial co-metabolism has also been considered as being a mechanism involved in ASD pathogenesis. Decreasing levels of 4-hydroxyphenyllactate were observed in the blood of children with ASD compared to typical children (West et al. 2014). This metabolite is related to bacteria: lactobacilli and bifidobacteria, which act as antioxidants (Beloborodova et al. 2012). This finding was supported by decreased antioxidant enzymes such as superoxide dismutase and glutathione peroxidase in the blood of children with ASD (Meguid et al. 2011; Yorbik et al. 2002).

The alteration of metabolomes in the blood samples could reflect the several abnormal metabolic pathways associated with children with ASD. The metabolism of some amino acids, such as tryptophan and BCAA, and neurotransmitters, such as glutamate, reactive oxygen species, and lipids, might play an important role in ASD pathogenesis. Further investigation on novel biomarkers from those abnormal metabolic pathways was needed.

Pharmacological interventions for ASD: metabolomic studies in animals

The research into pharmacological interventions for ASD from metabolomic studies in animals is limited, as shown in Table 3. The intervention in animal studies is suramin, an antipurinergic agent. Naviaux et al. (2014) gave suramin to mice from maternal immune activation (MIA) models. The concept of MIA model was to make the offspring mice having the features of ASD by letting them exposed to the poly(Inosine:Cytosine), which was intraperitoneally injected to the pregnant dams. Exposure to poly(Inosine:Cytosine) could lead to cell danger response (CDR), which is considered a metabolic response mechanism to a threat (Naviaux 2014). They found the near-normalization of blood metabolomic profiles in treated mice had improved social and novelty preference testing. (Naviaux et al. 2014). The metabolomics profiles alterations included increased levels of adenosine triphosphate and allantoin, whereas decreased levels of adenosine monophosphate, inosine monophosphate, deoxyadenosine monophosphate, deoxyinosine monophosphate, guanosine, inosine triphosphate, and guanosine diphosphate. The purinergic signaling regulates the CDR. When there was a perturbation in purine metabolism, it causes the persistence of CDR which leads to several chronic diseases such as ASD (Naviaux 2014). Hence, antipurinergic therapy has been thought to correct the dysregulation of purine metabolism. Antipurinergic therapy, in addition to mitigate autistic-like symptoms in ASD caused by the environmental factor (MIA model), could improve such symptoms in ASD caused by a genetic factor such as Fragile X (Fmr1 knockout) mouse model. Fragile X syndrome is the most common known inherited single-gene disorder associated to ASD (Cogram et al. 2020). The study of Naviaux et al. (2015) emphasized the association of ASD features and perturbed purinergic signaling pathway by showing the normalization of purine metabolism-related metabolites and restoration of typical social behaviors in Fmr1 knockout mice treated by antipurinergic therapy (Naviaux et al. 2015).

Table 3.

Pharmacological interventions for autism spectrum disorder: metabolomic studies in animals

Ref and Model Age Intervention Dose/
duration
Method Metabolic pathway Changes in metabolites compared to
controls
Outcomes Interpretation
Increase Decrease
Naviaux et al. 2014
Male mice from MIA model of ASD features vs. controls 6 mo Suramin 20 mg/kg intraperitonal single dose vs. NSS Targeted approach using LC-MS in blood Purine metabolism Inosine Adenosine monophosphate Improvement in social and novelty preferences testing Single dose of suramin could reverse social deficit in MIA model by normalization of metabolites in many metabolic pathways, mainly in purine metabolism.
Deoxyinosine Inosine monophosphate
Adenosine triphosphate Deoxyadenosine monophosphate
Allantoin Deoxyinosine monophosphate
Guanosine
Inosine triphosphate
Guanosine diphosphate
Microbiome metabolism Benzoic acid Hydroxyphenylacetic acid
Histidinol Sucrose-6-phosphate
dTDP-glucose 3-Hydroxyanthranilic acid
Naviaux et al. 2015
Fragile X (Fmr1) knockout mouse model vs. controls 9 wk Suramin 20 mg/kg intraperitonially weekly 16 wk vs. NSS Targeted approach using LC-MS/MS in blood Purine metabolism Xanthine
Hypoxanthine
Inosine
Guanosine
Restoration of social preference, social novelty, and spontaneous altered behavior Suramin could restore typical social behaviors in Fmr1 knockout mouse model by normalization of metabolites in many metabolic pathways, mainly in purine metabolism.
Fatty acid oxidation Hexanoylcarnitine
3,5-Tetradecadiencarnitine
Myristoylcarnitine
Eicosanoid metabolism Leukotriene B4


Epoxy-5,8,11-eicosatriencic acid
8,9-Epoxyeicosatrienoic acid
11-Dehydrothromboxane B2

LC–MS liquid chromatography-mass spectrometry, LC–MS/MS liquid chromatography–tandem mass spectrometry, APT antipurinergic therapy, RTT Rett syndrome, NSS normal saline solution, MIA model maternal immune activation model, mo months, wk weeks

In summary, the metabolomic studies using pharmacologic intervention for ASD demonstrated abnormalities in various metabolic pathways, as shown in Fig. 1, including purine metabolism, microbiome, fatty acid oxidation, TCA cycle, and eicosanoid metabolism. Near-normalization of the metabolite levels following those pharmacological interventions, such as suramin, was associated with a near-restoration of social behaviors in mice with autistic features.

Fig. 1.

Fig. 1

The altered metabolic pathways from the combination of pharmacologic interventions in Tables 3 and 4 were shared by animals with ASD features and humans with ASD. Four metabolic pathways were shared by animals with ASD features. Seventeen metabolic pathways were shared by humans with ASD after therapeutic trials. The potential common pathways shared by both animal and human studies were mammalian-microbial co-metabolite, purine metabolism, and fatty acid oxidation

Pharmacological interventions for ASD: metabolomic studies in humans

Details of the metabolomic studies of pharmacological interventions for ASD in humans are shown in Table 4. Those interventions include sulforaphane, and prebiotic: bimuno galactooligosaccharide (B-GOS).

Table 4.

Pharmacological interventions for autism spectrum disorder: metabolomic studies in humans

Ref and Model Age Intervention
Dose/duration
Method Categories of Metabolites/
Involved metabolic pathways
Changes in metabolites
Clinical Outcomes Interpretation
Increase Decrease
Bent et al. 2018
15 ASD children before and after intervention 5-22 y Sulforaphane 2.5 μmol glucoraphanin per lb/12 wk Untargeted approach using UHPLC-MS/MS in Urine Arginine N-acetylputrescine Improvement of behavior (ABC) and social (SRS) responsiveness score Sulforaphane supplementation statistically significantly improves behavior and social responsiveness by changes in urine metabolites involved in several pathways including amino acid, cholesterol, neurotransmitter, oxidative stress, polyol associated with uremia, sphingomyelin, TCA cycle, and phospholipid metabolism.
Tryptophan Tryptophan
Tyrosine Tyrosine
Leucine metabolism β-hydroxyisovalerate
α-hydroxyisocaproate
Isoleucine α-hydroxyisovalerate
Valine 3-hydroxyisobutyrate
Cysteine Taurine
Mammalian-microbial co-metabolism Tryptophan betaine
4-Hydroxybenzoate
3-Ethylphenylsulfate
Arabinose
Erythriol
Cholesterol Cholesterol
Cholate
12-Dehydrocholate
Cholesterol (bile acid), microbiome origin Glycocholenate sulfate
Cholesterol (hormone) Cortisone
Cortisol 21-glucuronide
Epiandrosterone glucuronide
17α-hydroxypregnanolone gluconide
5α-androstan-3β,17α-diol disulfate
Pregnen-diol disulfate
Dehydroepiandrosterone
11-Ketoetiocholanolone sulfate
5α-pregnan-3β,20α-diol disulfate
5α-pregnan-3(α/β),20β-diol disulfate
21-Hydroxypregnenolone disulfate
Neurotransmitter Glutamine
Serotonin
Homovanilate
Hypoxanthine
Oxidative stress γ-Glutamylglutamine
Methionine sulfone
Polyol associated with uremia 3-Carboxy-4-methyl-5-propyl-2-furanpropanoate
Sphingomyelin Stearoyl sphingomyelin
Lignoceroyl sphingomyelin
Behenoyl sphingomyelin
Sphingomyelin
TCA cycle Malate
Phospholipid 1-Palmitoyl-2-oleoyl--sn-glycero-3-phosphoethanolamine
Grimaldi et al. 2018
30 ASD on exclusion or unrestricted dwiet with intervention vs. placebo 4-11 y B-GOS® 6 wk vs. placebo Targeted approach using 1H-NMR in Feces and Urine Amino acid Dimethylglycine (urine) Phenylacetylglycine (urine) Improvement of antisociability and social skill scores Prebiotics (B-GOS) intervention, especially with exclusion diet, help improvements in antisocial behaviors in ASD by modulating gut microbiota and changing in fecal and urine metabolites.
Dimethylglycine (feces) Phenylalanine (urine)
Dimethylalanine (urine) Isoleucine (feces)
Leucine (feces)
Valine (feces)
Alanine (feces)
Glutamine (feces)
Fatty acid Adipate (urine)
Butyrate (feces)
Valerate (feces)
Organic acids Citrate (urine) Lactate (feces)
B-hydroxybutyrate (urine)
Others Creatinine (urine)
Creatine (urine)
Carnitine (urine)
Trimethylamine-N-oxide (urine)
Ethanol (feces)
Naviaux et al. 2017
10 ASD randomized to receive either suramin vs. placebo 4-17 y Suramin 20 mg/kg single dose VS saline infusion Targeted LC-MS/MS Blood Microbiome 2-Keto-L-gluconate
P-Hydroxyphenylacetic acid
4-Hydroxyphenyllactic acid
ADOS-2 comparison scores improved in suramin group but did not change in placebo group Low-dose suramin was associated with improvement of ASD symptoms measured by ADOS-2.
Sphingomyelin Sphingomyelin(d18:1/26:0 OH)
1-carbon, folate, glycine, serine Glycine
Purines 1-Methyladenine cAMP
Purine
Allantoin
dGDP
Amino acids Alanine
L-Asparagine
Pyrimidines Cytosine
Krebs cycle Citric acid
Cis-aconitic acid
GABA, Glutamate, Arginine, Ornithine, Proline 1-Pyrroline-5-carboxylic acid
Dipeptides Gamma-glutamyl-Alanine
Histidine, histamine Histamine
Nitric oxide, ROS Azelaic acid
SAM, SAH, Met, Cys, GSH Methionine sulfoxide
Cysteamine
S-Adenosylhomocysteine
Tryptophan, Kyurenine L-Kynurenine
Glycolysis Glycerol 3-phosphate
Bile acids Chenodeoxyglycocholic acid
Glycocholic acid
Vitamin C Hydroxyproline
Branch chain amino acids 2-Hydroxyisovaleric acid
Isovalerylglycine
Tiglylglycine
Disaccharides Hexose Disaccharide pool
Tyrosine, phenylalanine L-Phenylalanine
Cinnamoylglycine
Cholesterols Lathosterol
Fatty acid oxidation Octanoylcarnitine

ASD autism spectrum disorder, ADOS-2 The Autism Diagnostic Observation Schedule-Second Edition, B-GOS® Bimuno® galactooligosaccharide, LC–MS liquid chromatography-mass spectrometry, LC–MS/MS liquid chromatography–tandem mass spectrometry, 1H-NMR hydrogen-1 nuclear magnetic resonance, UHPLC-MS/MS ultra-high performance liquid chromatography-tandem mass spectrometry, SM sphingomyelin, cAMP cyclic adenosine monophosphate, dGDP 2’-deoxy-guanosine diphosphate, GABA Gamma-aminobutyric acid, SAM S-adenosylmethionine, SAH S-adenosylhomocysterin, Met methionine, Cys cysteine, GSH glutathione, TCA tricarboxylic acid cycle, ABC Aberrant Behavior Checklist, SRS Social Responsiveness Scale, y year, wk weeks

Sulforaphane

There has been a growing interest in sulforaphane, a substance extracted from broccoli sprouts, as a possible treatment option for ASD patients (McGuinness and Kim 2020). Sulforaphane is known for its proposed mechanism to reverse the perturbed metabolic pathways associating with ASD, including mitochondrial dysfunction, neuroinflammation, and oxidative stress (Singh et al. 2014). Bent et al. (2018) indicated an association between the changes in urine metabolites and improvement of ASD symptoms after treatment with sulforaphane in children and young adults with ASD (Bent et al. 2018). They found increased antioxidant-related substances such as γ-glutamylglutamine and methionine sulfone in the urinary after sulforaphane ingestion (Bent et al. 2018). They also found a strong correlation between the increased urinary levels of sphingomyelin and the improvement of ASD behaviors after treatment with sulforaphane. This was consistent with a previous study that demonstrated decreased levels of sphingomyelin in urine samples of children with ASD compared to age-matched controls (Kelly et al. 2019; Wang et al. 2016). Hence, restoration of the sphingomyelin metabolism may be a potential intervention for the improvement of ASD symptoms. Apart from its capacities of anti-inflammation and antioxidant, sulforaphane repairs immune dysfunction through activation of nuclear factor erythroid 2-related factor 2 (Nrf2). This mechanism mitigates ASD-like symptoms in the animal study (Nadeem et al. 2019).

B-GOS

Gut dysbiosis is one of the most likely causes of ASD pathogenesis. The possible link between alteration in gut microbiota and ASD symptom improvement has been investigated (Fattorusso et al. 2019; Kang et al. 2019; Li et al. 2017). Grimaldi et al. (2018) studied the metabolomic alterations in feces and urine between the groups receiving the prebiotic B-GOS and the controls taking a placebo of children with ASD either with exclusion or unrestricted diet (Grimaldi et al. 2018). They showed various changes in metabolites, including amino acids, fatty acids, and organic acids in the case of the ASD cohort with the prebiotic compared to those with placebo (Grimaldi et al. 2018). Interestingly, after B-GOS ingestion, the team observed the increased levels of butyrate in feces. Butyrate can be produced by bacteria in the Lachnospiraceae family, which is found in increased abundance after modulation of the composition of the gut microflora following B-GOS administration. In addition, decreased levels of some amino acids in children with ASD after treatment with B-GOS compared to those with placebo treatment were thought to be an effect of such prebiotic causing the increase in the absorption of amino acids through the intestine. (Grimaldi et al. 2018).

Suramin

An antipurinergic agent was shown to improve social behaviors in MIA and Fmr1 knockout mouse models by normalizing metabolites mainly in purine metabolism as presented in the previous animal studies (Naviaux et al. 2014, 2015). An explanation of the mechanism was discussed in the previous animal study section. For a human study, Naviaux et al. (2017) conducted a trial of single low dose suramin to children with ASD (Naviaux et al. 2017). They found the improvement of ASD symptoms associated with the alteration of several metabolic pathways. Notably, more than half of the pathways changed by suramin in children with ASD were also changed in the MIA mouse model. Therefore, suramin might be a promising agent for the treatment of ASD.

Due to limited trials on humans, we cannot conclude the strong evidence of the links between improved clinical symptoms of ASD and associated metabolic pathways. However, the potential intervention, such as sulforaphane, and B-GOS, may be beneficial for social behaviors in children with ASD by altering the perturbed ASD-related metabolites.

Conclusion and clinical implications

Previous metabolomic studies in urine and blood samples revealed that there were abnormalities of metabolism in children with ASD. These include the TCA cycle; urea cycle; oxidative stress; amino acids such as methionine, tryptophan, tyrosine, and BCAA; imbalance of neurotransmitters; mammalian-microbial co-metabolism; metabolic pathway of nucleotides, nicotinate and nicotinamide, lipids and fatty acids, carbohydrates, endocannabinoid, NAD; pentose phosphate pathway; protein glycation and oxidation, and oxidative phosphorylation. In animal studies, the pharmacological intervention that could improve ASD features by altering the metabolomic profiles was suramin. In human studies, the potential interventions that improved social behaviors in children with ASD include sulforaphane, and prebiotic B-GOS. The majority of studies that reported changes in metabolomic profiles after the treatment also demonstrated a favorable outcome of ASD symptoms. Several previous studies showed there are another promising pharmacological supplements for improving symptoms of ASD such as omega-3 fatty acid, folinic acid, and vitamin D. However, there is still a lack of evaluation in the metabolomics approach.

The most repeatedly proposed mechanisms and possible underlying pathophysiology of ASD were alterations of amino acids, reactive oxidative stress, neurotransmitters, and microbiota-gut-brain axis. For the interventional studies, it was less likely to explicitly present the trend of the most related metabolic pathways in relation to ASD, since only a few studies with various types of pharmacological interventions have been reported. However, our integrated metabolome data from both animal and human interventional studies reveal, to some extent, the shared potential pathways associated with the ASD phenotypic improvement. Those potential pathways were mammalian-microbial co-metabolite, purine metabolism, and fatty acid oxidation. Interestingly, all three interventional studies in humans also showed an increase in sphingomyelin, phospholipid-, and cholesterol-related metabolites, which were related to the improvement of ASD symptoms. This growing evidence of lipid alterations suggests that lipidomics might be another interesting field of investigation for a comprehensive explanation of ASD development in the future (El-Ansary et al. 2020).

Currently, the profiles of metabolic changes in previous studies have been inconsistent. Most notably, the ethnicity of the participants, which causes differences in both genetic and environmental exposure, may be the key factor in explaining unpredictable results. Much of the variation could be due to the differing populations, the types of samples, sample collection and sample preparation techniques, and data analysis techniques. Moreover, the varying degrees of symptom severity, the difference of co-morbidities, and various types of medications could influence the variety of metabolomic profiles among patients. The subgroup analysis investigating the association of those variations and metabolic profiles should be more researched. Prior studies opened the question of whether the heterogeneity, for example – symptom severity and age, could affect the change in metabolomic profiles among individuals (Mussap et al. 2020; Olesova et al. 2020). Mussap et al. (2020) showed the correlation between phenotypic severity of autism and urine metabolomes. Indeed, they found that the levels of some metabolites derived from diet, gut dysbiosis, tryptophan metabolism, and mitochondrial dysfunction of autistic children with severe core symptoms were distinguishable from those with less severe core symptoms (Mussap et al. 2020). In addition, Olesova et al. (2020) indicated that there was a significant difference in metabolomic profiles between ASD and non-ASD groups only in the school-aged children but not in the preschoolers (Olesova et al. 2020).

For further studies, confounding factors such as dietary intake, medication use, and illness in both children with ASD and children without ASD should be controlled. The severity of ASD, comorbid diseases, ethnicity, cognitive level, and environmental factors should also be collected for subgroup analysis to explain the pathogenesis of ASD more extensively. More metabolomic studies should focus on the pharmacological treatments and correlation of metabolomic profiles and ASD phenotypic changes after treatments. Importantly, there is a need for evaluation of the model sensitivity and specificity and cost-effectiveness for the practical use.

Funding

This work was supported by the Senior Research Scholar grant from the National Research Council of Thailand (S.C.C.), the Thailand Science Research and Innovation MRG6280014 (C.T.), the NSTDA Research Chair grant from the National Science and Technology Development Agency Thailand (N.C.), and the Chiang Mai University Center of Excellence Award Thailand (N.C.). National Institute of General Medical Sciences of the National Institutes of Health (P20GM125503) award to I.N.

Footnotes

Conflict of interest The authors have no conflicts of interest to disclose.

References

  1. Amminger GP, Berger GE, Schäfer MR et al. (2007) Omega-3 fatty acids supplementation in children with autism: a double-blind randomized, placebo-controlled pilot study. Biol Psychiatry 61:551–553 [DOI] [PubMed] [Google Scholar]
  2. Anwar A, Abruzzo PM, Pasha S et al. (2018) Advanced glycation endproducts, dityrosine and arginine transporter dysfunction in autism: a source of biomarkers for clinical diagnosis. Mol Autism 9:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Bae YS, Choi MK, Lee WJ (2010) Dual oxidase in mucosal immunity and host-microbe homeostasis. Trends Immunol 31:278–287 [DOI] [PubMed] [Google Scholar]
  4. Behnia F, Parets SE, Kechichian T et al. (2015) Fetal DNA methylation of autism spectrum disorders candidate genes: association with spontaneous preterm birth. Am J Obstet Gynecol 212:533. e531–533.e539 [DOI] [PubMed] [Google Scholar]
  5. Beloborodova N, Bairamov I, Olenin A et al. (2012) Effect of phenolic acids of microbial origin on production of reactive oxygen species in mitochondria and neutrophils. J Biomed Sci 19:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bent S, Lawton B, Warren T et al. (2018) Identification of urinary metabolites that correlate with clinical improvements in children with autism treated with sulforaphane from broccoli. Mol Autism 9:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bitar T, Mavel S, Emond P et al. (2018) Identification of metabolic pathway disturbances using multimodal metabolomics in autistic disorders in a middle eastern population. J Pharm Biomed Anal 152:57–65 [DOI] [PubMed] [Google Scholar]
  8. Cai J, Ding L, Zhang J-S et al. (2016) Elevated plasma levels of glutamate in children with autism spectrum disorders. NeuroReport 27:272. [DOI] [PubMed] [Google Scholar]
  9. Chang S, Linderholm A, Franzi L et al. (2013) Dual oxidase regulates neutrophil recruitment in allergic airways. Free Radic Biol Med 65:38–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Chaste P, Leboyer M (2012) Autism risk factors: genes, environment, and gene-environment interactions. Dialogues Clin Neurosci 14:281–292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Chauhan A, Chauhan V (2006) Oxidative stress in autism. Pathophysiology 13:171–181 [DOI] [PubMed] [Google Scholar]
  12. Chen Q, Qiao Y, Xu XJ et al. (2019) Urine organic acids as potential biomarkers for autism-spectrum disorder in Chinese children. Front Cell Neurosci 13:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Cogram P, Alkon DL, Crockford D et al. (2020) Chronic bryostatin-1 rescues autistic and cognitive phenotypes in the fragile x mice. Sci Rep 10:18058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Cohen BI (2002) The significance of ammonia/gamma-aminobutyric acid (gaba) ratio for normality and liver disorders. Med Hypotheses 59:757–758 [DOI] [PubMed] [Google Scholar]
  15. Cozzolino R, De Magistris L, Saggese P et al. (2014) Use of solid-phase microextraction coupled to gas chromatography–mass spectrometry for determination of urinary volatile organic compounds in autistic children compared with healthy controls. Anal Bioanal Chem 406:4649–4662 [DOI] [PubMed] [Google Scholar]
  16. da Silva Montenegro EM, Costa CS, Campos G et al. (2020) Meta-analyses support previous and novel autism candidate genes: outcomes of an unexplored Brazilian cohort. Autism Res 13:199–206 [DOI] [PubMed] [Google Scholar]
  17. Damodaran LPM, Arumugam G (2011) Urinary oxidative stress markers in children with autism. Redox Rep 16:216–222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Delaye J-B, Patin F, Lagrue E et al. (2018) Post hoc analysis of plasma amino acid profiles: towards a specific pattern in autism spectrum disorder and intellectual disability. Ann Clin Biochem 55:543–552 [DOI] [PubMed] [Google Scholar]
  19. Delwing D, Delwing D, Bavaresco CS et al. (2008) Protective effect of nitric oxide synthase inhibition or antioxidants on brain oxidative damage caused by intracerebroventricular arginine administration. Brain Res 1193:120–127 [DOI] [PubMed] [Google Scholar]
  20. Diémé B, Mavel S, Blasco H et al. (2015) Metabolomics study of urine in autism spectrum disorders using a multiplatform analytical methodology. J Proteome Res 14:5273–5282 [DOI] [PubMed] [Google Scholar]
  21. Dietert RR, Dietert JM, Dewitt JC (2011) Environmental risk factors for autism. Emerg Health Threats J 4:7111–7111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. El-Ansary A, Chirumbolo S, Bhat RS et al. (2020) The role of lipidomics in autism spectrum disorder. Mol Diagn Ther 24:31–48 [DOI] [PubMed] [Google Scholar]
  23. Emberti Gialloreti L, Curatolo P (2018) Autism spectrum disorder: why do we know so little? Front Neurol 9:670–670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Emond P, Mavel S, Aïdoud N et al. (2013) Gc-ms-based urine metabolic profiling of autism spectrum disorders. Anal Bioanal Chem 405:5291–5300 [DOI] [PubMed] [Google Scholar]
  25. Espinós C, Pineda M, Martinez Rubio MD et al. (2009) Mutations in the urocanase gene uroc1 are associated with urocanic aciduria. J Med Genet 46:407–411 [DOI] [PubMed] [Google Scholar]
  26. Fattorusso A, Di Genova L, Dell’Isola GB et al. (2019) Autism spectrum disorders and the gut microbiota. Nutrients 11:521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Frye RE (2015) Metabolic and mitochondrial disorders associated with epilepsy in children with autism spectrum disorder. Epilepsy Behav 47:147–157 [DOI] [PubMed] [Google Scholar]
  28. Gevi F, Belardo A, Zolla L (2020) A metabolomics approach to investigate urine levels of neurotransmitters and related metabolites in autistic children. Biochim Biophys Acta Mol Basis Dis 1866:165859. [DOI] [PubMed] [Google Scholar]
  29. Gevi F, Zolla L, Gabriele S et al. (2016) Urinary metabolomics of young Italian autistic children supports abnormal tryptophan and purine metabolism. Mol Autism 7:47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Ghezzo A, Visconti P, Abruzzo PM et al. (2013) Oxidative stress and erythrocyte membrane alterations in children with autism: correlation with clinical features. PLoS ONE 8:e66418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Glinton KE, Elsea SH (2019) Untargeted metabolomics for autism spectrum disorders: current status and future directions. Front Psych 10:647–647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Görker I, Tüzün U (2005) Autistic-like findings associated with a urea cycle disorder in a 4-year-old girl. J Psychiatry Neurosci 30:133–135 [PMC free article] [PubMed] [Google Scholar]
  33. Grimaldi R, Gibson GR, Vulevic J et al. (2018) A prebiotic intervention study in children with autism spectrum disorders (asds). Microbiome 6:133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Hannon E, Schendel D, Ladd-Acosta C et al. (2018) Elevated polygenic burden for autism is associated with differential DNA methylation at birth. Genome Med 10:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Hansen SN, Schendel DE, Parner ET (2015) Explaining the increase in the prevalence of autism spectrum disorders: the proportion attributable to changes in reporting practices. JAMA Pediatr 169:56–62 [DOI] [PubMed] [Google Scholar]
  36. Hassan T, Abdelrahman H, Fattah N et al. (2013) Blood and brain glutamate levels in children with autistic disorder. Res Autism Spectr Disord 7:541–548 [Google Scholar]
  37. Horder J, Petrinovic MM, Mendez MA et al. (2018) Glutamate and gaba in autism spectrum disorder—a translational magnetic resonance spectroscopy study in man and rodent models. Transl Psychiatry 8:106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Huerta M, Lord C (2012) Diagnostic evaluation of autism spectrum disorders. Pediatr Clin N Am 59:103–xi [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Kałużna-Czaplińska J (2011) Noninvasive urinary organic acids test to assess biochemical and nutritional individuality in autistic children. Clin Biochem 44:686–691 [DOI] [PubMed] [Google Scholar]
  40. Kang D-W, Adams JB, Coleman DM et al. (2019) Long-term benefit of microbiota transfer therapy on autism symptoms and gut microbiota. Sci Rep 9:5821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Kelly RS, Boulin A, Laranjo N et al. (2019) Metabolomics and communication skills development in children; evidence from the ages and stages questionnaire. Metabolites 9:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kuwabara H, Yamasue H, Koike S et al. (2013) Altered metabolites in the plasma of autism spectrum disorder: a capillary electrophoresis time-of-flight mass spectroscopy study. PLoS ONE 8:e73814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Ladd-Acosta C, Hansen KD, Briem E et al. (2014) Common DNA methylation alterations in multiple brain regions in autism. Mol Psychiatry 19:862–871 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Li Q, Han Y, Dy ABC et al. (2017) The gut microbiota and autism spectrum disorders. Front Cell Neurosci 11:120–120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Liang Y, Ke X, Xiao Z et al. (2020a) Untargeted metabolomic profiling using UHPLC-QTOF/MS reveals metabolic alterations associated with autism. BioMed Res Int 2020:6105608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Liang Y, Xiao Z, Ke X et al. (2020b) Urinary metabonomic profiling discriminates between children with autism and their healthy siblings. Med Sci Monit 26:e926634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Liu A, Zhou W, Qu L et al. (2019) Altered urinary amino acids in children with autism spectrum disorders. Front Cell Neurosci 13:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Lussu M, Noto A, Masili A et al. (2017) The urinary (1) h-nmr metabolomics profile of an Italian autistic children population and their unaffected siblings. Autism Res 10:1058–1066 [DOI] [PubMed] [Google Scholar]
  49. Lv QQ, You C, Zou XB et al. (2018) Acyl-carnitine, c5dc, and c26 as potential biomarkers for diagnosis of autism spectrum disorder in children. Psychiatry Res 267:277–280 [DOI] [PubMed] [Google Scholar]
  50. Mavel S, Nadal-Desbarats L, Blasco H et al. (2013) 1h–13c nmr-based urine metabolic profiling in autism spectrum disorders. Talanta 114:95–102 [DOI] [PubMed] [Google Scholar]
  51. Mazahery H, Stonehouse W, Delshad M et al. (2017) Relationship between long chain n-3 polyunsaturated fatty acids and autism spectrum disorder: systematic review and meta-analysis of case-control and randomised controlled trials. Nutrients 9:155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. McGuinness G, Kim Y (2020) Sulforaphane treatment for autism spectrum disorder: a systematic review. EXCLI J 19:892–903 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Meguid NA, Dardir AA, Abdel-Raouf ER et al. (2011) Evaluation of oxidative stress in autism: defective antioxidant enzymes and increased lipid peroxidation. Biol Trace Elem Res 143:58–65 [DOI] [PubMed] [Google Scholar]
  54. Melke J, Goubran Botros H, Chaste P et al. (2008) Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry 13:90–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Ming X, Stein TP, Barnes V et al. (2012) Metabolic perturbance in autism spectrum disorders: a metabolomics study. J Proteome Res 11:5856–5862 [DOI] [PubMed] [Google Scholar]
  56. Ming X, Stein TP, Brimacombe M et al. (2005) Increased excretion of a lipid peroxidation biomarker in autism. Prostaglandins Leukot Essent Fatty Acids 73:379–384 [DOI] [PubMed] [Google Scholar]
  57. Mitsubuchi H, Nakamura K, Matsumoto S et al. (2008) Inborn errors of proline metabolism. J Nutr 138:2016S–2020S [DOI] [PubMed] [Google Scholar]
  58. Monné M, Vozza A, Lasorsa FM et al. (2019) Mitochondrial carriers for aspartate, glutamate and other amino acids: a review. Int J Mol Sci 20:4456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Mussap M, Noto A, Fanos V (2016) Metabolomics of autism spectrum disorders: early insights regarding mammalian-microbial cometabolites. Expert Rev Mol Diagn 16:869–881 [DOI] [PubMed] [Google Scholar]
  60. Mussap M, Siracusano M, Noto A et al. (2020) The urine metabolome of young autistic children correlates with their clinical profile severity. Metabolites 10:476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Nadal-Desbarats L, Aïdoud N, Emond P et al. (2014) Combined 1h-nmr and 1h–13c hsqc-nmr to improve urinary screening in autism spectrum disorders. Analyst 139:3460–3468 [DOI] [PubMed] [Google Scholar]
  62. Nadeem A, Fayaz Ahmad S, Al-Harbi N et al. (2019) Nrf2 activator, sulforaphane ameliorates autism-like symptoms through suppression of th17 related signaling and rectification of oxidant-antioxidant imbalance in periphery and brain of btbr t+tf/j mice. Behav Brain Res 364:213–224 [DOI] [PubMed] [Google Scholar]
  63. Naushad SM, Jain JM, Prasad CK et al. (2013) Autistic children exhibit distinct plasma amino acid profile. Indian J Biochem Biophys 50:474–478 [PubMed] [Google Scholar]
  64. Naviaux JC, Schuchbauer MA, Li K et al. (2014) Reversal of autism-like behaviors and metabolism in adult mice with single-dose antipurinergic therapy. Transl Psychiatry 4:e400–e400 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Naviaux JC, Wang L, Li K et al. (2015) Antipurinergic therapy corrects the autism-like features in the fragile x (fmr1 knockout) mouse model. Mol Autism 6:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Naviaux RK (2014) Metabolic features of the cell danger response. Mitochondrion 16:7–17 [DOI] [PubMed] [Google Scholar]
  67. Naviaux RK, Curtis B, Li K et al. (2017) Low-dose suramin in autism spectrum disordera: a small, phase i/ii, randomized clinical trial. Ann Clin Transl Neurol 4:491–505 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Niu X, Zheng S, Liu H et al. (2018) Protective effects of taurine against inflammation, apoptosis, and oxidative stress in brain injury. Mol Med Rep 18:4516–4522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Noto A, Fanos V, Barberini L et al. (2014) The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J Maternal Fetal Neonatal Med 27(Suppl 2):46–52 [DOI] [PubMed] [Google Scholar]
  70. Novarino G, El-Fishawy P, Kayserili H et al. (2012) Mutations in bckdkinase lead to a potentially treatable form of autism with epilepsy. Science 338:394–397 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Olesova D, Galba J, Piestansky J et al. (2020) A novel UHPLC-MS method targeting urinary metabolomic markers for autism spectrum disorder. Metabolites 10:443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Orozco JS, Hertz-Picciotto I, Abbeduto L et al. (2019) Metabolomics analysis of children with autism, idiopathic-developmental delays, and down syndrome. Transl Psychiatry 9:243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Palmieri L, Papaleo V, Porcelli V et al. (2010) Altered calcium homeostasis in autism-spectrum disorders: evidence from biochemical and genetic studies of the mitochondrial aspartate/glutamate carrier agc1. Mol Psychiatry 15:38–52 [DOI] [PubMed] [Google Scholar]
  74. Parletta N, Niyonsenga T, Duff J (2016) Omega-3 and omega-6 polyunsaturated fatty acid levels and correlations with symptoms in children with attention deficit hyperactivity disorder, autistic spectrum disorder and typically developing controls. PLoS ONE 11:e0156432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Rangel-Huerta OD, Gomez-Fernández A, de la Torre-Aguilar MJ et al. (2019) Metabolic profiling in children with autism spectrum disorder with and without mental regression: preliminary results from a cross-sectional case–control study. Metabolomics 15:99. [DOI] [PubMed] [Google Scholar]
  76. Safiulina D, Peet N, Seppet E et al. (2006) Dehydroepiandrosterone inhibits complex i of the mitochondrial respiratory chain and is neurotoxic in vitro and in vivo at high concentrations. Toxicol Sci 93:348–356 [DOI] [PubMed] [Google Scholar]
  77. Singh K, Connors SL, Macklin EA et al. (2014) Sulforaphane treatment of autism spectrum disorder (ASD). Proc Natl Acad Sci USA 111:15550–15555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Smith AM, King JJ, West PR et al. (2019) Amino acid dysregulation metabotypes: potential biomarkers for diagnosis and individualized treatment for subtypes of autism spectrum disorder. Biol Psychiatry 85:345–354 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Tirouvanziam R, Obukhanych TV, Laval J et al. (2012) Distinct plasma profile of polar neutral amino acids, leucine, and glutamate in children with autism spectrum disorders. J Autism Dev Disord 42:827–836 [DOI] [PubMed] [Google Scholar]
  80. Toczylowska B, Zieminska E, Senator P et al. (2020) Hippocampal metabolite profiles in two rat models of autism: NMR-based metabolomics studies. Mol Neurobiol 57:3089–3105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Wang H, Liang S, Wang M et al. (2016) Potential serum biomarkers from a metabolomics study of autism. J Psychiatry Neurosci 41:27–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Wang L, Christophersen CT, Sorich MJ et al. (2012) Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig Dis Sci 57:2096–2102 [DOI] [PubMed] [Google Scholar]
  83. West PR, Amaral DG, Bais P et al. (2014) Metabolomics as a tool for discovery of biomarkers of autism spectrum disorder in the blood plasma of children. PLoS ONE 9:e112445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Willets JM, Lunec J, Williams AC et al. (1993) Neurotoxicity of nicotinamide derivatives. Biochem Soc Trans 21:299S–299S [DOI] [PubMed] [Google Scholar]
  85. Williams AC, Pall HS, Steventon GB et al. (1993) N-methylation of pyridines and Parkinson’s disease. Adv Neurol 60:194–196 [PubMed] [Google Scholar]
  86. Wu Z, Huang S, Zou J et al. (2020) Autism spectrum disorder (ASD): disturbance of the melatonin system and its implications. Biomed Pharmacother 130:110496. [DOI] [PubMed] [Google Scholar]
  87. Xiong X, Liu D, He W et al. (2019) Identification of gender-related metabolic disturbances in autism spectrum disorders using urinary metabolomics. Int J Biochem Cell Biol 115:105594. [DOI] [PubMed] [Google Scholar]
  88. Xiong X, Liu D, Wang Y et al. (2016) Urinary 3-(3-hydroxyphenyl)-3-hydroxypropionic acid, 3-hydroxyphenylacetic acid, and 3-hydroxyhippuric acid are elevated in children with autism spectrum disorders. Biomed Res Int 2016:9485412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Yap IKS, Angley M, Veselkov KA et al. (2010) Urinary metabolic phenotyping differentiates children with autism from their unaffected siblings and age-matched controls. J Proteome Res 9:2996–3004 [DOI] [PubMed] [Google Scholar]
  90. Yorbik O, Sayal A, Akay C et al. (2002) Investigation of antioxidant enzymes in children with autistic disorder. Prostaglandins Leukot Essent Fatty Acids 67:341–343 [DOI] [PubMed] [Google Scholar]
  91. Yui K, Koshiba M, Nakamura S et al. (2012) Effects of large doses of arachidonic acid added to docosahexaenoic acid on social impairment in individuals with autism spectrum disorders: a double-blind, placebo-controlled, randomized trial. J Clin Psychopharmacol 32:200–206 [DOI] [PubMed] [Google Scholar]
  92. Yui K, Tanuma N, Yamada H et al. (2017) Decreased total antioxidant capacity has a larger effect size than increased oxidant levels in urine in individuals with autism spectrum disorder. Environ Sci Pollut Res 24:9635–9644 [DOI] [PubMed] [Google Scholar]

RESOURCES