Elsevier

Anaerobe

Volume 49, February 2018, Pages 121-131
Anaerobe

Anaerobes in the microbiome
Differences in fecal microbial metabolites and microbiota of children with autism spectrum disorders

https://doi.org/10.1016/j.anaerobe.2017.12.007Get rights and content

Highlights

  • Isopropanol was higher in feces of children with ASD than neurotypical children.

  • Feces of children with ASD have different abundance of possible neurotransmitters.

  • Higher fecal p-cresol and possibly lower GABA concentrations for children with ASD.

  • Fisher Discriminant Analysis suggests a group of 5 fecal metabolites as a biomarker.

  • We confirmed lower microbial diversity and Prevotella in feces of children with ASD.

Abstract

Evidence supporting that gut problems are linked to ASD symptoms has been accumulating both in humans and animal models of ASD. Gut microbes and their metabolites may be linked not only to GI problems but also to ASD behavior symptoms. Despite this high interest, most previous studies have looked mainly at microbial structure, and studies on fecal metabolites are rare in the context of ASD. Thus, we aimed to detect fecal metabolites that may be present at significantly different concentrations between 21 children with ASD and 23 neurotypical children and to investigate its possible link to human gut microbiome. Using 1H-NMR spectroscopy and 16S rRNA gene amplicon sequencing, we examined metabolite profiles and microbial compositions in fecal samples, respectively. Of the 59 metabolites detected, isopropanol concentrations were significantly higher in feces of children with ASD after multiple testing corrections. We also observed similar trends of fecal metabolites to previous studies; children with ASD have higher fecal p-cresol and possibly lower GABA concentrations. In addition, Fisher Discriminant Analysis (FDA) with leave-out-validation suggested that a group of metabolites-caprate, nicotinate, glutamine, thymine, and aspartate-may potentially function as a modest biomarker to separate ASD participants from the neurotypical group (78% sensitivity and 81% specificity). Consistent with our previous Arizona cohort study, we also confirmed lower gut microbial diversity and reduced relative abundances of phylotypes most closely related to Prevotella copri in children with ASD. After multiple testing corrections, we also learned that relative abundances of Feacalibacterium prausnitzii and Haemophilus parainfluenzae were lower in feces of children with ASD. Despite a relatively short list of fecal metabolites, the data in this study support that children with ASD have altered metabolite profiles in feces when compared with neurotypical children and warrant further investigation of metabolites in larger cohorts.

Introduction

The prevalence of Autism Spectrum Disorders (ASD) continue to increase in the world [1,2]. Genetic, environmental and biological factors play a critical role in neurodevelopment during a mother's pregnancy and immediately after birth [3,4]. The comorbidity of GI symptoms (primarily chronic constipation and/or diarrhea) in ASD is generally estimated to be 30–50%, and is poorly understood [5,6]. The significantly increased presence of GI symptoms in ASD has motivated researchers to explore gut microbial composition in children with ASD and hypothesize about their potential role in contributing to/reflecting ASD symptoms [[7], [8], [9], [10]].

Researchers have focused on whether children with ASD possess a lack of beneficial or an increase of harmful microbes in their gut. Potentially harmful Clostridium species were observed abundant in feces of children with ASD [[11], [12], [13]]. One type of beneficial bacteria, i.e., Bifidobacterium, was reduced in children with ASD [[14], [15], [16]], while another probiotic, i.e., Lactobacillus, was reported to be present in higher concentrations in children with ASD [9,16]. Using next-generation sequencing technology, microbes that might otherwise be ignored have been detected and proposed as either potentially beneficial bacteria (e.g., Prevotella) [7] or harmful bacteria (e.g., Sutterella) [17] in the context of autism. Observations on individual bacterial taxa, however, have not always been congruent between studies. For example, Desulfovibrio and Akkermansia levels in children with ASD were found to be either higher [10,14] or lower [7,15], although multiple testing corrections was not always performed [10]. In some cohorts, no difference in gut microbiota was reported between children with ASD and their neurotypical siblings [18,19], and some studies have found differences between individuals with ASD and their siblings [14], but neurotypical siblings may be different from the general neurotypical population. As previously reviewed [20], these discrepancies may be attributed to the broad spectrum of ASD manifestations, varying molecular technologies used to investigate subject samples, geographical differences between participants (which may result in genetic and/or dietary differences), potential sub-types of gut microbiota within ASD groups, small sample size and inadequate statistical control for testing multiple-hypotheses. Considering the redundant functions of microbes [21], research must expand beyond simply cataloging microbial composition and instead also investigate microbial functions and interrelated pathways, since important microbial functions may be masked when gut microbes are considered individually.

Studies conducted in humans [5,6,16] and animal models of ASD [[22], [23], [24], [25], [26]] suggest that gut microbes and their metabolites may be linked not only to GI problems but also to ASD behavior symptoms. Hsiao et al. [22] demonstrated that 4-ethylphenylsulfate (4EPS) and indolepyruvate concentrations in serum were strikingly increased in the maternal immune activation (MIA) mouse model that displays ASD symptoms and induced anxiety-like behaviors in offspring mice. Bacteroides fragilis modulated 4EPS concentrations most likely by restoring tight-junction integrity [22]. Likewise, stress-induced corticosterone levels were reduced when stressed mice were treated with Lactobacillus rhamnosus [25]. The treatment with beneficial bacteria, Lactobacillus reuteri, ameliorated deficient social behaviors in maternal high-fat diet offspring mice, and oxytocin levels were restored [24]. Oxytocin is a crucial hormone in social behavior and cognition [27], but its therapeutic effect on ASD needs more human clinical trials to draw definite conclusions [28,29].

Despite these compelling observations, studies that examine human gut microbial metabolites are rare in the context of ASD. Functional level analyses such as metagenomics, metatranscriptomics, and metabolomics should follow studies of microbial composition: metabolomics has the advantage that it can provide information about the final products of microbial functions. Metabolomics with fecal samples can provide clues on gut microbial metabolism. However, most metabolomics studies focus on urine and blood metabolites [[30], [31], [32], [33], [34], [35]]. Only a few studies have investigated fecal metabolites and attempted to correlate them with gut microbial structure in children with ASD. Wang et al. [36] observed elevated short-chain fatty acids (SCFA) and ammonia concentrations in feces of children with ASD, although De Angelis et al. [14] and Adams et al. [16] reported reduced total SCFA in children with ASD. De Angelis et al. [14] also observed altered levels of neurotransmitters-glutamate and GABA in fecal samples. Glutamate was highest in children with autism and GABA was lowest in children with Pervasive developmental disorder not otherwise specified (PDD-NOS). In addition, phenol substances including p-cresol were higher in feces of children with autism and PDD-NOS [14], but phenol and p-cresol levels were comparable between children with ASD and controls in the other cohort [36].

In this study, we received fecal samples from children with ASD and neurotypical controls, and obtained quantitative levels of 59 fecal metabolites and gut microbial profiles using 1H-NMR spectroscopy and pyrosequencing, respectively. We detected some fecal metabolites that may be present at significantly different concentrations between children with ASD and neurotypical children. We also performed correlation tests between fecal metabolite and individual bacterial phylotypes, and postulated their potential implications in the detection, etiology, and treatment of GI and ASD symptoms in children with ASD.

Section snippets

Ethics statement

The study was approved by the Institutional Review Board (IRB) at Arizona State University (ASU IRB Protocol #: 1206007979). We advertised the study by email to families with family members with ASD in Arizona, USA, and mailed the consent form to people who showed an interest in participating in the study. Once the signed consent forms were returned, we screened them for eligibility criteria and sent questionnaires and sample collection kits to participants.

Subject recruitment and sample collection

21 neurotypical children and 23

Subject characteristics

21 neurotypical children and 23 children with ASD participated in the study (Table 1). All children were between 4 and 17 years old, with a mean age (±SD) of 8.4 (±3.4) years for neurotypical children and 10.1 (±4.1) years for children with ASD (Table 1). We assessed the severity of GI symptoms using the 6 item Gastrointestinal Severity Index (6-GSI) [16], and GI symptoms were significantly more severe for children with ASD compared to controls (two-tailed Mann-Whitney U test, p < 0.005) (

Conclusion

In summary, we obtained fecal metabolite profiles in children with ASD and neurotypical children. Notably, isopropanol concentrations were significantly higher in feces of children with ASD after multiple testing corrections. Consistent with previous studies, we observed similar trends regarding higher p-cresol in feces of children with ASD, whereas concentrations of GABA were relatively lower in feces of children with ASD. We also confirmed lower gut microbial diversity in children with ASD in

Data deposition

The 16S rRNA gene sequence reads analyzed in this paper were deposited in the open-source microbiom database “Qiita” with the study ID number 11169 (https://qiita.microbio.me).

Conflicts of interest

JBA, D-WK, and RKB have pending/approved patents related to the use of fecal microbiota transplant and/or probiotics for various conditions including autism. JBA is a part-time consultant for Crestovo (a microbiome-related company).

Acknowledgements

We gratefully would like to thank all the children with ASD, neurotypical children, and their families for participating in the study. This study was financially supported by the BHARE (Brenen Hornstein Autism Research & Education) Foundation, the Emch Foundation, and Autism Research Institute. We thank Dr. Jay Park and the DNASU Genomics Core Facility at Arizona State University for bioinformatics. We also thank D. Hagner for help with study coordination. A portion of the research was

References (87)

  • L. Dou et al.

    The uremic solutes p-cresol and indoxyl sulfate inhibit endothelial proliferation and wound repair

    Kidney Int.

    (2004)
  • T. Shiba et al.

    Effects of intestinal bacteria-derived p-cresyl sulfate on Th1-type immune response in vivo and in vitro

    Toxicol. Appl. Pharmacol.

    (2014)
  • M. Andriamihaja et al.

    The deleterious metabolic and genotoxic effects of the bacterial metabolite p-cresol on colonic epithelial cells

    Free Radic. Biol. Med.

    (2015)
  • L.Y. Peng et al.

    Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers

    J. Nutr.

    (2009)
  • T.W. Stone et al.

    NMDA receptors and ligands in the vertebrate CNS

    Prog. Neurobiol.

    (1988)
  • E.J. Lee et al.

    NMDA receptor dysfunction in autism spectrum disorders

    Curr. Opin. Pharmacol.

    (2015)
  • D.Y. Kim et al.

    Serotonin: a mediator of the brain-gut connection

    Am. J. Gastroenterol.

    (2000)
  • J.S. Lim et al.

    Modeling environmental risk factors of autism in mice induces IBD-related gut microbial dysbiosis and hyperserotonemia

    Mol. Brain

    (2017)
  • M. Rajilic-Stojanovic et al.

    Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome

    Gastroenterology

    (2011)
  • F. Backhed et al.

    Dynamics and stabilization of the human gut microbiome during the first year of life

    Cell Host Microbe

    (2015)
  • D.M. Saulnier et al.

    Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome

    Gastroenterology

    (2011)
  • CDC

    Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2010

    (2014)
  • S. Kim et al.

    Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring

    Nature

    (2017)
  • T. Buie et al.

    Evaluation, diagnosis, and treatment of gastrointestinal disorders in Individuals with ASDs: a consensus report

    Pediatrics

    (2010)
  • B.O. McElhanon et al.

    Gastrointestinal symptoms in autism spectrum disorder: a meta-analysis

    Pediatrics

    (2014)
  • D.W. Kang et al.

    Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children

    PLoS One

    (2013)
  • B.L. Williams et al.

    Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances

    PLos One

    (2011)
  • S.M. Finegold et al.

    Pyrosequencing study of fecal microflora of autistic and control children

    Anaerobe

    (2010)
  • Y.L. Song et al.

    Real-time PCR quantitation of Clostridia in feces of autistic children

    Appl. Environ. Microbiol.

    (2004)
  • H. Parracho et al.

    Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children

    J. Med. Microbiol.

    (2005)
  • S.M. Finegold et al.

    Gastrointestinal microflora studies in late-onset autism

    Clin. Infect. Dis.

    (2002)
  • M. De Angelis et al.

    Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified

    PLos One

    (2013)
  • L. Wang et al.

    Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism

    Appl. Environ. Microbiol.

    (2011)
  • J.B. Adams et al.

    Gastrointestinal flora and gastrointestinal status in children with autism-comparisons to typical children and correlation with autism severity

    BMC Gastroenterol.

    (2011)
  • B.L. Williams et al.

    Application of novel PCR-based methods for detection, quantification, and phylogenetic characterization of Sutterella species in intestinal biopsy samples from children with autism and gastrointestinal disturbances

    mBio

    (2012)
  • J.S. Son et al.

    Comparison of fecal microbiota in children with autism spectrum disorders and neurotypical siblings in the Simons simplex sollection

    PLos One

    (2015)
  • S.V. Gondalia et al.

    Molecular characterisation of gastrointestinal microbiota of children with autism (with and without gastrointestinal dysfunction) and their neurotypical siblings

    Autism Res.

    (2012)
  • R. Krajmalnik-Brown et al.

    Gut bacteria in children with autism spectrum disorders: challenges and promise of studying how a complex community influences a complex disease

    Microb. Ecol. Health Dis.

    (2015)
  • C. Huttenhower et al.

    Structure, function and diversity of the healthy human microbiome

    Nature

    (2012)
  • J.A. Bravo et al.

    Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • K.A. Foley et al.

    Pre- and neonatal exposure to lipopolysaccharide or the enteric metabolite, propionic acid, alters development and behavior in adolescent rats in a sexually dimorphic manner

    PLos One

    (2014)
  • Z.R. Donaldson et al.

    Oxytocin, vasopressin, and the neurogenetics of sociality

    Science

    (2008)
  • L.J. Young et al.

    Can oxytocin treat autism?

    Science

    (2015)
  • Cited by (240)

    View all citing articles on Scopus
    View full text