Download as pdf or txt
Download as pdf or txt
You are on page 1of 59

Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.

com

Journal Pre-proof

Microbiota and gut-brain axis dysfunction in Autism Spectrum Disorder:


Evidence for Functional Gastrointestinal disorders

I. Lasheras, P. Seral, E. Latorre, E. Barroso, P. Gracia-Garcı́a, J.


Santabárbara

PII: S1876-2018(19)30747-6
DOI: https://doi.org/10.1016/j.ajp.2019.101874
Reference: AJP 101874

To appear in: Asian Journal of Psychiatry

Received Date: 12 August 2019


Revised Date: 4 November 2019
Accepted Date: 4 November 2019

Please cite this article as: Lasheras I, Seral P, Latorre E, Barroso E, Gracia-Garcı́a P,
Santabárbara J, Microbiota and gut-brain axis dysfunction in Autism Spectrum Disorder:
Evidence for Functional Gastrointestinal disorders, Asian Journal of Psychiatry (2019),
doi: https://doi.org/10.1016/j.ajp.2019.101874

This is a PDF file of an article that has undergone enhancements after acceptance, such as
the addition of a cover page and metadata, and formatting for readability, but it is not yet the
definitive version of record. This version will undergo additional copyediting, typesetting and
review before it is published in its final form, but we are providing this version to give early
visibility of the article. Please note that, during the production process, errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal
pertain.

© 2019 Published by Elsevier.


Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

Microbiota and gut-brain axis dysfunction in Autism Spectrum

Disorder: Evidence for Functional Gastrointestinal disorders

Lasheras I a, Seral P a, Latorre E b,c,d*


, Barroso E e, Gracia-García P d,f,g
,
Santabárbara J a,d,g

a
Department of Preventive Medicine and Public Health, Universidad de Zaragoza,
Zaragoza, Spain.
b
Department of Biochemistry and Molecular and Cell Biology, Universidad de Zaragoza,
Zaragoza, Spain

of
c
Instituto Agroalimentario de Aragón – IA2- (Universidad de Zaragoza – CITA),
Zaragoza, Spain

ro
d
Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
e
Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Madrid,
Spain
f
-p
Psychiatry Service. Hospital Clínico Universitario Miguel Servet, Zaragoza, Spain
re
g
Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Ministry of
Science and Innovation, Madrid, Spain.
lP

*Corresponding and reprint author:


Dr. Eva Latorre
na

Department of Biochemistry and Molecular and Cell Biology


University of Zaragoza
ur

Pedro Cerbuna 12, 50009, Zaragoza, Spain


evalatorre@unizar.es
Jo

1
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

Highlights
 Autism spectrum disorders (ASD) are associated with functional gastrointestinal
disorders
 Gut microbiota dysbiosis could be a potential factor for ASD pathogenesis
 Microbiome may be an interface between genetic and environmental risk factors
in ASD
 We review evidence of gut microbial shifts in ASD and gastrointestinal
disorders
 Gut microbiota-brain axis studies could identify novel targets for ASD

ABSTRACT

of
INTRODUCTION: The high frequency of functional gastrointestinal disorders (FGIDs)

in autism spectrum disorders (ASD) has drawn attention to the composition of gut

ro
microbiota as a possible factor in ASD pathogenesis. However, characterization of a

distinctive ASD microbial pattern is still unclear.


-p
OBJECTIVE: To conduct a narrative review on ASD microbial profile and diversity
re
changes relative to NT children and FGID comorbidity and ASD pathogenesis.
lP

METHODOLOGY: First, we searched the PubMed database in peer-reviewed journals

for evidence regarding the current epidemiological evidence on FGID comorbidity. For
na

the identification of a microbial profile in ASD children, only original studies examining

gut bacterial and fungal abundances and diversity in ASD children and adolescents were
ur

included. Lastly, research on the role of microbial dysbiosis as an interface between

genetic and environmental risk factors in the pathogenesis of neuropsychiatric disorders,


Jo

and specifically ASD, was examined.

RESULTS: Prevalence and risk of FGIDs is significantly higher in ASD children and

correlates with the severity of ASD. Bacterial and fungal diversity differ between ASD

and NT children, indicating a difference in taxonomic abundance profiles, which have

2
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

been reported at all bacterial phylogenetic levels. However, studies analyzing gut

microbiota have a heterogeneous methodology and several limitations that could account

for the variety of findings for each taxon. Also, covariate analysis reveals influence of

demographics, diet, disease severity, GI comorbidity and allergies. Integration of these

findings with changes in metabolome and genetic risk factors allowed for a better

understanding of microbiota involvement in ASD pathogenesis for future research.

Keywords: Autism Spectrum Disorder, Microbiota, Gut-brain Axis, Functional

of
Gastrointestinal Disorders, Dysbiosis

ro
1. INTRODUCTION
-p
re
Autism spectrum disorders (ASD) is an umbrella term that incorporates a set of

neurodevelopmental disorders previously referred to as autistic disorder (AD), Asperger´s


lP

syndrome, pervasive developmental disorder not otherwise specified (PDD-NOS), and

childhood disintegrative disorders (Gyawali and Patra 2019). The global prevalence of
na

ASD was estimated at 1 in 160 in 2000 (Elsabbagh et al. 2012) and rates have shown an

increasing trend over time. In 2016, ASD was found to affect 1 in every 37 US children
ur

(Xu et al. 2018), with a significantly higher prevalence in boys (Christensen et al. 2018).

Although this upward trend seems to have leveled off (Tsai 2014), this group of disorders
Jo

has become a major concern in the scientific community.

Along with the core diagnostic traits, a wide range of comorbid conditions varying in

severity and combination can be observed in people with ASD. Among the more

commonly reported ones are sleep problems (Elrod and Hood 2015), atypical feeding
3
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

patterns (Sharp et al. 2013), psychiatric disorders such as obsessive compulsive disorder,

bipolar disorder, psychotic spectrum disorders, anxiety and depression (Nahar et al.

2019), epilepsy and gastrointestinal (GI) disorders (McElhanon et al. 2014). Several

emerging studies have identified an imbalanced composition of the intestinal microbiota

in ASD individuals, often reported under the controversial term “dysbiosis”, providing a

plausible explanation for the more frequent occurrence of functional GI disorders

(FGIDs) in ASD (Coury et al. 2012; Ding, Taur, and Walkup 2016; Roman, Rueda-

ruzafa, and Cardona 2018). Three large millionaire research projects on the link between

of
gut microbiota to neurodevelopmental and psychiatric disorders have been launched since

2013. Recent studies support that changes in gut microbiota could affect the brain

ro
functions and development through the gut-brain axis (H. X. Wang and Wang 2016),
-p
which refers to the bidirectional interaction pathways between the central nervous system

(CNS) and the trillions of microorganisms that inhabit the gut. However, characterization
re
of a distinctive ASD microbial pattern and its possible role on ASD remains unclear
lP

(Hsiao 2014; Luna, Savidge, and Williams 2016; Yang, Tian, and Yang 2018).

In view of this emerging evidence in recent years, this narrative review seeks to synthesize
na

what is currently known about: 1) FGIDs in ASD, 2) evidence of a distinctive gut

microbial signature in ASD patients, and 3) the possible role of gut microbiota dysbiosis
ur

and gut-brain axis dysfunction in ASD pathogenesis.


Jo

1.1 Functional Gastrointestinal Disorders in Autism Spectrum Disorder

FGIDs comprise a heterogeneous group of chronic recurrent gastrointestinal symptoms

which could not be explained by any underlying anatomical or biochemical abnormalities

4
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

(Gorrindo et al. 2012). Several studies found that the prevalence of any GI disorder in

patients with ASD ranged from 23% to 70% (Chaidez, Hansen, and Hertz-picciotto 2014;

Coury et al. 2012). The wide variation for each symptom across studies has been

attributed to methodological limitations, such as retrospective study design and

inappropriate control groups, enrollment of clinically heterogeneous ASD children, bias

incase selection and reliance on parental reports of symptoms (Holingue et al. 2018). This

breadth of estimates was also apparent in a meta-analysis, where it was reported that ASD

children had a four-fold higher risk for general GI concerns, a three-fold risk for

of
constipation and diarrhea, and a two-fold risk for abdominal pain (McElhanon et al.

2014); in most cases, these complaints received a diagnosis of a FGID.

ro
In the absence of any identified objective biomarker, the diagnosis of GI disorders is
-p
usually foreshadowed by the emergence or exacerbation of certain behavioral problems

which could indicate a child’s attempt to communicate the discomfort (Maenner et al.
re
2012). In fact, it has been found that ASD patients who showed signs of GI comorbidity
lP

displayed increased sleep difficulties, abnormal mood and argumentative, oppositional,

defiant or destructive behavior, anxiety, sensory responsivity, rigid compulsive


na

behaviors, self-injury, aggression, lack of expressive language and social impairments

when compared to ASD patients without GI comorbidities (Nikolov et al. 2009).


ur

Moreover, in some studies, the presence and intensity of abdominal pain was directly

associated with the severity of ASD core symptoms (Ding, Taur, and Walkup 2016;
Jo

Vuong and Hsiao 2016), and the occurrence of constipation correlated with rigid-

compulsive behavior (Srikantha and Hasan Mohajeri 2019). Despite the fact that

behavioral disturbances are not efficient predictors of GI problems, as they are already

frequent in ASD children without GI complaints (Maenner et al. 2012), they could

5
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

indicate a more than subtle association between GI dysfunction in ASD and behavioral

output through the gut-brain axis (Hsiao 2014; Vuong and Hsiao 2016).

2. EVIDENCE OF AN ALTERED GUT MICROBIOTA IN AUTISM SPECTRUM

DISORDERS

2.1 Description of included studies

A summary of the characteristics of 33 original studies examining gut bacterial and fungal

of
profile in children and adolescents with ASD is shown in Table 1.

ro
-p
In most cases (69.7%), unrelated NT children were recruited for the control group (Adams

et al. 2011; Carissimi et al. 2019; Coretti et al. 2018; Finegold et al. 2002; Inoue et al.
re
2016; Iovene et al. 2017; D.-W. Kang et al. 2013; D. W. Kang et al. 2018; Kantarcioglu,
lP

Kiraz, and Aydin 2016; Kong et al. 2019; Kushak et al. 2017; Lee et al. 2017; Liu et al.

2019; Luna et al. 2017; Ma et al. 2019; Pärtty et al. 2015; Plaza-Díaz et al. 2019; Sandler

et al. 2000; Song et al. 2003; Song, Liu, and Finegold 2004; Strati et al. 2017; Williams
na

et al. 2011; Williams, Hornig, and Parekh 2012; Zhang et al. 2018). However, some

studies contrast their results with a group of NT siblings (9%) (De Angelis et al. 2013a;
ur

Gondalia et al. 2012; Son et al. 2015), NT family members (6%) (Kong et al. 2019;
Jo

Pulikkan et al. 2018), or both related and unrelated controls (15.2%) (Finegold et al. 2010;

Helena M R T Parracho et al. 2005; Tomova et al. 2015; Lv Wang et al. 2011, 2013). In

some cases, ASD sample was stratified by severity of ASD (Finegold et al. 2010;

Gondalia et al. 2012; D.-W. Kang et al. 2013; Ma et al. 2019; Strati et al. 2017; Tomova

et al. 2015). Likewise, two studies were confined to specific clinical subtypes, such as
6
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

mild (Lee et al. 2017), severe (Pulikkan et al. 2018) and late-onset (Finegold et al. 2002;

Sandler et al. 2000; Song et al. 2003a; Song, Liu, and Finegold 2004) ASD forms, subjects

with Aspergers and attention deficit hyperactivity disorder (ADHD) (Pärtty et al. 2015)

or ASD children with developmental delay (Carissimi et al. 2019). Similarly, two studies

stratified their sample by PDD-NOS and AD (De Angelis et al. 2013), and ASD with and

without mental regression (Plaza-Díaz et al. 2019) respectively.

Most studies suffered from methodological issues, since 81.8% had small sample size

(n<50 subjects on the ASD group), 15.2% had a modest amount of subjects enrolled

of
(Adams et al. 2011; Gondalia et al. 2012; H M R T Parracho et al. 2010; Son et al. 2015;

ro
Strati et al. 2017), and the one study with a larger sample size included subjects with

suspected ASD diagnosis (Kantarcioglu, Kiraz, and Aydin 2016). Moreover, all but the
-p
latter suffered from sex bias, as female representation in sample was either low (Adams
re
et al. 2011; Coretti et al. 2018; Finegold et al. 2010b; Gondalia et al. 2012; Iovene et al.

2017b; D.-W. Kang et al. 2013; D. W. Kang et al. 2018; Kong et al. 2019; Kushak et al.
lP

2017; Lee et al. 2017; Liu et al. 2019; Ma et al. 2019; H M R T Parracho et al. 2010;

Pulikkan et al. 2018; Son et al. 2015; Strati et al. 2017; Tomova et al. 2015; Lv Wang et
na

al. 2011, 2013; Zhang et al. 2018), none (Carissimi et al. 2019; Luna et al. 2017; Pärtty

et al. 2015b; Williams et al. 2011; Williams, Hornig, and Parekh 2012) or unspecified
ur

(De Angelis et al. 2013; Finegold et al. 2002; Inoue et al. 2016; Plaza-Díaz et al. 2019;

Sandler et al. 2000; Song et al. 2003; Song, Liu, and Finegold 2004). Regarding study
Jo

design, only 15.2% of studies were age and gender-matched (Liu et al. 2019; Ma et al.

2019; Plaza-Díaz et al. 2019; Song et al. 2003a; Song, Liu, and Finegold 2004), 12.1%

were age-matched only (Kushak et al. 2017; Lee et al. 2017; Pärtty et al. 2015; Pulikkan

et al. 2018) and 6% were family-matched (Pulikkan et al. 2018; Son et al. 2015).

7
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

However, it should be noted that subjects enrolled in ASD and control groups were often

of similar ages.

Verification of ASD diagnosis was often carried out in the selection process by clinical

observation or using several validated tests. One study included suspected ASD patients

(Kantarcioglu, Kiraz, and Aydin 2016), whereas other three performed no validating

assessment of a previous ASD diagnosis (Adams et al. 2011; Gondalia et al. 2012; Son et

al. 2015), directly studying severity of disease. Four earlier studies claimed to include

children diagnosed with ASD but did not report any diagnostic method (Finegold et al.

of
2002; Helena M R T Parracho et al. 2005; Song et al. 2003; Song, Liu, and Finegold

ro
2004).

-p
Microbiota changes and overall bacterial diversity were assessed by quantification of

targeted bacterial and/or fungal species from a single or several stool samples (84.8%),
re
urine (3%) and/or biopsies (15.2%), which were taken from the stomach and small bowel

(Finegold et al. 2002) cecum and ileum (Williams et al. 2011; Williams, Hornig, and
lP

Parekh 2012), duodenum (Kushak et al. 2017) and rectum (Luna et al. 2017). One study

analyzed stool fungal profile exclusively (Kantarcioglu, Kiraz, and Aydin 2016), four
na

examined both stool bacteria and fungi (Adams et al. 2011; Finegold et al. 2002; Iovene

et al. 2017; Strati et al. 2017) and the remaining 28 studied bacterial population only. The
ur

most common methods for bacterial identification were sequencing or pyrosequencing of

the 16s rRNA gene, a very sensitive technique to obtain reliable results (Hang et al. 2014;
Jo

Rosselli et al. 2016). By contrast, all but one study (Strati et al. 2017) investigating fungi

relied on cultural approaches for yeast detection, given that molecular biology methods

were not available until very recently (Iovene et al. 2017). It should also be noted that,

although statistical analyses were performed on nearly all studies, covariates adjustment

8
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

was only carried out in some of the latest ones, reaching more conservative significance

levels. Moreover, in some cases, some microbiota-modulating factors were not taken into

consideration in the exclusion criteria, such as previous and/or concomitant intake of

antibiotics (Iovene et al. 2017; Kushak et al. 2017; Lee et al. 2017; Pärtty et al. 2015; Lv

Wang et al. 2011, 2013), probiotics and prebiotics, also known as functional foods

(Finegold et al. 2002, 2010; Iovene et al. 2017; D.-W. Kang et al. 2013; Kantarcioglu,

Kiraz, and Aydin 2016; Kushak et al. 2017; Luna et al. 2017; Pulikkan et al. 2018; Sandler

et al. 2000; Tomova et al. 2015; Lv Wang et al. 2011; Williams et al. 2011; Williams,

of
Hornig, and Parekh 2012); presence of concomitant pathologies (Gondalia et al. 2012;

Kantarcioglu, Kiraz, and Aydin 2016; Kushak et al. 2017; Pärtty et al. 2015; Sandler et

ro
al. 2000; Lv Wang et al. 2011, 2013; Williams et al. 2011; Williams, Hornig, and Parekh
-p
2012) and dietary status (Adams et al. 2011; De Angelis et al. 2013; Finegold et al. 2002;

Gondalia et al. 2012; Inoue et al. 2016; Iovene et al. 2017; D.-W. Kang et al. 2013; D. W.
re
Kang et al. 2018; Kantarcioglu, Kiraz, and Aydin 2016; Kushak et al. 2017; Lee et al.
lP

2017; Luna et al. 2017; H M R T Parracho et al. 2010; Pärtty et al. 2015; Sandler et al.

2000; Son et al. 2015; Tomova et al. 2015; Lv Wang et al. 2011, 2013; Williams et al.

2011; Williams, Hornig, and Parekh 2012). Nevertheless, some of these studies analyzed
na

their association or effect on bacterial outcomes, as will be discussed later. In addition,

studies that did consider prior antibiotic intake displayed variable discontinuation periods,
ur

ranging from 2 weeks to 3 months, with the average amount of days being 32 days and 1
Jo

month being the most frequent duration (52%), followed by 3 months (30%). In some

studies, other medications were assessed, and subjects taking antipsychotics (Lee et al.

2017), dietary supplements (Helena M R T Parracho et al. 2005), steroids (Luna et al.

2017), anti-inflammatories and antioxidants (Pulikkan et al. 2018), sedatives and muscle

relaxants (Plaza-Díaz et al. 2019) were ruled out. However, antifungal-taking subjects
9
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

10

were only excluded in two studies (Finegold et al. 2010b; D.-W. Kang et al. 2013; D. W.

Kang et al. 2018; Liu et al. 2019), none of which was among those analyzing fungal

populations (Adams et al. 2011; Iovene et al. 2017b; Kantarcioglu, Kiraz, and Aydin

2016; Strati et al. 2017). Furthermore, some studies did not provide information on some

of these inclusion or exclusion criteria (Finegold et al. 2002, 2010; Inoue et al. 2016; D.

W. Kang et al. 2018; Helena M R T Parracho et al. 2005; Song et al. 2003; Song, Liu,

and Finegold 2004; Tomova et al. 2015).

Following microbial sequencing, assessment of bacterial diversity was performed in over

of
60% of the cases. Species richness and diversity, which make up alpha diversity, were

ro
estimated in 58% and 33% of studies respectively. For the former, bacterial count was

performed by assemblage into operational taxonomic unit (OTU) clusters, abundance-


-p
based coverage (ACE) estimator metric or Chao1 index, whereas, for the latter, microbial
re
evenness was measured by Shannon, phylogenic diversity (PD) or Fisher indexes. On the

other hand, beta diversity, measured by unweighted and weighted UniFrac distances,
lP

Jaccard distance and Bray–Curtis dissimilarity was carried out in 45% of studies and

allowed for identification of differences among microbial communities within groups,


na

which enabled analysis of the effect of covariates in microbial composition. Only two

studies analyzed fungal diversity (Kantarcioglu, Kiraz, and Aydin 2016; Strati et al.
ur

2017).

Lastly, additional assessment of GI function was carried out in 26 studies using parental
Jo

symptom reports and standardized questionnaires in all cases but five (Inoue et al. 2016;

Lee et al. 2017; Plaza-Díaz et al. 2019; Pulikkan et al. 2018; Zhang et al. 2018), whose

measuring methods were not provided, and one ground-breaking earlier study, which only

considered loose stool history (Sandler et al. 2000). Strict inclusion or exclusion of

10
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

11

children with GI disturbances or stratification by this variable prompted classification of

the studies in Table 1 for further analysis of the implication of these GI conditions in

ASD microbial profile.

2.2 Major findings of included studies

The results of studies focusing on bacterial diversity in ASD patients are inconsistent.

20% of them reported a significant reduction of species richness (Carissimi et al. 2019;

of
D.-W. Kang et al. 2013; D. W. Kang et al. 2018; Ma et al. 2019), one of which attributed

ro
this finding to a generally lower number of taxa in ASD, rather than the depth of

sequencing, since this reduction was not supported by the reads count within groups
-p
(Carissimi et al. 2019). Opposite changes were found in 10% of studies (De Angelis et al.

2013; Finegold et al. 2010). This, however, is not supported by the remaining 70% of
re
studies reporting no significant differences in species richness, a trend that seemed to be
lP

consistent regardless of the kinship shared between ASD and controls or GI function,

since 29% of them included an unrelated group of NT children without GI dysfunction


na

(Coretti et al. 2018; Liu et al. 2019; Plaza-Díaz et al. 2019; Zhang et al. 2018) 21%

established comparisons with unrelated NT-GI children (Kushak et al. 2017; Strati et al.
ur

2017; Williams et al. 2011), 29% of them had a control group of related individuals

(Gondalia et al. 2012; Kong et al. 2019; Pulikkan et al. 2018; Son et al. 2015) and 21%
Jo

analyzed both unrelated and sibling controls (Helena M R T Parracho et al. 2005; Tomova

et al. 2015; Lv Wang et al. 2013). Similar tendencies were found for species diversity,

with a decreased reported by 33% of studies (Carissimi et al. 2019; D.-W. Kang et al.

2013; D. W. Kang et al. 2018; Liu et al. 2019; Ma et al. 2019), an increase in only one

study (7%) (Coretti et al. 2018) and no overall differences in the remaining 60%
11
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

12

(Gondalia et al. 2012; Kong et al. 2019; Kushak et al. 2017; Plaza-Díaz et al. 2019;

Pulikkan et al. 2018; Son et al. 2015; Strati et al. 2017; Williams et al. 2011; Zhang et al.

2018). It should be noted that only one study with a control group of relatives found

significant changes in species richness (De Angelis et al. 2013), and none regarding

species diversity or beta diversity (Gondalia et al. 2012; Kong et al. 2019; Pulikkan et al.

2018; Son et al. 2015). This underpins the hypothesis that changes in siblings to ASD

children could be intermediate between NT and ASD ones (Finegold et al. 2010). As

microbiota is influenced by genetic and environmental factors, certain similarities in

of
microbiome between ASD and their NT siblings could be expected. By contrast, analysis

of the beta diversity between ASD and unrelated NT children unanimously revealed a

ro
separate clustering for healthy and autistic samples (Coretti et al. 2018; D. W. Kang et al.
-p
2018; Liu et al. 2019; Luna et al. 2017; Ma et al. 2019; Strati et al. 2017; Zhang et al.

2018). In one case, however, this distinction was only significant in the unweighed
re
UniFrac analysis (Ma et al. 2019), which exclusively considers the presence or absence
lP

of bacterial reads and does not account for their abundance.


na

Overall, all but one study (Kushak et al. 2017) agreed that bacterial diversity did not show

any significant differences regarding age (D.-W. Kang et al. 2013; Luna et al. 2017;
ur

Helena M R T Parracho et al. 2005; Pulikkan et al. 2018) and sex (D.-W. Kang et al.

2013; Luna et al. 2017; Helena M R T Parracho et al. 2005). This could be ascribed to the
Jo

fact that samples in the discrepant study are collected from adolescents aged 12.7 to 17.3

years, who could be in pubertal age, and sexual maturation and hormones are considered

as major determinants for gender differences. Likewise, no effect was caused by antibiotic

history, intake of probiotics and other supplements (Helena M R T Parracho et al. 2005),

12
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

13

body mass index (Pulikkan et al. 2018), diet pattern (D.-W. Kang et al. 2013; Helena M

R T Parracho et al. 2005) or the severity of the autistic phenotype (Gondalia et al. 2012;

Strati et al. 2017). Moreover, half of the studies analyzing the effect of GI dysfunction

concluded that bacterial diversity variations could not be attributed to the latter. However,

in some cases, richness was found to negatively correlate with GI severity (D.-W. Kang

et al. 2013) and stratification by constipation was found to increase alpha diversity within

ASD subjects (Kong et al. 2019), as well as to separate clustering of samples within NT

subjects (Strati et al. 2017) and between the ASD-C, ASD-NC and NT-NC groups (Liu

of
et al. 2019) in regard to beta diversity.

ro
Changes at the phylum level are shown in Figure 1 and Supplementary Table S1. The

gut microbiota is mainly defined by two bacterial phylotypes: Bacteroidetes and


-p
Firmicutes, whose ratio was only reported to vary significantly in five studies (Coretti et
re
al. 2018; Kong et al. 2019; Strati et al. 2017; Tomova et al. 2015; Williams et al. 2011),

which found opposite tendencies. When both phyla are examined separately,
lP

contradictory findings replicate in all cases but when comparing ASD individuals to their

relatives. Indeed, the only study including family members to ASD in the cohort which
na

found a significant phylum change relative to ASD, specifically an increase in Firmicutes,

claimed to have a heterogeneous control group regarding kinship (Pulikkan et al. 2018).
ur

Likewise, replication of findings was rare within the class, order, family, genera and

species levels, which are shown in Figure 2, Figure 3 and Table S1.
Jo

Covariate analysis for correlation between demographics and microbial profile

predominantly concluded that there were no associations between age (Coretti et al. 2018;

Finegold et al. 2002; D.-W. Kang et al. 2013; Helena M R T Parracho et al. 2005), gender

(Adams et al. 2011; Coretti et al. 2018; Finegold et al. 2002; D.-W. Kang et al. 2013;

13
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

14

Luna et al. 2017; Helena M R T Parracho et al. 2005) or ethnicity (D.-W. Kang et al.

2013) and any specific taxa. However, a prospective follow-up study of newborns for 13

years revealed that, while no single constant microbiota composition component or

change was detected, some significant differences were found within the ASD and NT

groups between the third and eighteenth month of life, namely, decreased levels of

Bifidobacterium and decreased Bacteroides and cumulative levels of Enterococcus and

Lactobacillus (Pärtty et al. 2015) at 6 and 18 months respectively. Since microbial

colonization in offspring achieves its stability between the 6th and 36th month of life

of
(Mangiola et al. 2016), these changes could partake in a chain of pathological events

leading to ASD development, which will be further discussed in the following section.

ro
Although these differences found by Pärtty et al 2015 were not upheld at older ages
-p
(Pärtty et al. 2015), changes in those genera have been reported by several studies in

young children and adolescents (see Figure 2). Similarly, after stratification by age, Luna
re
et al 2017 found decreased Parabacteroides distasonis and increased Alistipes putrednis

and Clostridium perfringens in the 13-18th year old group in contrast with higher
lP

Ruminococcus spp. counts in the <6 years old cohort (Luna et al. 2017).
na

The effect of diet on ASD has been gaining broad attention, since it is one of the key

modulators of gut microbiota and has been linked to the development of ASD in humans
ur

and mice (Buffington et al. 2017; Coury et al. 2012). In fact, elimination diets and intake

of dietary supplements such as probiotics, prebiotics and symbiotics has gained popularity
Jo

among ASD children. Early studies included in our analysis conclude that there is no

relationship between the absolute levels of any bacterial populations and diet type

(Finegold et al. 2002; Helena M R T Parracho et al. 2005). This finding has been

contradicted by a recent and thorough study using the mathematical model principal

14
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

15

component analysis (PCA), which found that dissimilarity between ASD and NT children

was driven by distinct dietary components and bacterial variables within each cohort.

Furthermore, relationships have been established between Bifidobacterium and a high

consumption of dairy products, Prevotella and a plant-rich diet and, specifically in ASD

children, fish consumption and Lactobacillus (Adams et al. 2011), as well as pastry,

processed cold meat and fish, and low vegetable consumption with Hespellia (Plaza-Díaz

et al. 2019). This essentially implies that diet variability could be a confounding factor

for microbiota assessment and might contribute to the heterogeneity of results across the

of
studies analyzed, since they were conducted in different geographical areas. For instance,

the genus Sutterella, which has been put forward as relevant in ASD dysbiosis (D.-W.

ro
Kang et al. 2013; Lv Wang et al. 2013; Williams et al. 2011), was absent from all subjects
-p
consuming a Mediterranean diet (Plaza-Díaz et al. 2019).
re
Half of the studies assessing the correlation between ASD severity and bacterial taxa did

not observe any association (Carissimi et al. 2019; D.-W. Kang et al. 2013; Strati et al.
lP

2017). On the contrary, Iovene et al 2017 found a significant positive correlation between

autism severity measured by Childhood Autism Rating Scale (CARS) with Clostridium
na

spp. and calprotectin value, which is a marker of gut inflammation (Iovene et al. 2017).

Likewise, Autism Diagnostic Observation Schedules (ADOS) scores positively


ur

correlated with Faecalibacterium prasusnitzii and Bacteroides uniformis in another study

(Coretti et al. 2018). Involvement of these two species in autism is, however, unclear,
Jo

since evidence on the direction of change in ASD relative to NT is inconsistent (De

Angelis et al. 2013; D. W. Kang et al. 2018). Interestingly, Tomova et al 2015 observed

fluctuating results depending on the tool used to measure the severity of symptoms. That

is, children with CARS>50 had a nearly significant higher Clostridia and Desulfovibrio

15
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

16

amount and lower Bacteroidetes/Firmicutes ratio, while Autism Diagnostic Interview

(ADI) scores tended to a positive correlation with the relative amount of Desulfovibrio,

emanating from the strong association between this genus and the ADI

restricted/repetitive behavior subscale score (Tomova et al. 2015). Conflictingly, other

studies revealed that levels of metabolically active Desulfovibrio genus and

Desulfovibrionaceae family were 11 and 9.7 times greater in mildly autistic children (Lee

et al. 2017), compared to an increase of 2.7 of the relative family levels in severely

affected ones (Pulikkan et al. 2018).

of
The onset of behavioral signs of autism is usually conceptualized as occurring in either

ro
an early onset pattern, in which children show abnormalities in social and communicative

development in the first years of life, or a regressive pattern (previously referred to as


-p
“late onset”), in which children develop typically for a certain period of time and then
re
lose previously acquired skills. Information about microbial changes between these two

phenotypes is scarce. In this review, we found three early studies to exclusively include
lP

a group of late-onset ASD (Finegold et al. 2002; Song et al. 2003a; Song, Liu, and

Finegold 2004), and a recent one which stratified its ASD sample into subgroups by
na

mental regression (AMR) and no regression (ANMR) phenotype (Plaza-Díaz et al. 2019).

Although no obvious similarities among them were found for the differentially present
ur

taxa in ASD relative to NT, the latter study showed a higher relative abundance of

Proteobacteria in AMR and augmented levels in Actinobacteria phylum and class in


Jo

ANMR. Likewise, isolated results suggested differences in microbiota between different

clinical subtypes included under the term ASD; more specifically, it was found that

changes in microbiota and bacterial diversity in PDD-NOS were intermediate between

those found in AD and those found in NT siblings (De Angelis et al. 2013).

16
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

17

For the purposes of this paper, we reviewed studies that stratified their ASD sample by

comorbidity with FGIDs. In the ASD-FGIDs group, an increase was observed in the

absolute numbers or Ruminococcus torques and the relative abundances of Bacteroides

fragilis (Lv Wang et al. 2011). Interestingly, the increase of R. torques did not reach

significance before stratification by FGID presence (Lv Wang et al. 2011) or in studies

with a mixed ASD cohort in terms of GI function (D. W. Kang et al. 2018; Lv Wang et

al. 2011). The rise of B. fragilis, however, was also significant in another study where

neither ASD nor NT children reported FGIDs (De Angelis et al. 2013). Another study

of
found the mean relative abundance of the Chloroplast genus pertaining the

Cyanobacteria phylum to be increased in ASD-FGIDs and to dominate first-order

ro
interactions between ASD and FGIDs, a finding that could have been confounded by chia
-p
seed consumption (Son et al. 2015). Additionally, the severity of GI symptoms inversely

correlated with Clostridia and Desulfovibrio amounts and the Bacteroidetes/Firmicutes


re
ratio in one study (Tomova et al. 2015), although this finding did not reach statistical
lP

significance and was not supported by a posterior study (Carissimi et al. 2019).

Furthermore, cecal and ileal relative levels of Clostridiales and cumulative levels of

Lachnospiraceae and Ruminococcaceae in the FGIDs subset were significantly higher in


na

children whose first episode of GI symptoms occurred before or at the same time (within

the same month) as the onset of autism rather than after, and this difference remained
ur

significant after adjustment by age of onset of GI symptoms (Williams et al. 2011).


Jo

When FGID symptoms are assessed individually, significant associations are found

between irritable bowel syndrome and aerophagia and increased Clostridium aldenense;

aerophagia and decreased Blautia luti, Bifidobacterium adolescentis, Eubacterium

ventriosum, Anoxystipes fissicatena, Coprococcus comes, Eubacterium ramulus, and

17
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

18

Phascolarctobacterium faecium; as well as abdominal migraine and decreases in

Akkermansia muciniphila, Coprococcus catus, Odoribacter splanchnicus, Clostridium

lactatifermentans and Ruminococcus lactaris. Notably, none of these organisms

contributed to the separation of the ASD and NT groups (Luna et al. 2017). A greater

amount of evidence has focused on functional constipation in ASD children, which has

been associated with higher relative abundances of Escherichia/Shigella and Clostridium

cluster XVIII (Strati et al. 2017), the genera Fusobacterium, Barnesiella, Coprobacter,

Olsenella and Allisonella, the family Actinomycetaceae and the order Fusobacteriales

of
(Liu et al. 2019), as well as decreases in Oscillospira plautii, Bacteroides eggerthii,

Bacteroides uniformis, Faecalibacterium prausnitzii and Clostridium clariflavum (Luna

ro
et al. 2017). Remarkably, non-constipated NT children harbored differentiating bacteria
-p
from ASD and NT constipated individuals, with higher abundances of Gemmiger (Strati

et al. 2017) and a preponderance of Eubacterium rectale group, Streptococcus,


re
Butyricicoccus, Eubacterium ventriosum group, Propionibacterium and
lP

Lachnospiraceae NC2004 group (Liu et al. 2019). Moreover, children suffering from

abdominal pain had higher levels of Turicibacter sanguinis, Clostridium lituseburense,

Clostridium disporicum, C. aldenense and O. plautii. The latter two, whose link to other
na

GI symptoms has already been reflected in this review, together with Tyzzerella sp. and

Parasutterella excrementihominis were further enriched in the ASD-FGID pain group


ur

compared to the ASD-FGID no pain group (Luna et al. 2017). On the contrary, ASD and
Jo

NT subjects without this symptom had higher levels of Roseburia and Bacteroides, the

latter emanating from an increase in ASD patients without abdominal pain, but not in NT

(Kong et al. 2019).

18
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

19

Interestingly, some of the positively correlated bacteria with GI symptoms have been

found at very high levels in some ASD children (i.e. Turicibacter sanguinis) (De Angelis

et al. 2013; D.-W. Kang et al. 2013), whereas they displayed lower levels of the negatively

correlated ones such as Bifidobacterium adolescentis (De Angelis et al. 2013),

Phascolarctobacterium faecium (Ma et al. 2019), Blautia luti (Luna et al. 2017),

Ruminococcus lactaris (non-significant) (Finegold et al. 2002) and Roseburia (D.-W.

Kang et al. 2013; D. W. Kang et al. 2018). For the latter, at a species level, an increase of

R. inulinovorans (De Angelis et al. 2013) and the decrease of R. faecis, R. hominis and R.

of
intestinalis (De Angelis et al. 2013; Luna et al. 2017) has been observed. In regard to the

pathogenesis of GI disorders, some studies have hypothesized a role of disaccharidase

ro
activity as a connection between microbial abundance, dysbiosis and malabsorption

(Kushak et al. 2017; Williams et al. 2011). -p


re
Finally, given the higher prevalence of allergies in the ASD population, especially food

relates ones, two studies evaluated the effect of this comorbidity on cecal and ileal
lP

(Williams et al. 2011) and stool (Kong et al. 2019) microbial profile. ASD subjects with

allergies showed increased relative abundance of stool Proteobacteria, a phylum


na

previously associated with autoimmune conditions (Kong et al. 2019). Also, food

allergies seemed to increase cecal Betaproteobacteria, as well as ileal and cecal


ur

Firmicutes and the Firmicutes/Bacteroidetes ratio (Williams et al. 2011). This effect was

stronger after stratification by milk related allergies, whereas wheat-related ones only
Jo

exerted a significant effect on ileal proportions. Further evidence in ASD children was

provided by the negative correlation of the Firmicutes/Bacteroidetes ratio with

allergy/immune function in stool (Kong et al. 2019) and with overall atopic disease

19
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

20

manifestations (asthma, allergic rhinitis and atopic dermatitis) in the cecum (Williams et

al. 2011).

Fungal diversity was only specifically studied by Strati et al 2017, who, unlike previous

culture-based studies by Kantarcioglu et al 2016 or Iovene et al 2017, did not observe

significant differences in alpha diversity between ASD and NT children. Nonetheless, he

found significant dissimilarity when beta diversity was analyzed (Strati et al. 2017), a

distinction that seemed to be mainly driven by a relative two times increase of the genus

Candida in ASD (uncorrected p value = 0.006, FDR-corrected p value = 0.09), which is

of
consistent with the two previously mentioned studies, where a higher number of isolates

ro
was recovered for every species studied and Candida was isolated from 79.8% of ASD

versus 19.6% of NT children, and 57.5% versus 0% respectively (Iovene et al. 2017;
-p
Kantarcioglu, Kiraz, and Aydin 2016). Candida albicans was largely the most
re
represented species in these two studies, whereas Strati et al 2017 did not provide a

species-level classification of Candida. Correlation analyses among the most abundant


lP

fungi and bacteria showed no significant association within ASD individuals, but a

significant positive correlation between the genera Aspergillus and Bifidobacterium was
na

found within NT subjects (Strati et al. 2017). Candida levels seemed to be increased in

ASD independent of GI symptoms (Iovene et al. 2017) and, specifically, constipation


ur

status (Strati et al. 2017), since no difference was found when ASD and NT groups were

stratified by this symptom, and difference remained significant when ASD-not


Jo

constipated (NC) and NT-NC and AD-constipated (C) vs NT-C populations were

compared (p=0.007 and p=0.05 respectively) (Strati et al. 2017). Furthermore, correlation

analysis with the most abundant and widely distributed fungi genera revealed that,

whereas Aspergillus and Malassezia did not differ between NT and ASD children,

20
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

21

regardless of constipation status, Penicillum levels showed a strong correlation with this

symptom in NT children (p<0.01), and a trend to correlate in ASD ones (p=0.07). Further

underpinning this association, higher levels of this genus in NT versus ASD children

(p=0.08) lost significance when NT-C and ASD-C children were compared (p=0.88) but

remained in the absence of constipation (p=0.07) (Strati et al. 2017).

of
3. GUT MICROBIOTA DYSBIOSIS AND GUT-BRAIN-AXIS DYSFUNCTION IN

ro
AUTISM SPECTRUM DISORDER

Currently, there seems to be a consensus about the multifactorial etiology of ASD, and
-p
that heritability and environmental contributors seem to equally influence its occurrence
re
and interact through epigenetics (Kim and Leventhal 2015; Sandin et al. 2014).

Microbiota lies at the intersection between genes and environment, as its role and
lP

composition are dependent on genetic background and crucially shaped by environmental

factors (Vuong and Hsiao 2016).


na

Regarding genetic factors, there are some gene polymorphisms that potentially increase

the risk of ASD and are distinctively associated with ASD individuals with co-occurring
ur

GI dysfunction. For instance, a variant of the Chromodomain helicase DNA binding


Jo

protein 8 gene (CHD8) has been associated with GI complaints, especially constipation,

among ASD children (Bernier et al. 2014). CHD8 is a chromatin regulator enzyme which

is essential during human fetal development. As evidenced on zebrafish and mice,

disruptive mutations of this gene result in a reduced colonization of the GI tract by enteric

neurons, and hence, slower intestinal transit and reduced intestine length along with

21
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

22

altered social interaction, increased anxiety and higher brain weight (Bernier et al. 2014;

Nithianantharajah et al. 2017), a feature that occurs in 15-35% of ASD children (Bernier

et al. 2014). The variant of c-MET is another example of gene that has been related to

ASD and comorbid FGID. It encodes the MET tyrosine kinase, leading to MET

hypofunction in the GI tract, whose endogenous ligand is called hepatocyte growth factor

(HGF), and decreases in MET expression in temporal cortex (Hsiao 2014). Similarly, the

autism-associated polymorphisms in SLC6A4, encoding the serotonin transporter

(SERT), can lead to SERT hyperfunction not only in the brain but also in the

of
gastrointestinal tract, resulting in abnormalities in behavior and gastrointestinal function,

as well as autism-related increase in blood serotonin levels (Hsiao 2014). As shown in

ro
animal models, transgenic mice expressing a human ASD-associated SERT variant
-p
exhibit the hyperserotonemia phenotype, along with core ASD-related behavioral

abnormalities and altered serotonergic signaling, causing slower motility and GI transit,
re
reduced intestinal permeability and a decrease in the number of neurons in both the
lP

myenteric and submucosal plexuses of the enteric nervous system (Adamsen et al. 2014;

Hsiao 2014).
na

On the other hand, several environmental risk factors for the development or aggravation

of ASD have been associated with a dysbiotic microbial community. It has been
ur

hypothesized that environmental factors that favor Candida colonization, such as a

prolonged antibiotic usage or reduced early life encounters with foodborne and
Jo

environmental bacteria and fungi in urban areas, could be a risk factor for ASD

development (Adams et al. 2011; Allen et al. 2017; Ding, Taur, and Walkup 2016; Strati

et al. 2017). According to these hypotheses, the increased incidence of ASD cases in the

last decades may be partially attributable to “Western” habits (i.e., high-fat diet,

22
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

23

gestational maternal obesity and diabetes, and excessive overall hygiene). Changes in gut

microbiota in babies born by Cesarean section delivery, from mothers taking drugs such

as Valproate (VPA) and ethanol during pregnancy, and in formula-fed infants have also

been proposed as possible factors related to the increased incidence of ASD (Coury et al.

2012; Li et al. 2017; Macfabe 2012; Nithianantharajah et al. 2017; Strati et al. 2017).

However, changes in reporting practices are likely to account for most of the increasing

ASD rates over the years (Hansen, Schendel, and Parner 2015; Roman, Rueda-ruzafa,

and Cardona 2018; Sadelhoff et al. 2019).

of
Maternal infections during pregnancy might be a factor linked to ASD (Patterson 2012).

ro
Recent findings showed that offspring have an altered fecal microbiota similar to ASD

individuals in a MIA model (mimic viral infection) (Hsiao et al. 2013). In addition,
-p
different microbial patterns were found in male and female ASD-like mice, but both
re
presented increased gut permeability, evident inflammatory cells infiltration and ASD-

like behavioral abnormalities (Coretti et al. 2017). These disturbances were reversed by
lP

treatment with Bacteroides fragilis (Hsiao et al. 2013). The relevance of the findings in

the MIA model is due to the clinically-common etiology and sex-specificity incidence of
na

ASD after maternal infections, which is 4 times higher in males (Ruskin et al. 2017).

Surprisingly, early in life, male infants are more likely to suffer infections than females
ur

(Klein and Flanagan 2016). In fact, it has been postulated that this sex-specific immune

sensitivity could be related to higher male prevalence in early-onset neurodevelopmental


Jo

disorders (Schwarz and Bilbo 2013). There is strong evidence that early life infections in

males can alter microglia, the resident immune cells of the brain, resulting in an

exaggerated pro-inflammatory response from microglia and impairment of learning and

memory (Bilbo et al. 2005; Williamson et al. 2011). Interestingly, microglial

23
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

24

development and myelination in the prefrontal cortex seems to be regulated by microbiota

(Roman, Rueda-ruzafa, and Cardona 2018). Adult mixed-sex groups of mice raised in a

germ-free background or depleted gut microbes have perpetually “immature” microglia

(Erny et al. 2015). This immature profile, characterized by increased proliferation and

decreased immune reactivity, can be reversed by re-colonization with a diverse bacterial

population or by treatment with metabolites derived from commensal bacteria (Erny et

al. 2015).

The gut microbiota interacts with the brain through the named gut-brain axis, that refers

of
to the bidirectional interaction pathways comprising the autonomic nervous,

ro
neuroendocrine, immunological and metabolic (via microbial toxin production) systems.

Herein, we describe the different hypothesized pathways and, when available, specific
-p
finding for ASD patients. Figure 4 shows the integration of the impaired gut-brain-axis
re
pathways in ASD individuals.
lP

3.1 Increased permeability of barrier paths


na

In general terms, homeostasis of gut microbiota is achieved when 70% of microorganisms

are Gram-negative, and the remaining 30% are Gram-positive (Mirielys and Gutiérrez
ur

2018). The microbial signature of ASD individuals, with a higher proportion of Gram-

negative bacteria (Coretti et al. 2018), increases production of lipopolysaccharides (LPS)


Jo

and pro-inflammatory cytokines (see section 3.4), which have been suggested as

contributing factors for the impairment of both the intestinal and blood-brain barrier

(BBB) found in ASD patients (Fiorentino et al. 2016). In fact, by measuring serum levels

of adhesion proteins, it has been shown that increased abnormal intestinal permeability

24
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

25

or “leaky gut” is present in 37% of ASD patients (Hsiao et al. 2013). This could be related

to lower number of Lactobacilli in patients with ASD (De Angelis et al. 2013; Iovene et

al. 2017; Ma et al. 2019; Pärtty et al. 2015), since they contribute to the maintenance of

tight junction in the intestinal epithelial barrier (Srikantha and Hasan Mohajeri 2019) and

its depletion has been directly related to chronic constipation in NT children (Kushak et

al. 2017). As a result of the impairment of the intestinal barrier, entrance of toxins and

bacterial products into the bloodstream is allowed (see below, metabolic pathway) and

bacterial translocation into the mesenteric lymphoid tissue is favored, where they activate

of
the immune system.

It should be noted that, though mucosal barrier impairment is the most studied mechanism

ro
connecting GI comorbidity and intestinal dysbiosis in ASD, this relationship appears to
-p
be much more complex. In light of this, recent research suggests a relationship between

ASD microbial profile and an altered metabolism and absorption of disaccharides in their
re
gut epithelium (Kushak et al. 2017; Srikantha and Hasan Mohajeri 2019).
lP

3.2 Anatomical pathway: The gut-brain’s neural network


na

Two neuroanatomical routes are known to deliver the signals from the intestine to the

brain. The first is the autonomic nervous system and the vagus nerve, and the second is
ur

the enteric nervous system, including the enteroglial cells and the autonomic nervous
Jo

system and vagal nerve in the spinal cord. The gut luminal contents and events and the

mucosal constituents create the signals to be transmitted by hierarchic integrative levels

cephalically (Lerner, Neidhöfer, and Matthias 2017). Regarding this pathway, certain

anatomical changes have been found in the brains of autistic individuals, such as impaired

GABAergic functioning (Horder et al. 2018) and increased activation of microglial cells
25
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

26

and a lower count of Purkinje cells in the cerebellum, which could be related to the first

(Srikantha and Hasan Mohajeri 2019).

3.3 Neuroendocrine pathway:

3.3.1 Hypothalamic-pituitary-adrenal (HPA) axis

The primary function of the HPA axis is to regulate the response to environmental factors,

such as stress. Under stress conditions, the hypothalamus releases Corticotropin-

of
releasing-hormone (CRH) and vasopressin that signal the release of Adrenocorticotropin

ro
(ACTH) from the pituitary gland, which in turn influences the secretion of hormones from

the adrenal glands, such as cortisol, a glucocorticoid that affects many human organs,
-p
including the brain, where it can regulate the activity of the intestinal functional effector

cells, modulating GI motility, permeability, immunity and mucus. These same cells are
re
under the influence of the gut microbiota (Carabotti et al. 2015) (Figure 5). Germ free
lP

(GF) mice show an increased stress response with augmented levels of ACTH and

cortisol, which can be reversed after germ colonization in young mice(Lerner, Neidhöfer,

and Matthias 2017), by fecal microbial transplant or by Bifidobacterium infantis


na

(Evrensel and Ceylan 2016). GF mice also display reduced brain-derived neurotrophic

factor (BDNF) and N-methyl-D-aspartate (NMDA) receptor expressions in cortex and


ur

hippocampus, which affect the release and expression of the CRH in the hypothalamus
Jo

and thus change the function of the HPA axis (Sudo et al. 2004). Some studies,

specifically in ASD patients, have found altered mRNA levels of the glucocorticoid

receptor and CRH receptor 1 (Patel et al. 2016), which essentially implies an alteration

of this pathway.

26
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

27

3.3.2 Neurotransmitters and Neural regulators

Gut bacteria seem to regulate several key neurotransmitters such as gamma amino butyric

acid (GABA), glutamate, serotonin, dopamine (Yano et al. 2015; Yunes et al. 2016),

which have shown altered levels in ASD patients (Mohamadkhani 2018; Srikantha and

Hasan Mohajeri 2019). In fact, an imbalance in the CNS between excitation (glutamate)

and inhibition (GABA) has been postulated to contribute to ASD (El-Ansary et al. 2018;

Horder et al. 2018; Srikantha and Hasan Mohajeri 2019). In addition, bacteria can produce

a wide range of neuroendocrine hormones which can intervene in intestinal homeostasis

of
and modulate mood and behavior (Lyte 2014; Oleskin, Shenderov, and Rogovsky 2017).

ro
More specifically, it has been theorized that both the SLC6A4 gene polymorphism

discussed above and the increase of serotonin-producing microbes, such as Candida,


-p
Streptococcus, Escherichia, Enterococcus and Clostridiales in ASD may rise intestinal
re
production of serotonin at the expense of a lower synthesis in the brain (due to

consumption of its precursor tryptophan), leading to hyperserotoninemia, intestinal


lP

dysmotility and a still inconsistently reported behavioral outcome (Fattorusso et al. 2019;

Luna et al. 2017; Srikantha and Hasan Mohajeri 2019).


na

3.4 Immunological pathway: cytokine release


ur

The development of the immune system heavily relies on gut microbiota; hence, GF mice

have limited immune activity (Lerner, Neidhöfer, and Matthias 2017). Most bacteria and
Jo

host communication is carried out by TLRs (Toll-Like Receptors), which are found in a

variety of cells from the innate immune system, to intestinal epithelial cells or neurons

(both from the ENS and CNS). TLRs can recognize the microbiota triggering appropriate

intracellular signaling pathways and immune responses. Among other bacterial products,

27
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

28

increased LPS in ASD subjects could activates TLR4 in the ENS, where stimulates pro-

inflammatory cytokine production, and in the CNS, where it can lead to inflammation

through activating microglia once crossed the impaired mucosal and blood-brain-barriers

(Li et al. 2017; Srikantha and Hasan Mohajeri 2019), as it has been shown in postmortem

ASD brain biopsies (Morgan et al. 2010). Likewise, cytokines can bind TLRs in neurons,

inducing changes in the electric layout of neuronal membranes and altering the regulation

of emotion and behavior (Evrensel and Ceylan 2016).

The microbial imbalance in ASD patients could induce an inadequate immune activation

of
leading to cytokine dysregulation, thereby activating the HPA axis and altering the

ro
permeability of the gut and blood-brain barrier through MLCK and MAPCK-mediated

regulation of tight junction (Ahmad et al. 2017; Barrier et al. 2014; Carabotti et al. 2015;
-p
Ulluwishewa et al. 2018) (Figure 6). Further evidence is provided by studies carried out
re
in both ASD models and ASD individuals who displayed increased LCR, plasma or serum

pro-inflammatory and Th2 cell-derived cytokines, such as IL-1, IL-4, IL-5, IL-6, IL- 8,
lP

TNF-α, IFN- and IL-17 , as well as low IL-10, TGF- and Treg cells levels, which are

involved in synapse, survival and differentiation of neurons, and correlate with the
na

severity of ASD symptoms (Eftekharian, Ghafouri-fard, and Noroozi 2018; Sadelhoff et

al. 2019). Further correlation between faecal TNF-α and the severity of GI symptoms was
ur

reported in ASD children (Srikantha and Hasan Mohajeri 2019). Moreover,

Faecalibacterium abundance in ASD children was strongly correlated with a greater


Jo

number of differentially expressed genes involved in both the interferon (IFN)-γ and type-

I IFN signaling pathways, which were enriched in this cohort (Inoue et al. 2016).

T-helper 1 and T-helper 17 cells affect the reactivity of peripheral immune cells in the

CNS and the integrity of blood-brain barrier, induce apoptosis in oligodendrocytes and

28
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

29

increase glutamate excitotoxicity (Eftekharian, Ghafouri-fard, and Noroozi 2018;

Kamada et al. 2013; Stolp et al. 2005); their hyper-activation has been linked to the

increase of Clostridiales (Luna et al. 2017) and disruptions in the mammalian target of

rapamycin (mTOR) pathway in some ASD phenotypes. Enhanced mTOR activity is also

responsible for increased autophagy in the intestine and disbalance in allergy-associated

Th2 and Treg cells (Sadelhoff et al. 2019). This evidence, together with a ten-fold

incidence of mastocytosis found in ASD patients, the similarities in gut biopsies of

children with ASD and individuals with immunodeficiency and food allergies, and the

of
previously mentioned increased prevalence of food allergies in this population, points to

a relationship between intestinal allergic responses and brain circuits involved in social

ro
functioning, which can be targeted by rapamycin and amino-acid administration

(Fattorusso et al. 2019; Sadelhoff et al. 2019). -p


re
3.5 Metabolic pathway: microbial metabolome
lP

The gut microbiota produces constantly changing metabolites that impact host physiology

and susceptibility to disease (Lerner, Neidhöfer, and Matthias 2017). ASD patients show
na

altered levels of potentially toxic phenol compounds (Alabdali, Al-Ayadhi, and El-

Ansary 2014) produced, among others, by Bifidobacterium, C. difficile, C. histolyticum


ur

clusters II and I and some Lactobacillus (Ding, Taur, and Walkup 2016; Fattorusso et al.
Jo

2019; Mohamadkhani 2018). Among them, the increased abundance of urinary and fecal

para-cresol (p-cresol) and its conjugated derivative p-cresylsulfate, which might impair

oligodendrocyte differentiation in vitro and can inhibit the enzyme dopamine-beta-

hydroxylase, are considered putative metabolic markers for ASD, especially useful in

females and severely autistic males (Persico and Napolioni 2013; Roman, Rueda-ruzafa,
29
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

30

and Cardona 2018). Along with it, increased Clostridia derived metabolite 3-(3-

hydroxyphenyl)-3-hydroxypropionic acid (HPHPA) could lead to a depletion of

catecholamines in the CNS and its reduction has been shown to decrease ASD severity

(Roman, Rueda-ruzafa, and Cardona 2018; Xiong et al. 2016). Another significant

metabolite increased in stool of children with ASD is isopropanol, which has been related

to GI disturbances (Mohamadkhani 2018), whereas relevant findings in tue urinary profile

point to the alteration of nicotinamide, tryptophan, vitamine B6 and purine metabolism

(Srikantha and Hasan Mohajeri 2019).

of
Other metabolites plausibly involved in ASD pathogenesis could be short-chain fatty

ro
acids (SCFAs), which are fermentation products of dietary carbohydrates produced by

intestinal bacteria such as Clostridia, Bifidobacteria, Bacteroidetes, the families


-p
Ruminococcaceae and Lachnospiraceae, and the genus Desulfovibrio (Fattorusso et al.
re
2019; Roman, Rueda-ruzafa, and Cardona 2018; Williams et al. 2011). Interestingly, the

overall structure of ASD children was found to be dominated by carbohydrate


lP

metabolism, among other functions (Ma et al. 2019), and the abundance of SCFA-

producing bacteria has been reported to be altered in ASD individuals (Zhang et al. 2018).
na

Despite data being somewhat discordant, findings suggest decreased butyrate, and

increased acetate and propionate (PPA) levels in ASD patients (De Angelis et al. 2013b;
ur

Grimaldi et al. 2016; L Wang 2010). Although PPA is beneficial at appropriate levels

(den Besten et al. 2013), excessive amounts are associated with the severity of ASD (Lv
Jo

Wang et al. 2012). In fact, its acidic properties may make them liposoluble enough to

cross the gut and blood-brain barriers and gain access to the brain, where it could induce

several ASD-linked behavioral and neurochemical changes, such as neurotransmitter

alteration (dopamine, serotonin and glutamate), increased oxidative stress,

30
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

31

neuroinflammation, and intracellular acidification, which can inhibit mitochondrial

function and may contribute to causing gut dysmotility and altering development and

behavior (Foley et al. 2014; Macfabe 2012; Roman, Rueda-ruzafa, and Cardona 2018;

Srikantha and Hasan Mohajeri 2019).

4. CONCLUSIONS

The lack of consistent knowledge inherent to the emerging nature of the field and

of
incomplete understanding of the gut-brain biological signaling pathways, together with

ro
the disparity of sample sizes, demographics and methodologies used, translates into

discordant differences reported for bacterial gut microbiota in ASD individuals. In fact,
-p
measuring tools varied widely, not only for autism diagnosis, microbial and GI

characterization, but also for the statistical analysis of findings, leading to heterogeneous
re
p-values in terms of conservatism. Besides, the effect of change for each taxon targeted
lP

could not always be given, since not all studies provided calculations or enough data to

calculate them. In one case, this data, while existing, could not be accessed (Carissimi et

al. 2019). Finally, it should be kept in mind that bacterial abundances should be
na

segregated by absolute and relative amounts in order to accurately establish comparisons.

All in all, inferences regarding changes of individual bacteria at each level should be
ur

cautiously made.
Jo

Despite more research being needed to shed light on the many questions that remain

unanswered, the present review concludes that there is general agreement on the existence

of a distinctive microbial pattern in ASD individuals that can be affected by

demographics, autistic phenotypical profile and symptomatology, diet pattern as well as

GI and autoimmune comorbidity. However, for a deeper clarification regarding the


31
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

32

relationship between GI disorders and ASD, studies investigating the correlation between

GI symptoms and gut microbiota in NT groups should have been included.

Overall, there is a need for further research on the interactions between gut bacterial and

fungal microbiota, the role of the bacterial and fungal components in the neuroplastic

changes and gastrointestinal physiology in ASDs, and the integration of such data with

genetics, immunology, and metabolomics. Not only can this provide novel insights into

the intricate gut microbiota-brain axis, whose relevance to ASD is herein evaluated, but

it can also help us thoroughly identify the distinctive ASD microbiome and ASD specific

of
biomarkers, useful in the determination of at-risk population, early ASD detection and

treatment development.

ro
Declarations of interest: none
-p
re
Acknowledgements
lP

The authors are grateful to Dr Manuel Maynar for his decisive role in the genesis and

development of this research through his critical reading of the first drafts. Also, we thank
na

Dr Jeffery for her critical reading of the manuscript. This research did not receive any

specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
ur
Jo

32
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

33

REFERENCES

Adams, James B et al. 2011. “Gastrointestinal Flora and Gastrointestinal Status in


Children with Autism – Comparisons to Typical Children and Correlation with
Autism Severity.” BMC Gastroenterology 11(1): 22.
http://www.biomedcentral.com/1471-230X/11/22.
Adamsen, Dea et al. 2014. “Autism Spectrum Disorder Associated with Low Serotonin
in CSF and Mutations in the SLC29A4 Plasma Membrane Monoamine Transporter
(PMAT) Gene.” Molecular Autism: 1–11.
Ahmad, Sheikh F et al. 2017. “Imbalance between the Anti- and pro-Inflammatory Milieu
in Blood Leukocytes of Autistic Children.” Molecular Immunology 82: 57–65.
Alabdali, Altaf, Laila Al-Ayadhi, and Afaf El-Ansary. 2014. “Association of Social and
Cognitive Impairment and Biomarkers in Autism Spectrum Disorders.” Journal of

of
Neuroinflammation 11(1): 4.
Allen, Andrew P., Timothy G. Dinan, Gerard Clarke, and John F. Cryan. 2017. “A
Psychology of the Human Brain–Gut–Microbiome Axis.” Social and Personality

ro
Psychology Compass 11(4): 1–22.
De Angelis, Maria et al. 2013. “Fecal Microbiota and Metabolome of Children with
Autism and Pervasive Developmental Disorder Not Otherwise Specified.” PLoS
-p
ONE 8(10): 1–18.
Barrier, Blood-brain et al. 2014. “The Gut Microbiota Influences Blood-Brain Barrier
re
Permeability in Mice.” 6(263).
Bernier, Raphael et al. 2014. “Disruptive CHD8 Mutations Define a Subtype of Autism
Early in Development.”
lP

Cell 158(2): 263–76.


http://dx.doi.org/10.1016/j.cell.2014.06.017.
den Besten, Gijs et al. 2013. “The Role of Short-Chain Fatty Acids in the Interplay
between Diet, Gut Microbiota, and Host Energy Metabolism.” Journal of Lipid
na

Research 54(9): 2325–40. http://www.jlr.org/lookup/doi/10.1194/jlr.R036012.


Bilbo, Staci D et al. 2005. “Neonatal Infection-Induced Memory Impairment after
Lipopolysaccharide in Adulthood Is Prevented via Caspase-1 Inhibition.” 25(35):
ur

8000–8009.
Buffington, Shelly A et al. 2017. “Microbial Reconstitution Reverses Maternal Diet-
Induced Social and Synaptic Deficits in Offspring.” 165(7): 1762–75.
Jo

Carabotti, Marilia, Annunziata Scirocco, Maria Antonietta Maselli, and Carola Severi.
2015. “The Gut-Brain Axis: Interactions between Enteric Microbiota, Central and
Enteric Nervous Systems.” Annals of Gastroenterology 28(2): 203–9.
Carissimi, Claudia et al. 2019. “Functional Analysis of Gut Microbiota and
Immunoinflammation in Children with Autism Spectrum Disorders.” Digestive and
Liver Disease 51: 1366–74.
Chaidez, Virginia, Robin L Hansen, and Irva Hertz-picciotto. 2014. “Gastrointestinal
33
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

34

Problems in Children with Autism , Developmental Delays or Typical


Development.” Journal of Autism and Developmental Disorders 44(5): 1117–27.
Christensen, Deborah L et al. 2018. “Prevalence and Characteristics of Autism Spectrum
Disorder Among Children Aged 8 Years - Autism and Developmental Disabilities
Monitoring Network, 11 Sites, United States, 2012.” Morbidity and mortality weekly
report. Surveillance summaries (Washington, D.C. : 2002) 65(13): 1–23.
Coretti, Lorena et al. 2017. “Sex-Related Alterations of Gut Microbiota Composition in
the BTBR Mouse Model of Autism Spectrum Disorder.” Nature Publishing Group
(November 2016): 1–10.
———. 2018. “Gut Microbiota Features in Young Children with Autism Spectrum
Disorders.” Frontiers in Microbiology 9(December): 1–12.
Coury, Daniel L. et al. 2012. “Gastrointestinal Conditions in Children With Autism
Spectrum Disorder: Developing a Research Agenda.” Pediatrics 130(Supplement

of
2): S160–68. http://pediatrics.aappublications.org/lookup/doi/10.1542/peds.2012-
0900N.

ro
Ding, Helen T, Ying Taur, and John T Walkup. 2016. “Gut Microbiota and Autism : Key
Concepts and Findings.” Journal of Autism and Developmental Disorders 0(0): 0.

-p
Eftekharian, Mohammad Mahdi, Soudeh Ghafouri-fard, and Rezvan Noroozi. 2018.
“Cytokine Cytokine pro Fi Le in Autistic Patients.” 108(November 2017): 120–26.
El-Ansary, Afaf et al. 2018. “Probiotic Treatment Reduces the Autistic-like
re
Excitation/Inhibition Imbalance in Juvenile Hamsters Induced by Orally
Administered Propionic Acid and Clindamycin.” Metabolic Brain Disease: 1–10.
Elrod, Marilisa G, and Bradley S Hood. 2015. “Sleep Differences Among Children With
lP

Autism Spectrum Disorders and Typically Developing Peers: A Meta-Analysis.”


Journal of Developmental & Behavioral Pediatrics 36(3, April): 166–77.
Elsabbagh, Mayada et al. 2012. “Global Prevalence of Autism and Other Pervasive
na

Developmental Disorders.” Aurism Research 5(3): 160–79.


Erny, Daniel et al. 2015. “Host Microbiota Constantly Control Maturation and Function
of Microglia in the CNS.” Nature Neuroscience 18: 965.
ur

Evrensel, Alper, and Mehmet Emin Ceylan. 2016. “Fecal Microbiota Transplantation and
Its Usage in Neuropsychiatric Disorders.” Clinical psychopharmacology and
neuroscience : the official scientific journal of the Korean College of
Jo

Neuropsychopharmacology 14(3): 231–37.


http://www.ncbi.nlm.nih.gov/pubmed/27489376%5Cnhttp://www.pubmedcentral.n
ih.gov/articlerender.fcgi?artid=PMC4977816.
Fattorusso, Antonella et al. 2019. “Autism Spectrum Disorders and the Gut Microbiota.”
Nutrients 11(3).
Finegold, Sydney M. et al. 2002. “Gastrointestinal Microflora Studies in Late‐ Onset
Autism.” Clinical Infectious Diseases 35(s1): S6–16.
https://academic.oup.com/cid/article-lookup/doi/10.1086/341914.
34
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

35

Finegold, Sydney M. et al. 2010a. “Pyrosequencing Study of Fecal Microflora of Autistic


and Control Children.” Anaerobe 16(4): 444–53.
http://dx.doi.org/10.1016/j.anaerobe.2010.06.008.
Finegold, Sydney M et al. 2010b. “Pyrosequencing Study of Fecal Microflora of Autistic
and Control Children.” Anaerobe 16(4): 444–53.
Fiorentino, Maria et al. 2016. “Blood–Brain Barrier and Intestinal Epithelial Barrier
Alterations in Autism Spectrum Disorders.” Molecular Autism 7(1): 49.
Foley, Kelly A., Klaus Peter Ossenkopp, Martin Kavaliers, and Derrick F. MacFabe.
2014. “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 9(1): 1–13.
Gondalia, Shakuntla V. et al. 2012. “Molecular Characterisation of Gastrointestinal
Microbiota of Children With Autism (With and Without Gastrointestinal

of
Dysfunction) and Their Neurotypical Siblings.” Autism Research 5(6): 419–27.
Gorrindo, Phillip, Kent C Williams, Evon B Lee, and Lynn S Walker. 2012.

ro
“Gastrointestinal Dysfunction in Autism.” Autism Res 5(2): 101–8.
Grimaldi, Roberta et al. 2016. “In Vitro Fermentation of B-GOS: Impact on Faecal
-p
Bacterial Populations and Metabolic Activity in Autistic and Non-Autistic
Children.” FEMS Microbiology Ecology 93(2): 1–10.
Gyawali, Shreeya, and Bichitra Nanda Patra. 2019. “Trends in Concept and Nosology of
re
Autism Spectrum Disorder: A Review 1.” Asian Journal of Psychiatry.
https://doi.org/10.1016/j.ajp.2019.01.021.
Hang, Jun et al. 2014. “16S RRNA Gene Pyrosequencing of Reference and Clinical
lP

Samples and Investigation of the Temperature Stability of Microbiome Profiles.”


2(1): 1–15.
Hansen, Stefan N, Diana E Schendel, and Erik T Parner. 2015. “Explaining the Increase
na

in the Prevalence of Autism Spectrum Disorders The Proportion Attributable to


Changes in Reporting Practices.” JAMA Pediatrics 169(1): 56–62.
Holingue, Calliope et al. 2018. “Gastrointestinal Symptoms in Autism Spectrum
Disorder: A Review of the Literature on Ascertainment and Prevalence.” 11(1): 24–
ur

36.
Horder, Jamie et al. 2018. “Glutamate and GABA in Autism Spectrum Disorder — a
Jo

Translational Magnetic Resonance Spectroscopy Study in Man and Rodent Models.”


Translational Psychiatry: 1–11.
Hsiao, Elaine Y. et al. 2013. “Microbiota Modulate Behavioral and Physiological
Abnormalities Associated with Neurodevelopmental Disorders.” Cell 155(7): 1451–
63. http://dx.doi.org/10.1016/j.cell.2013.11.024.
———. 2014. “Gastrointestinal Issues in Autism Spectrum Disorder.” Harvard Review
of Psychiatry 22(2): 104–11.

35
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

36

Inoue, Ryo et al. 2016. “A Preliminary Investigation on the Relationship between Gut
Microbiota and Gene Expressions in Peripheral Mononuclear Cells of Infants with
Autism Spectrum Disorders.” Bioscience, Biotechnology and Biochemistry 80(12):
2450–58. http://dx.doi.org/10.1080/09168451.2016.1222267.
Iovene, Maria Rosaria et al. 2017. “Intestinal Dysbiosis and Yeast Isolation in Stool of
Subjects with Autism Spectrum Disorders.” Mycopathologia 182(3–4): 349–63.
Kamada, Nobuhiko, Sang Uk Seo, Grace Y. Chen, and Gabriel Núñez. 2013. “Role of
the Gut Microbiota in Immunity and Inflammatory Disease.” Nature Reviews
Immunology 13(5): 321–35.
Kang, Dae-Wook et al. 2013. “Reduced Incidence of Prevotella and Other Fermenters in
Intestinal Microflora of Autistic Children.” PLOS ONE 8(7): e68322.
https://doi.org/10.1371/journal.pone.0068322.
Kang, Dae Wook et al. 2018. “Differences in Fecal Microbial Metabolites and Microbiota

of
of Children with Autism Spectrum Disorders.” Anaerobe 49: 121–31.
https://doi.org/10.1016/j.anaerobe.2017.12.007.

ro
Kantarcioglu, A. Serda, Nuri Kiraz, and Ahmet Aydin. 2016. “Microbiota–Gut–Brain
Axis: Yeast Species Isolated from Stool Samples of Children with Suspected or
Diagnosed Autism Spectrum Disorders and In Vitro Susceptibility Against Nystatin
-p
and Fluconazole.” Mycopathologia 181(1–2): 1–7.
Kim, Young Shin, and Bennett L. Leventhal. 2015. “Genetic Epidemiology and Insights
into Interactive Genetic and Environmental Effects in Autism Spectrum Disorders.”
re
Biological Psychiatry 77(1): 66–74.
Klein, Sabra L, and Katie L Flanagan. 2016. “Sex Differences in Immune Responses.”
lP

Nature Publishing Group 16(10): 626–38.


Kong, Xuejun et al. 2019. “New and Preliminary Evidence on Altered Oral and Gut
Microbiota in Individuals with Autism Spectrum Disorder (ASD): Implications for
ASD Diagnosis and Subtyping Based on Microbial Biomarkers.” Nutrients 11(9):
na

2128.
Kushak, Rafail I. et al. 2017. “Analysis of the Duodenal Microbiome in Autistic
Individuals; Association with Carbohydrate Digestion.” Journal of Pediatric
ur

Gastroenterology and Nutrition 64: e110–16.


Lee, Yunjin et al. 2017. “Rapid Assessment of Microbiota Changes in Individuals with
Autism Spectrum Disorder Using Bacteria-Derived Membrane Vesicles in Urine.”
Jo

Experimental Neurobiology 26(5): 307–17.


Lerner, Aaron, Sandra Neidhöfer, and Torsten Matthias. 2017. “The Gut Microbiome
Feelings of the Brain: A Perspective for Non-Microbiologists.” Microorganisms
5(4): 66. http://www.mdpi.com/2076-2607/5/4/66.
Li, Qinrui, Ying Han, Angel Belle C. Dy, and Randi J. Hagerman. 2017. “The Gut
Microbiota and Autism Spectrum Disorders.” Frontiers in Cellular Neuroscience
11(April). http://journal.frontiersin.org/article/10.3389/fncel.2017.00120/full.

36
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

37

Liu, Simeng et al. 2019. “Altered Gut Microbiota and Short Chain Fatty Acids in Chinese
Children with Autism Spectrum Disorder.” Scientific Reports 9(1): 1–9.
http://dx.doi.org/10.1038/s41598-018-36430-z.
Luna, Ruth Ann et al. 2017. “Distinct Microbiome-Neuroimmune Signatures Correlate
With Functional Abdominal Pain in Children With Autism Spectrum Disorder.”
Cellular and Molecular Gastroenterology and Hepatology 3(2): 218–30.
http://dx.doi.org/10.1016/j.jcmgh.2016.11.008.
Luna, Ruth Ann, Tor C Savidge, and Kent C Williams. 2016. “The Brain-Gut-
Microbiome Axis : What Role Does It Play In.” Current Developmental Disorders
Reports 3(1): 75–81.
Lyte, Mark. 2014. “Host-Microbiota Neuroendocrine Interactions Influencing Brain and
Behavior Microbial Endocrinology.” Gut Microbes 5(3): 381–89.
Ma, Bingjie et al. 2019. “Altered Gut Microbiota in Chinese Children with Autism

of
Spectrum Disorders.” Frontiers in Cellular and Infection Microbiology 9(MAR): 1–
10.

ro
Macfabe, Derrick F. 2012. “Short-Chain Fatty Acid Fermentation Products of the Gut
Microbiome: Implications in Autism Spectrum Disorders.” Microbial Ecology in
Health & Disease 1: 1–25.
-p
Maenner, Matthew J et al. 2012. “Brief Report : Association Between Behavioral
Features and Gastrointestinal Problems Among Children with Autism Spectrum
Disorder.” : 1520–25.
re
Mangiola, Francesca et al. 2016. “Gut Microbiota in Autism and Mood Disorders.” World
Journal of Gastroenterology 22(1): 361–68.
lP

McElhanon, Barbara O, Courtney McCracken, Saul Karpen, and William G Sharp. 2014.
“Gastrointestinal Symptoms in Autism Spectrum Disorder: A Meta-Analysis.”
Pediatrics.
http://pediatrics.aappublications.org/content/early/2014/04/22/peds.2013-
na

3995.abstract.
Mirielys, Lic, and Pérez Gutiérrez. 2018. “Nuevos Aportes Desde La Microbiología Para
Entender El Autismo New Contributions from the Microbiology in Order to
ur

Understand the Autism.” 22(2).


Mohamadkhani, Ashraf. 2018. “Gut Microbiota and Fecal Metabolome Perturbation in
Children with Autism Spectrum Disorder.” Middle East Journal of Digestive
Jo

Diseases 10(4): 205–12.


Morgan, John T et al. 2010. “Microglial Activation and Increased Microglial Density
Observed in the Dorsolateral Prefrontal Cortex in Autism.” BPS 68(4): 368–76.
Nikolov, Roumen N. et al. 2009. “Gastrointestinal Symptoms in a Sample of Children
with Pervasive Developmental Disorders.” 39: 405–13.
Nithianantharajah, Jess, Gayathri K. Balasuriya, Ashley E. Franks, and Elisa L. Hill-
Yardin. 2017. “Using Animal Models to Study the Role of the Gut–Brain Axis in
37
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

38

Autism.” Current Developmental Disorders Reports 4(2): 28–36.


http://link.springer.com/10.1007/s40474-017-0111-4.
Oleskin, Alexander V, Boris A Shenderov, and Vladimir S Rogovsky. 2017. “Role of
Neurochemicals in the Interaction between the Microbiota and the Immune and the
Nervous System of the Host Organism.” Probiotics and Antimicrobial Proteins 9(3):
215–34.
Parracho, H M R T et al. 2010. “A Double-Blind, Placebo-Controlled, Crossover-
Designed Probiotic Feeding Study in Children Diagnosed with Autistic Spectrum
Disorders.” International Journal of Probiotics and Prebiotics 5(2): 69–74.
http://centaur.reading.ac.uk/17353/.
Parracho, Helena M R T, Max O. Bingham, Glenn R. Gibson, and Anne L. McCartney.
2005. “Differences between the Gut Microflora of Children with Autistic Spectrum
Disorders and That of Healthy Children.” Journal of Medical Microbiology 54(10):

of
987–91.
Pärtty, Anna et al. 2015. “A Possible Link between Early Probiotic Intervention and the
Risk of Neuropsychiatric Disorders Later in Childhood: A Randomized Trial.”

ro
Pediatric Research 77(6): 823–28.
Patel, Neil et al. 2016. “Altered MRNA Levels of Glucocorticoid Receptor,
-p
Mineralocorticoid Receptor, and Co-Chaperones (FKBP5 and PTGES3) in the
Middle Frontal Gyrus of Autism Spectrum Disorder Subjects.” Molecular
Neurobiology 53(4): 2090–99.
re
Patterson, Paul H. 2012. “Maternal Infection and Immune Involvement in Autism.”
Trends in Molecular Medicine 17(7): 389–94.
Persico, Antonio M, and Valerio Napolioni. 2013. “Neurotoxicology and Teratology
lP

Urinary p -Cresol in Autism Spectrum Disorder.” Neurotoxicology and Teratology


36(2012): 82–90.
Plaza-Díaz, Julio et al. 2019. “Autism Spectrum Disorder (ASD) with and without Mental
na

Regression Is Associated with Changes in the Fecal Microbiota.” Nutrients 11(2).


Pulikkan, Joby et al. 2018. “Gut Microbial Dysbiosis in Indian Children with Autism
Spectrum Disorders.” Microbial Ecology 76(4): 1102–14.
ur

Roman, Pablo, Lola Rueda-ruzafa, and Diana Cardona. 2018. “Gut – Brain Axis in the
Executive Function of Austism Spectrum Disorder.” : 654–63.
Jo

Rosselli, Riccardo et al. 2016. “Direct 16S RRNA-Seq from Bacterial Communities : A
PCR-Independent Approach to Simultaneously Assess Microbial Diversity and
Functional Activity Potential of Each Taxon.” Nature Publishing Group (February):
1–12. http://dx.doi.org/10.1038/srep32165.
Ruskin, David N., Michelle I. Murphy, Sierra L. Slade, and Susan A. Masino. 2017.
“Ketogenic Diet Improves Behaviors in a Maternal Immune Activation Model of
Autism Spectrum Disorder.” PLoS ONE 12(2): 1–14.
Sadelhoff, Joris H J Van, Paula Perez Pardo, Jiangbo Wu, and Johan Garssen. 2019. “The
38
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

39

Gut-Immune-Brain Axis in Autism Spectrum Disorders ; A Focus on Amino Acids.”


10(April): 1–15.
Sandin, Sven et al. 2014. “The Familial Risk of Autism.” JAMA - Journal of the American
Medical Association 311(17): 1770–77.
Sandler, Richard H. et al. 2000. “Short-Term Benefit from Oral Vancomycin Treatment
of Regressive-Onset Autism.” Journal of Child Neurology 15(7): 429–35.
Schwarz, Jaclyn M., and Staci D. Bilbo. 2013. “Sex, Glia, and Development: Interactions
in Health and Disease.” Hormones and Behavior 62(3): 243–53.
Sharp, William G et al. 2013. “Feeding Problems and Nutrient Intake in Children with
Autism Spectrum Disorders : A Meta-Analysis and Comprehensive Review of the
Literature.”
Son, Joshua S. et al. 2015. “Comparison of Fecal Microbiota in Children with Autism

of
Spectrum Disorders and Neurotypical Siblings in the Simons Simplex Collection.”
PLoS ONE 10(10): 1–19.

ro
Song, Yuli et al. 2003. “Clostridium Bolteae Sp. Nov., Isolated from Human Sources.”
Systematic and Applied Microbiology 26(1): 84–89.
Song, Yuli, Chengxu Liu, and Sydney M Finegold. 2004. “Real-Time PCR Quantitation
-p
of Clostridia in Feces of Autistic Children.” 70(11): 6459–65.
Srikantha, Piranavie, and M. Hasan Mohajeri. 2019. “The Possible Role of the
Microbiota-Gut-Brain-Axis in Autism Spectrum Disorder.” International Journal of
re
Molecular Sciences 20(9): 14–19.
Stolp, H B et al. 2005. “Long-Term Changes in Blood–Brain Barrier Permeability and
lP

White Matter Following Prolonged Systemic Inflammation in Early Development in


the Rat.” European Journal of Neuroscience 22(11): 2805–16.
Strati, Francesco et al. 2017. “New Evidences on the Altered Gut Microbiota in Autism
Spectrum Disorders.”
na

Microbiome 5(1): 24.


http://microbiomejournal.biomedcentral.com/articles/10.1186/s40168-017-0242-1.
Sudo, Nobuyuki et al. 2004. “Postnatal Microbial Colonization Programs the
Hypothalamic – Pituitary – Adrenal System for Stress Response in Mice.” 1: 263–
ur

75.
Tomova, Aleksandra et al. 2015. “Gastrointestinal Microbiota in Children with Autism
in Slovakia.”
Jo

Physiology and Behavior 138: 179–87.


http://dx.doi.org/10.1016/j.physbeh.2014.10.033.
Tsai, Luke Y. 2014. “Research in Autism Spectrum Disorders Impact of DSM-5 on
Epidemiology of Autism Spectrum Disorder.” Research in Autism Spectrum
Disorders 8(11): 1454–70. http://dx.doi.org/10.1016/j.rasd.2014.07.016.
Ulluwishewa, Dulantha et al. 2018. “Regulation of Tight Junction Permeability by
Intestinal Bacteria and Dietary Components 1 , 2.” (September): 769–76.

39
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

40

Vuong, Helen E, and Elaine Y Hsiao. 2016. “Author ’ s Accepted Manuscript Emerging
Roles for the Gut Microbiome in Autism Spectrum DisorderThe Microbiome in
ASD Emerging Roles for the Gut Microbiome in Autism Spectrum Disorder.”
Biological Psychiatry. http://dx.doi.org/10.1016/j.biopsych.2016.08.024.
Wang, Hong Xing, and Yu Ping Wang. 2016. “Gut Microbiota ‑ Brain Axis.” 129(19):
2373–80.
Wang, L. 2010. 25 J Gastroenterol Hepatol Gut Bacterial and Fermentation Profiles Are
Altered in Children with Autism.
Wang, Lv et al. 2011. “Low Relative Abundances of the Mucolytic Bacterium
Akkermansia Muciniphila and Bifidobacterium Spp. in Feces of Children with
Autism.” Applied and Environmental Microbiology 77(18): 6718–21.
———. 2012. “Elevated Fecal Short Chain Fatty Acid and Ammonia Concentrations in
Children with Autism Spectrum Disorder.” Digestive Diseases and Sciences 57(8):

of
2096–2102.
———. 2013. “Increased Abundance of Sutterella Spp . and Ruminococcus Torques in

ro
Feces of Children with Autism Spectrum Disorder.” Molecular Autism 4(1): 1.
Molecular Autism.
Williams, Brent L. et al. 2011. “Impaired Carbohydrate Digestion and Transport and
-p
Mucosal Dysbiosis in the Intestines of Children with Autism and Gastrointestinal
Disturbances.” PLoS ONE 6(9).
re
Williams, Brent L, Mady Hornig, and Tanmay Parekh. 2012a. “Application of Novel
PCR-Based Methods for Detection, Quantitation, and Phylogenetic Characterization
Of.” mBIo 3(1): 1–11.
lP

Williamson, Lauren L et al. 2011. “Microglia and Memory : Modulation by Early-Life


Infection.” 31(43): 15511–21.
Xiong, Xiyue et al. 2016. “Urinary 3-(3-Hydroxyphenyl)-3-Hydroxypropionic Acid, 3-
na

Hydroxyphenylacetic Acid, and 3-Hydroxyhippuric Acid Are Elevated in Children


with Autism Spectrum Disorders.” BioMed Research International 2016.
Xu, Guifeng, Lane Strathearn, Buyun Liu, and Wei Bao. 2018. “Corrected Prevalence of
Autism Spectrum Disorder Among US Children and Adolescents.” Jama 28: 4–5.
ur

http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.2018.0001.
Yang, Yongshou, Jinhu Tian, and Bo Yang. 2018. “Targeting Gut Microbiome: A Novel
Jo

and Potential Therapy for Autism.” Life Sciences 194(August 2017): 111–19.
https://doi.org/10.1016/j.lfs.2017.12.027.
Yano, Jessica M et al. 2015. “Indigenous Bacteria from the Gut Microbiota Regulate Host
Serotonin Biosynthesis.” Cell 161: 264–76.
Yunes, R A et al. 2016. “Anaerobe GABA Production and Structure of GadB / GadC
Genes in Lactobacillus and Bi Fi Dobacterium Strains from Human Microbiota.”
Anaerobe 42: 197–204.

40
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

41

Zhang, Mengxiang et al. 2018. “Analysis of Gut Microbiota Profiles and Microbe-
Disease Associations in Children with Autism Spectrum Disorders in China.”
Scientific Reports 8(1): 1–9.

of
ro
-p
re
lP
na
ur
Jo

41
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

42

Figure 1: overview of phyla whose change in ASD was reported as significant in at least

one study. The rooted tree shows the phylogenetic relationships of the different phyla. In

the heatmap, colors blue and red denote direction of change (decreased and increased

respectively), whereas color intensity refers to the strength of the evidence, light meaning

p<0.05 and dark meaning p<0.01. Squares colored in grey correspond to studies finding

no significant differences between ASD and controls. Folds change relative to controls

are given when data was significant and available and control values differed from 0.

Letters R and A in the first row indicate if studies provide relative or absolute bacterial

of
levels. Abbreviations: mild=mild ASD, sib=sibling, unr=unrelated, sev=severe ASD,

ANMR=ASD with no mental regression, AMR=ASD with mental regression. Asterisks:

ro
*= Gene Ontology classification used to assign each gene to a GO term, depending on its
-p
functional characteristics, rather than calculating a relative abundance for each gene;

*1=cumulative level of Firmicutes and Proteobacteria.


re
lP
na
ur
Jo

Figure 2: overview of genera whose change in ASD was reported as significant in at least

one study. The rooted tree shows the phylogenetic relationships of the different genera.

Color ranks indicate the pertaining phylum: dark blue = Verrucomicrobia, purple =
42
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

43

Bacteroidetes, green = Proteobacteria; brown = [Thermi]; red = Armatimonadetes, light

blue = Actinobacteria, orange = Firmicutes. In the heatmap, colors blue and red denote

direction of change (decreased and increased respectively), whereas color intensity refers

to the strength of the evidence, light meaning p<0.05 and dark meaning p<0.01. Squares

colored in grey correspond to studies finding no significant differences between ASD and

controls. Folds change relative to controls are given when data was significant and

available and control values differed from 0. Letters R and A in the first row indicate if

studies provide relative or absolute bacterial levels. Abbreviations: mild=mild ASD,

of
sib=sibling, unr=unrelated, sev=severe ASD, PDD=pervasive developmental disorder,

AD=autism disorder, ANMR=ASD with no mental regression, AMR=ASD with mental

ro
regression. Asterisks: *1=cumulative level of Clostridium and Ruminococcus;
-p
*2=cumulative level of Bacteroides, Porphyromonas and Prevotella; *3=cumulative level

of Pseudomonas and Aeromonas; *4=cumulative level of Enterococcus and


re
Lactobacillus; *5=cumulative level of Pseudomonas and Aeromonas; *6= retrieved at 18

months old; *7= retrieved at 6 months old; *8= cumulative levels of Enterococcus and
lP

Lactobacillus retrieved at 18 months old; *9= quotient calculated after removal of outlier

(previous value of 83).


na
ur
Jo

43
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

44

of
ro
-p
re
lP
na
ur
Jo

44
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

45

Figure 3: overview of species whose change in ASD was reported as significant in at

least one study. The rooted tree shows the phylogenetic relationships of the different

species. Color ranks indicate the pertaining phylum: dark blue = Verrucomicrobia, yellow

= Fusobacteria, green = Proteobacteria, purple = Bacteroidetes, light blue =

Actinobacteria, orange = Firmicutes. In the heatmap, colors blue and red denote direction

of change (decreased and increased respectively), whereas color intensity refers to the

strength of the evidence, light meaning p<0.05 and dark meaning p<0.01. Squares colored

in grey correspond to studies finding no significant differences between ASD and

of
controls. Folds change relative to controls are given when data was significant and

available and control values differed from 0. Letters R and A in the first row indicate if

ro
studies provide relative or absolute bacterial levels. Abbreviations: mild=mild ASD,
-p
sib=sibling, unr=unrelated, sev=severe ASD, PDD=pervasive developmental disorder,

AD=autism disorder, FGIDs= functional gastrointestinal disorders.


re
lP
na
ur
Jo

45
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

46

of
ro
-p
re
lP
na
ur
Jo

46
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

47

of
ro
-p
re
lP
na
ur
Jo

Figure 4: Integration of the impaired gut-brain-axis pathways in ASD individuals and

mouse models.

Abbreviations:

47
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

48

BBB: blood-brain barrier; CHD8: chromodomane helicase DNA binding protein 8

gene; CNS: central nervous system; CRH: corticotropin-releasing hormone; G-

bacteria: Gram negative bacteria; G x E: interactions between genes and environmental

factors; GABA: gamma amino butyric acid; GI: gastrointestinal; HGF: hepatocyte

growth factor, ligand of the MET receptor; LPS: lipopolysaccharides; MET

rs1858830: variant of the MET gene which encodes the c-MET tyrosine-protein

kinase, also called hepatocyte growth factor receptor; poly I:C: polyinosinic-

polycytidylic acid, used to mimic maternal viral infection in mice; PPA: propionic

of
acid; SERT: serotonin transporter; SLC6A4: variant of the gene which encodes the

serotonin transporter; 5-HT: serotonin; Th1, 2, 17: T-helper cell 1, 2 and 17; VN: vagus

ro
nerve; VPA: Valproate.

Color code:
-p
re
Navy: increased permeability of barrier pathways: intestinal mucosal barrier and BBB.

Orange: anatomical pathways: the gut-brain’s neural network.


lP

Green: neuroendocrine pathway: HPA axis (colored in grey) and neurotransmitters

and neural regulators (colored in light green).


na

Light blue: immunological pathway: cytokine release.

Red: metabolic pathway: microbial metabolome.


ur

Yellow: microbial and clinical outcomes in ASD.


Jo

48
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

49

of
ro
Figure 5: Hypothalamic-pituitary-adrenal (HPA) axis. Under stress conditions, the

hypothalamus releases vasopressin and corticotropin-releasing hormone (CRH) that


-p
signal the release of adrenocorticotropic hormone (ACTH) from the pituitary gland,

which in turn influences the secretion of hormones from the adrenal glands, such as
re
cortisol. Cortisol is a glucocorticoid that affects many human organs, including the brain,
lP

where it can regulate the activity of the intestinal cells, such as the immune cells, the

epithelial cells, the enterochromaffin cells, the interstitial cells of Cajal, the enteric
na

neurons and the smooth muscle cells. These same cells are under the influence of the gut

microbiota, which, when dysbiotic, can induce an early immune activation leading to
ur

increased pro-inflammatory cytokines. As will be discussed later, cytokines gain access

to the hypothalamus throughout the impaired blood-brain barrier, acting on the HPA axis
Jo

similarly to stress.

49
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

50

of
Figure 6: Interaction pathway between bacterial and viral components and the nervous

and immune systems: subepithelial dendritic cells, one of the basic cells of the gut

ro
immune system, extend their dendrites through epithelial cells into the gut lumen and
-p
collect bacteria and their metabolites, which are then processed into proteins, nucleic

acids, sugars and lipids and carried in exosomes. This content is transferred from dendritic
re
cells to T cells in the lymph nodes, allowing exosomes to enter the systemic circulation
lP

via the lymphatic system. Once they have reached the brain by passing through the blood-

brain barrier, they bind toll-like receptors (TLRs) in neurons and induce changes in the

electrical layout of neuronal membranes which are responsible for the regulation of
na

emotion and behavior. On the upper side of the diagram, interactions between bacterial

and viral components and the immune system are represented, as they are the first step to
ur

produce cytokine response. When cytokines are spread in the bloodstream and have
Jo

gained access to the brain, they can activate the HPA axis.

50
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

51

of
ro
-p
re
lP
na
ur
Jo

51
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

Author/ yeara Sample size and ASD diagnostic and Measuring tools and sample for Measuring tools for Limitations 52
demographics categorizing tools microbial analysis GI function

GI function not studied


Song et al 2003 Late-onset AD (n=13); Not reported. Bacterial culture, genotypic (HPLC, NA 1. Small sample size.

f
NT unrelated (n=8) PCR) and phenotypic 2. Unknown sample age and sex

oo
characterization of 15 isolates on 3. Unknown exclusion criteria
stool, blood and an intra- 4. No stratification by ASD severity
abdominal abscess. 5. Number of samples per subject not specified (presumably
one).

Song et al 2004 Late-onset AD (n=13); Not reported. Clostridium quantification by qPCR NA 1. Small sample size.

pr
NT unrelated (n=8). on stool 2. Unknown sample age and sex
3. Unknown exclusion criteria
4. No stratification by ASD severity
5. Number of samples per subject not specified.

e-
Tomova et al ASD (m:f = 9:1, aged 2-9y); ICD-10, CARS, ADI qPCR on stool bacterial population NA 1. Small sample size.
2015 NT siblings (m:f = 7:2, aged 5- 2. Sex bias.
17y); 3. Not considered as exclusion criteria: concomitant intake of
NT unrelated (m:f = 10:0; 2- other medication besides antibiotics
11y). 4. Number of samples per subject not specified.

Pr
Pärtty et al 2015 Aspergers and ADHD (m:f = ICD-10 FISH and qPCR on stool bacterial NA 1. Small sample size.
6:0); population 2. Sex bias.
NT unrelated (m:f = 34:35). 3. Not considered as exclusion criteria: concomitant antibiotic
and intake of other medications, such as probiotics
(supplemented in 40 NT subjects), dietary status, concomitant
pathologies

l 4. No stratification by ASD severity


na
5. Single sample per subject.
Kantarcioglu et al ASD suspected/ diagnosed No verification of ASD Yeast culture from stool samples NA 1. Unverified diagnosis of ASD subjects
2015 (m:f = 879:676, aged 9mo- diagnosis and identification by 2. Not considered as exclusion criteria: functional foods besides
17y); morphological and biochemical kefir, dietary status, concomitant pathologies
NT unrelated (m:f = 234:169; tests. 3. No stratification by ASD severity
2-18y). 4. Number of samples per subject not specified.
ur

Ma et al 2019 ASD (m:f = 39:6, aged 6.2- DSM-V, CARS 16S rRNA gene pyrosequencing by NA 1. Small sample size.
8.34y); PCR on stool bacterial population 2. Sex bias
NT unrelated-wGI (m:f = 39:6; 3. Not considered as exclusion criteria: dietary status, but p-value
aged 5.85-8.23y) adjusted accordingly
4. Number of samples per subject not specified.
Jo

Mixed sample (not stratified by GI function)

52
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

53

f
Finegold et al ASD (m:f = 24:9 – 3 excluded); Clinical observation (no Bacterial tag-encoded FLX Presence of 1. Small sample size.
2010 NT siblings (m:f= 5:9); specific test used) amplicon pyrosequencing on stool symptoms. 2. Sex bias.

oo
NT unrelated (m:f=5:3). samples 3. Not considered as exclusion criteria: functional food intake,
Age range: 2-13y, *some with dietary status, unclear if concomitant pathologies
GI 4. Number of samples per subject not specified.
Gondalia et al AD or Asperger (m:f = 42:9, No validating assessment Bacterial tag-encoded FLX Presence of 1. Modest sample size.
2012 *28 ASD-GI); (usage of CARS scores amplicon pyrosequencing on stool symptoms. 2. Sex bias.

pr
NT siblings (m:f = 19:34, * dated up to 12 months samples 3. Not considered as exclusion criteria: concomitant medication
NTsib-GI). prior) intake (antibiotic intake only of one child on the control
Age range: 2-12y group), dietary status, food allergies
5. No unrelated NT control group
6. Number of samples per subject not specified.

e-
Iovene et al 2016 ASD (m:f=40:7; aged 6±2.8y, DSM-V-TR, ADI-R, CARS, Bacterial and yeast culture from QPGS-RIII and 1. Small sample size.
*33 ASD-GI) ADOS stool samples with identification LA/MA for intestinal 2. Sex bias.
NT unrelated-wGI (m:f=23:9; of colonies by VITEK2 system. permeability 3. Not considered as exclusion criteria: concomitant medication
aged 7.3±3.1y) intake (including antibiotics), dietary status

Pr
4. No stratification by ASD severity
5. Number of samples per subject not specified.
Lee et al 2017b ASD (m:f = 18:2, aged 17.5- Diagnosis: DSM-V Isolation and 16S rRNA Not reported 1. Small sample size.
27.3y); Characterization: K-CARS pyrosequencing of bacteria- 2. Sex bias
NT unrelated-wGI (m:f = 24:4; derived extracellular membrane 3. Not considered as exclusion criteria: concomitant antibiotic in
aged 11.6-30.6y vesicles from urine samples ASD group, other medication besides antipsychotics, dietary
status
l 4. Number of samples per subject not specified.
na
Kang et al 2018 ASD (m:f = 22:1, aged 6-14.2y, ATEC, PDD-BI 16S rRNA gene pyrosequencing by 6-GSI 1. Small sample size.
*21 ASD-GI); Genome Sequencer FLX-Titanium 2. Sex bias
NT unrelated (m:f = 15:6; System on stool bacterial 3. Not considered as exclusion criteria: functional foods, dietary
aged 5-11.8y, *10 NT-GI) population status, unclear if concomitant pathologies
4. No stratification by ASD severity
Pulikkan et al Severe ASD (m:f = 28:2, aged CARS, INDT-ASD, ISAA 16S rRNA gene sequencing (NGS) Not reported 1. Small sample size.
ur

2018b 3-16y); on stool bacterial population 2. Sex bias.


NT mostly related - wGI (m:f = 3. Not considered as exclusion criteria: functional foods and
15:9; 3.5-16y) antifungal intake
4. No unrelated NT control group
Jo

5. Single sample per subjects


Zhang et al 2018 ASD (m:f = 29:6, aged 3.4- DSM-V 16S rRNA gene sequencing (NGS) Not reported 1. Small sample size.
6.4y, *21 ASD-GI); on stool bacterial population 2. Sex bias
NT unrelated-wGI (m:f = 5:1; 3. Not considered as exclusion criteria: antifungal intake
aged 3.5-5.7y) 4. No stratification by ASD severity
5. Number of samples per subject not specified.

53
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

54

f
Coretti et al 2018 ASD (m:f = 9:2, aged 2.4-3.4y, Diagnosis: DSM-V, ADOS-2 16S rRNA and rDNA gene QPGS-RIII 1. Small sample size.
*2 ASD-GI); Characterization: ADI-R, sequencing by droplet digital PCR 2. Sex bias

oo
NT unrelated-wGI (m:f = 8:6; GMDS, VABS, CARS on stool bacterial population 3. Not considered as exclusion criteria: antifungal intake
aged 2.2-3.6y)
Plaza-Díaz 2019 ASD-MR (n=18); DSM-V, ADI-R, ICD-10, 16S rRNA gene pyrosequencing by Not reported 1. Small sample size.
ASD-wMR (n=30); ADOS, PDDBI, CARS, PCR on stool bacterial population 2. Unknown sample sex
NT unrelated (n=48; ged 3.5- Battelle developmental test 3. Not considered as exclusion criteria: concomitant medication

pr
3.9y; *2 ASD-abdominal pain) (developmental delay), 5- intake (including antibiotics), dietary status (but analysis of its
item questionnaire effect on microbial pattern).
(developmental regression) 4. Number of samples per subject not specified.

e-
AD-GI versus NT-GI
Williams et al AD-GI (m:f= 15:0, aged 3.5- DSM-IV-TR, ADI-R, 16S rRNA gene pyrosequencing by Standardized 1. Small sample size.
2011 5.9y); Shortened CPEA qPCR on bacterial population from questionnarie 2. Sex bias.
NT-GI (m:f= 7:0, aged 3.9- Regression Interview ileal and cecal biopsies. 3. Not considered as exclusion criteria: concomitant medication

Pr
5.5y); (regression status) intake (antibiotic intake only of one child on the control
group), dietary status, concomitant pathologies (but analysis
of their effect on microbial pattern)
4. No stratification by ASD severity.
Williams et al AD-GI (m:f= 15:0, aged 3.5- DSM-IV-TR, ADI-R, 16S rRNA gene pyrosequencing by Standardized 1. Small sample size.
2012 5.9y + 8m aged 6-10 for Shortened CPEA qPCR on bacterial population from questionnarie 2. Sex bias.
Suterella assessment); Regression Interview ileal and cecal biopsies. 3. Not considered as exclusion criteria: concomitant medication
l
NT-GI (m:f= 7:0, aged 3.9-5.5y (regression status) intake (antibiotic intake only of one child on the control
na
+ 2m aged 6-10 for Suterella group), dietary status, concomitant pathologies
assessment). 4. No stratification by ASD severity.

Kushak et al 2017 ASD-GI (m:f = 19:2, aged DSM-IV 16S rRNA gene pyrosequencing on Presence of 1. Small sample size.
12.7—16.1y); duodenal bacterial population symptoms. 2. Sex bias.
ur

NT-GI (m:f = 10:9, aged 14.8- 3. Not considered as exclusion criteria: concomitant medication
17.3y). intake (including antibiotics), dietary status, concomitant
pathologies
4. No stratification by ASD severity
5. Number of samples per subject not specified.
Jo

AD-wGI versus NT-wGI


De Angelis et al PDD-NOS-wGI (n=10); DSM-IV-TR, ADI-R, ADOS, Bacterial tag-encoded FLX Presence of 1. Small sample size.
2013 AD-wGI (n=10); CARS amplicon (16S rDNA and 16S symptoms. 2. No information about age and sex distribution across groups
NT siblings-wGI (n=10); rRNA) pyrosequencing on stool 3. Not considered as exclusion criteria: antifungal intake, dietary
Age range: 4-10y, m:f = 14:16 samples status
4. No stratification by ASD severity
5. No unrelated NT control group

54
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

55

f
Inoue et al 2016 ASD-wGI (n=6); DSM-V, PARS, M-CHAT 16S rRNA gene pyrosequencing by Not reported 1. Small sample size.
NT unrelated - wGI (n=6) PCR on stool bacterial population 2. Unknown sample sex

oo
Age range: 3-5y 3. No information about age and sex distribution across groups
4. Not considered as exclusion criteria: dietary status, unclear if
concomitant pathologies
5. No stratification by ASD severity
6. Number of samples per subject not specified.

pr
AD-GI versus NT-wGI
Sandler et al Late-onset AD + antecedents DSM-IV, behavioral Quantification of aerobic and Loose stool history 1. Small sample size.
2000 of antibiotic exposure questionnaire anaerobic species on stool 2. Unknown age from control group
followed by chronic diarrhea (unreported exact method). 3. Unknown sample sex
(m:f = 10:1 – 7 excluded, aged 4. Not considered as exclusion criteria: dietary status,

e-
2-8y); concomitant pathologies
NT adults n=104. 5. No stratification by ASD severity
6. Number of samples per subject not specified (presumably
one).

Pr
7. No statistical analysis between groups
Finegold et al Late-onset AD-GI n = 13 (stool Not reported (possibly not Bacterial culture, analysis of Presence of 1. Small sample size.
2002 data) and n = 7 (gastric, small validated) bacterial metabolites and cellular symptoms. 2. Unknown sample age and sex
bowel data); fatty acids, PCR of the 16S and 16S 3. Not considered as exclusion criteria: concomitant intake of
NT unrelated n = 8 and n= 4 rRNA gene sequencing on stool, other medication besides antibiotics, dietary status, unclear If
respectively. gastric and small-bowel samples. concomitant pathologies
4. No stratification by ASD severity
l 5. Number of samples per subject not specified.
na
Adams et al 2011 AD (55), Aspergers (3)-GI (m:f No validating assessment Bacterial culture, quantification 6-GSI 1. Modest sample size.
= 50:8, aged 3.5-10.3) for diagnosis (previous and identification of bacteria and 2. Sex bias.
NT unrelated-wGI (m:f = diagnosis by physician), yeast from stool samples. 3. Not considered as exclusion criteria: antifungal intake, dietary
18:21, aged 3.3-12.1) ATEC (severity) status
4. No stratification by ASD severity.
5. Single sample per subject.
ur

Carissimi et al ASD-severe develop-mental Clinical observation 16S rRNA gene sequencing (NGS) GIH 1. Small sample size.
2019 delay (m:f = 16:0, aged 2-6y, GMDS, ADOS-2 on stool bacterial population 2. Sex bias
*12 ASD-GI, *3mild, 9 3. Not considered as exclusion criteria: antifungal intake, dietary
moderate, 4 severe); status, unclear if concomitant pathologies
Jo

NT unrelated-wGI (m:f = 2:5; 4. Number of samples per subject not specified.


aged 5-16y)
AD-GI versus AD-wGI
Wang et al. 2011 AD (17), Aspergers (6) (m:f = DSM-V, CARS qPCR on stool bacterial population FGID questionnaire 1. Small sample size.
21:2, aged 3-17y, *9 ASD-GI); 2. Sex bias.
NT siblings (m:f = 11:11, aged 3. Not considered as exclusion criteria: concomitant medication
4.5-18.5y, *6 NTsib-GI); intake (including antibiotics), dietary status, concomitant

55
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

56

f
NT unrelated (m:f = 4:5, aged pathologies (epilepsy and intellectual disability included in ASD
3.5-15y, *1 NT-GI). group)

oo
1. No stratification by ASD severity.

Wang et al. 2013 AD (17), Asperger (6) (m:f = DSM-V, CARS qPCR on stool bacterial population FGID questionnaire 2. Small sample size.
21:2, aged 3-17y, *9 ASD-GI); 3. Sex bias.
NT siblings (m:f = 11:11, aged 4. Not considered as exclusion criteria: concomitant medication
4.5-18.5y, *6 NTsib-GI); intake (including antibiotics), dietary status, concomitant

pr
NT unrelated (m:f = 4:5, aged pathologies (epilepsy and intellectual disability included in ASD
3.5-15y, *1 NT-GI). group Epilepsy and intellectual disability included in ASD
group)
5. No stratification by ASD severity.

e-
AD-GI versus NT-GI versus NT-wGI
Parracho et al. ASD (m:f = 48:10, aged 3-16y, Not reported FISH analysis and microscopic Questionnaire 1. Modest sample size.
2005 *53 ASD-GI); examination of stool bacterial 2. Sex bias.
NT siblings (m:f = 7:5, aged 2- population 3. Not considered as exclusion criteria: concomitant medication

Pr
10y, *3 NTsib-GI); intake (including antibiotics), dietary status, but analysis of
NT unrelated - wGI (m:f = 6:4; their association with bacterial profiles. Unclear if concomitant
aged 3-12y) pathologies were considered.
4. No stratification by ASD severity
5. Number of samples per subject not specified.
Kang et al. 2013 AD-GI (m:f = 18:12, aged 3- ADI-R, ADOS, PDD-BI qPCR analysis of 16S rDNA stool Modified version of 1. Small sample size.
11y); bacterial population. GSI 2. Sex bias.
NT unrelated (m:f = 17:3,
l 3. Not considered as exclusion criteria: functional foods, dietary
na
aged 3-16y, *7 NT-GI). status and dietary supplements
4. Single sample per subject.
Liu et al 2019 ASD (m:f = 25:5, aged 3-5.9y, DSM-V, ICD-10 16S rRNA gene pyrosequencing by Modified versión of 1. Small sample size.
*15 ASG-GI); PCR on stool bacterial population 6-GSI 2. Sex bias
NT unrelated-wGI (m:f = 16:4; 3. No stratification by ASD severity
aged 3.3-5.3y) 4. Number of samples per subject not specified.
ur

Kong et al 2019 ASD (m:f = 15:5, aged 13- DSM-V 16S rRNA gene pyrosequencing by GSI 1. Small sample size.
18y); PCR on stool bacterial population 2. Sex bias
NT related (m:f = 8:11, aged 3. Not considered as exclusion criteria: antifungal intake, dietary
11-50y) status and allergies, but analysis of the effect of the last two
Jo

on bacterial profiles
4. No stratification by ASD severity
5. Number of samples per subject not specified.
AD-wGI versus NT-GI versus NT-wGI

56
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

57

f
Luna et al 2017c ASD-GI (m:f = 14:0, aged 4- ADOS; SRS for NT children 16S rDNA characterization from QPGS-RIII 1. Small sample size.
13y); rectal bacterial population 2. Sex bias.

oo
NT unrelated-GI (m:f = 12:3, 3. Not considered as exclusion criteria: concomitant intake of
aged 3-18y); other medication besides antibiotics and steroids, dietary
NT unrelated-wGI (m:f = 6:0; status
3-14y) 4. No stratification by ASD severity
AD-GI versus AD-wGI versus NT-GI versus NT-wGI

pr
Son et al 2015 ASD (m:f = 52:7, *25 ASD-GI); Unclear verification of qPCR and 16S rRNA gene QPGS-RIII 1. Modest sample size.
NT siblings (m:f = 21:23, *13 diagnosis, behavior pyrosequencing of stool bacterial 2. Sex bias.
NT-GI). problems assessed by population. 3. Not considered as exclusion criteria: antifungal intake, dietary
Age range: 7-14y, only 37 CBCL status, concomitant pathologies
family-matched pairs. 4. No stratification by ASD severity

e-
5. No unrelated NT control group
6. Number of samples per subject not specified.
Strati et al 2017 AD (m:f = 31:9, aged 5-17y, *5 DSM-V, ADOS, ABC, CARS 16S rRNA gene pyrosequencing on Constipation 1. Modest sample size.
constipated, *29 non-cons., bacterial and fungal stool sample defined according 2. Sex bias.

Pr
*36 severe AD); populations. to Rome III 3. Not considered as exclusion criteria: antifungal intake, dietary
NT unrelated (m:f = 28:12, status, celiac disease (present in 2 ASD subjects
aged 3.6-12y, *11 cons.). 4. Number of samples per subject not specified.
6-GSI: 6-item gastrointestinal severity index; ABC: Autism Behavior Checklist; AD: autism disorder; ADI/ ADI-R: Autism Diagnostic Interview/ Revised Version; ADOS-2: Autism Diagnostic Observation Schedules – 2;
ASD: autism spectrum disorder; ATEC: Autism Treatment Evaluation Checklist; CARS: Childhood Autism Rating Scale; CBCL: Child Behavior Checklist; DSM-IV/V/TR: Diagnostic and Statistical Manual of Mental
Disorders 4th Edition/ 5th Edition/ Text revision; FGID: functional gastrointestinal disorders; FGIDs: functional gastrointestinal disorders; FISH: fluorescence in situ hybridization; GI: gastrointestinal disorders; GIH:
CHARGE Gastrointestinal History Questionnaire; GMDS: Griffiths Mental Development Scales; HPLC: high performance liquid chromatography; ICD-10: International Statistics Classification of Diseases and Related
l
Health Problems, 10th Revision; INDT-ASD: INCLEN Diagnostic Tool for Autism Spectrum Disorder (DSM-V approved); ISAA: Indian Scale for Assessment of Autism; KCARS: Korean Childhood Autism Rating Scale;
na
LA/MA: lactulose/mannitol test; M-CHAT: Modified Check-list for Autism in Toddlers; m:f: male/female ratio; MR: mental regression; NA: not applicable; NGS: next-generation sequencing; NT: neurotypical; PARS:
Pervasive Developmental Disorders Autism Society Japan Rating Scale; PCoA: principal coordinate analysis; PCR: Polymerase Chain Reaction; PDD-NOS: pervasive developmental disorder not otherwise specified;
PDDBI: Pervasive Developmental Disorders Behavior Inventory; qPCR quantitative polymerase chain reaction, also known as real-time PCR (RT-PCR), QPGS-RIII: Questionnaire on Pediatric Gastrointestinal
Symptoms-Rome III version; SRS: Social Responsiveness Scale; VABS: Vineland Adaptive Behavior Scales; wGI: without gastrointestinal disorders; wMR: without mental regression
ur

Table 1: gut bacterial and fungal profile in children and adolescents with ASD. a Studies classified by assessment of GI function on microbial analysis and
displayed in ascending order of publication date; b Unclear whether ASD children suffer from GI conditions, c For specific analysis of correlation between
Jo

microbiota and abdominal pain, stratification on four groups was conducted: ASD-GI with no pain reported (n=5), ASD-GI with abdominal pain (n=9), NT with
no pain reported (n=10), and NT with abdominal pain (n=11).

57
Licensed to Alícia Gleides Fontes Gonçalves - aliciafontes.nutri@gmail.com

58

f
oo
pr
e-
l Pr
na
ur
Jo

58

You might also like