Sie sind auf Seite 1von 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/318165543

Metabolic Association Between the Gut–Brain Axis in Autism Spectrum


Disorders

Chapter · July 2017


DOI: 10.1007/978-3-319-53126-7_33

CITATIONS READS

0 145

4 authors, including:

Andrea Delgado Adriana Fochesato


National University of Cordoba, Argentina Fundacion J Robert Cade
11 PUBLICATIONS   42 CITATIONS    1 PUBLICATION   0 CITATIONS   

SEE PROFILE SEE PROFILE

Pascual Angel Gargiulo


Pontifical Catholic University of Argentina
28 PUBLICATIONS   20 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Both chapters are included in our Book "Neurosciences and Psychiatry. Bridging the divide." View project

INCIDENCIA DE EFECTOS ADVERSOS EN PERROS Y GATOS CON PROTOCOLO ANESTÉSICO COMPUESTO POR KETAMINA Y XILAZINA View project

All content following this page was uploaded by Andrea Delgado on 01 November 2018.

The user has requested enhancement of the downloaded file.


Metabolic Association Between
the Gut–Brain Axis in Autism
33
Spectrum Disorders

María Andrea Delgado, Adriana Fochesato,


Luis Isaías Juncos, and Pascual Ángel Gargiulo

Abstract
Autism spectrum disorder (ASD) is a severe, complex neurodevelopmental
disorder, characterized by impairments in social interaction and communi-
cation with restricted and stereotyped behavior patterns. ASD symptoms
result from a complex interaction between genetic and environment factors.
Food intolerances, allergies, altered intestinal permeability (leaky gut),
immune dysregulation, neuroinflammation and oxidative stress may trigger
ASD symptoms. ASD patients have shown increased urinary levels of
β-casomorphin and gliadorphin peptides produced by incomplete digestion
of gluten proteins and milk casein. “Leaky gut” may facilitate the transport
of these peptides into the central nervous system (CNS) inducing direct
“opioid activity” and thus affecting neurotransmission. ASD patients on
gluten and/or casein-free diet have shown improvement in most behavior
and cognitive scores. Immune dysregulation leads to a neuroinflammatory
response that correlates between immune dysfunction with behavioral and
cognitive impairments in ASD patients. Genetic variants of the MET gene
(7q31.2) are risk factors for ASD. The MET receptor participates in brain
cortex and cerebellum development and in gastrointestinal and immuno-
logical functions. A high percentage of ASD children have shown non-
celiac gluten sensitivity, an immune r­ eaction against gluten in subjects not
affected with celiac disease with prominent mucosal eosinophil infiltration
and increased blood eosinophilia. ASD patients have shown alterations in

M.A. Delgado, PhD (*) • A. Fochesato


L.I. Juncos
Fundación J Robert Cade,
Pedro de Oñate 253, Córdoba X5003DDZ, Argentina
e-mail: delgadoandrea73@gmail.com
P.Á Gargiulo
Laboratorio de Neurociencias y Psicología
Experimental, Área de Farmacología, Facultad de
Ciencias Médicas, Universidad Nacional de Cuyo,
CONICET, Mendoza, Argentina

© Springer International Publishing AG 2017 465


P.Á. Gargiulo, H.L. Mesones-Arroyo (eds.), Psychiatry and Neuroscience Update - Vol. II,
DOI 10.1007/978-3-319-53126-7_33
466 M.A. Delgado et al.

brain anatomy involved in language and social interaction skills, correlat-


ing with specific aspects of ASD symptoms. ASD behavior results from
abnormal interactions between the opioid system and various pathways
together with anatomical alterations in the CNS. Individualized diagnosis
and prognostic predictions should provide effective personalized therapies
in ASD patients.

Keywords
Autism spectrum disorders • Opioid peptides • Leaky gut • Genetics
Immune dysregulation

Introduction borderline disability (IQ = 71–85), and 46% were


in the average or higher range of intellectual abil-
Autism spectrum disorders (ASDs) (Online ity (IQ >85). In addition, there were differences
Mendelian Inheritance in Man—(OMIM)— between ethnic groups in the rate of intellectual
209850) is increasingly recognized as a systemic disability: 48% of non-Hispanic black children,
and complex disease process whose main features 38% of Hispanic children, and 25% of non-­
are displayed by a severe and complex neurodevel- Hispanic white children were classified with
opmental disorder. Indeed, this state is character- intellectual disability [2].
ized by impairment in reciprocal social interaction This recent and rapid increase in ASD preva-
and communication, and restricted and stereo- lence highlights the importance of furthering
typed patterns of interests and behaviors [1]. research, including risk factors, etiology, bio-
The latest studies, according to the Centers for markers, and effective interventions [2].
Disease Control and Prevention and Autism and ASD refers to a broader definition of autism
Developmental Disabilities Monitoring (ADDM) that includes classical and atypical autism,
Network, indicated that the prevalence was 1 in Asperger syndrome, and pervasive developmen-
68 children, aged up to 8 years from 11 commu- tal disorder [3]. Although the etiology of autism
nities within the United States, who had been is not yet understood, patients with ASD show
identified with ASD in 2012 [2]. manifestations of a systemic and complex dis-
ASD is a highly heritable disorder, with onset ease process, and symptoms persisting through-
usually after the 3rd year of age. Estimations of out life [4].
ASD prevalence diverge by sex and ethnicity. It The symptoms of autism are thought to result
affects predominantly males, with a sex ratio of from a complex and variable interaction between
approximately 4.5:1. Indeed, approximately 1 in genetic and environmental factors [5]. Food intol-
42 boys and 1 in 189 girls living in the ADDM erances or allergies, altered intestinal permeability
Network communities were identified as having (leaky gut), dietary opioid peptides in bloodstream,
ASD. All 11 sites reported higher prevalence neuroinflammation, immune dysregulation, and
among white children than among black (1.2%) oxidative stress by mitochondrial dysfunction may
or Hispanic children (1.5%), while ASD was trigger ASD symptoms [6–10]. (Fig. 33.1).
more prevalent in black children than in Hispanic Scientific evidence has confirmed the improvement
children (1.3%) [2]. The intellectual ability of of ASD symptoms after early interventions to pre-
children with ASD was classified in different pro- vent common biomedical abnormalities believed to
portions: 31% of the children fell within the lead to harmful consequences on behavior and neu-
range of intellectual disability (IQ ≤70), 23% had rological functioning [11–13].
33  Metabolic Association Between the Gut–Brain Axis in Autism Spectrum Disorders 467

Fig. 33.1 Schematic
representation of
different mechanisms Opioid
thought to be implicated Inmune activity
in the pathogenesis of dysregulation Neuroinflammation
autism

Autism
spectrum Oxidative
Leaky gut disorders stress

Food intolerancies Genetic


or alterations
allergies

Metabolic Insights in ASD


1 2 3 4 5 6 7
B-Casomorphin tyr pro phe pro gly pro lle
In 1966, Dohan postulated that schizophrenia is Gliadorphin tyr pro gln pro gln pro phe
the cause of an overload of peptides derived from
dietary gluten [14]. This hypothesis was later
extended to include autism [15, 16]. Fig. 33.2  Amino acid sequence for β-casomorphin-7 and
The normal dietary protein digestion entails gliadorphin-7 peptides. Amino acids: tyrosine (tyr), pro-
their breakdown to smaller molecules until line (pro), phenylalanine (phe), glycine (gly), isoleucine
reaching the basic unit (amino acid). Therefore, (ile), glutamine (gln)
an incomplete protein breakdown generates
bigger structural compounds, namely peptides, ASD patients are four times more likely to
composed by 2–8 amino acids. Indeed, suffer from gastrointestinal problems than the
increased urinary β-casomorphin and gliador- rest of the population [24]. This is a fact known
phin peptides have been reported in ASD since the first clinical description of autism in
patients as a result of an incomplete digestion the 1940s [25]. Moreover, there is evidence of a
of casein and gluten respectively [6]. Both of strong correlation between gastrointestinal
these peptides show structural similarities in symptoms and autism severity that could be
fragment tyrosine–proline (tyr–pro) in position explained by three different mechanisms: (1) an
1–2 and proline (pro) amino acid in positions 4 increased permeability of the intestinal mucosa
and 6 (Fig. 33.2). not related to inflammatory processes, such as
Altered intestinal permeability has been impli- tight junction defects or alterations in the intes-
cated in ASD patients, and is thought to be the tinal flora composition (gut microbiome). (2) a
link between the gut and brain in autism patho- possible second mechanism involving inade-
genesis [17–19]. Supporting this hypothesis, gas- quate or deficient enzymes for either intestinal
trointestinal disorders, such as diarrhea, chronic or blood breakdown of β-casomorphins or glia-
constipation, abdominal pain, ulcerative colitis, dorphins which allows exorphins in large
and others, have been reported in ASD patients amounts to gain access to the blood and circu-
[20–23]. late until taken up by the brain, (3) a possible
468 M.A. Delgado et al.

a b c

Gut

Blood

Brain

Fig. 33.3  Passage of opioid peptides through gut, blood, tions (gut pH alterations or enzyme system deficiencies)
and central nervous system (CNS). (a) Normal gut–brain may show increased intestinal peptide levels that will go
transport of peptides. In healthy subjects, with no major through the bloodstream and later the BBB reaching the
problems with intestinal wall permeability, a portion of brain. (c) Excessive permeability of the intestinal mem-
peptides (green image) normally crosses from the intes- brane. Subjects with normal gut peptide levels with a flaw
tine into the bloodstream. A portion of these peptides in intestinal membrane permeability. This situation allows
bloodstream will cross through the blood–brain barrier greater amounts of biologically active peptides to enter
(BBB), accessing the brain. (b) Excessive peptide produc- the brain through the bloodstream
tion in the gut. Subjects with abnormal intestinal condi-

third mechanism involving an inflammatory autistic children within 3 months in most of the
process causing increased intestinal wall perme- behavior categories [6]. Other researchers have
ability [6, 17, 18, 26] (Fig. 33.3). shown similar results [34–39].
Both peptides, β-casomorphin and gliador-
phin, induce direct “opioid activity” [27, 28]. The
“opioid excess” theory suggests that ASD symp- Genetic Insights in ASD
toms are a consequence of the action of exoge-
nous peptides affecting neurotransmission within Direct Brain Impacts  Genetic heterogeneity and
the central nervous system (CNS) [15, 29–32]. gene–environment interactions have been found
In the absence of an established pathogenesis in patients with ASD. Many ASD risk genes
for autism, some authors have proposed explana- encode regulatory proteins of the glutamatergic
tions and treatments based on diet and nutrients. system involved in the synaptic process and
Reports on the use of a gluten-and casein-free excitatory/inhibitory brain networks, namely:
diet have shown improvement of ASD symp- NRXN1, PTEN, SHANK3, UBE3a, NF1,
toms, such as social isolation, and overall ability NLGN3/4, CNTNAP2, SYNGAP1, and FMR1
to communicate and interact [33]. Moreover, [40–48]. Mutations in these genes during CNS
Cade et al. showed that a gluten- and casein-free development trigger disruptions in excitatory/
diet was accompanied by improvement in 81% of inhibitory brain circuits [45, 47, 48].
33  Metabolic Association Between the Gut–Brain Axis in Autism Spectrum Disorders 469

Actually, before synaptogenesis and brain net- stimulate disrupted behavioral and cognitive
work formation, critical developmental events functions in ASD [59].
such as neurogenesis, migration, cellular differ- Interestingly, MET also contributes to gastro-
entiation, and polarization take place [49, 50]. intestinal and immunological functions [71–74].
Therefore, alterations in genes involved in these Alterations in both systems frequently co-occur
early stages of CNS development may contribute in ASD patients [18, 19, 22, 60].
to a serious damage in brain function, particu- Several studies have found a relationship
larly in relation to genes responsible for posterior between MET promoter variants and the risk
synaptic function [47, 51]. for developing ASD. Furthermore, functional
The International Molecular Genetic Study of imaging and animal studies tie alterations in
Autism Consortium (IMGSAC) was the first to MET gene to morphological and functional
identify a genome linkage with the autism sus- disruptions in brain regions related with ASD
ceptibility locus1 (AUTS1) on the chromosome [59, 75].
7q region [52, 53]. Moreover, evidence of genetic MET gene polymorphisms “rs1858830” and
association with autism has been reported for “rs38845” have frequently been found in indi-
genes located in three different regions of chro- viduals with ASD in Europe and USA, and may
mosome 7q; 7q21 (RELN, SERPINE1), 7q31 contribute to ASD susceptibility [63, 76]. MET
(MET), and 7q35–36 (CNTNAP2, EN2). Each of gene promoter “rs1858830” is a common gua-
these chromosome 7q genes, and possibly others, nine (G) to cytosine (C) single nucleotide poly-
may contribute to autism risk [43, 54–56]. morphism, with significant association in ASD
The human gene MET (proto-oncogene hepa- families reported in Italian and USA cohorts.
tocyte growth factor receptor, OMIM 164860) The “C allele” is more common in individuals
that is located on chromosome 7q31.2 and covers with ASD than in the general population [59].
approximately 126 kb, encodes a high-affinity Expression studies have shown that “C allele”
transmembrane receptor tyrosine kinase that results in decreased MET promoter activity, thus
binds to the hepatocyte growth factor (HGF) [57, reducing transcription of the MET gene as well
58]. Protein receptor tyrosine kinases (RTKs) are as the specific transcription factor complexes
cell-membrane receptors that participate in key binding “SP1” [60]. The functional ASD risk
stages of CNS development, such as neurogene- variant rs1858830 (“C allele”), which reduces
sis, neuronal differentiation, migration, connec- MET protein expression, specifically impacts on
tivity, and plasticity [59]. the network of connections of different areas of
Different studies have reported this gene as a the brain involved in social behavior, including
risk factor for ASD based on genetic variants recognizing emotions shown on people’s faces
overrepresented in individuals with ASD com- [75]. Moreover, rs1858830 has shown dimin-
pared with control populations [60–64]. ished functional and structural connectivity in
Furthermore, the expression of the MET temporo-­parietal lobes brain areas that present a
receptor and HGF protein was reduced in post-­ large MET expression, which leads to decreased
mortem brains of individuals with ASD [65]. MET protein in the brains of individuals with
MET and HGF have been involved in neuronal ASD [59, 75].
development and maturation of functional cir- Sousa et al. described an association of another
cuits, particularly in cerebral cortex, cerebel- polymorphism located in intron 1 of MET gene
lum, and hippocampus [59, 66–68]. With dis- “rs38845” with a potential risk of autism suscep-
tinct spatial and temporal profiles, these brain tibility [63]. These authors suggested that
regions may be altered in autism [69, 70]. rs38845 “A allele” may regulate gene expression
Cellular signaling through the MET receptor interfering in the transcription of MET gene by a
contributes to neuronal migration and synapto- transcriptional activator “IRF1”. Thereby, IRF1
genesis, among other developmental processes. would bind to MET promoter, thus altering MET/
Therefore, alterations in MET/HGF would HGF signaling in ASD patients [63].
470 M.A. Delgado et al.

Gastrointestinal Impacts  As stated before, high symptoms of the disorder and enhance overall
rates of gastrointestinal conditions in individuals functioning of affected individuals [85, 86].
with ASD have been reported in several studies
[77, 78]. MET gene polymorphism rs1858830 “C
allele” has been associated with ASD patients  ssociation Between Allergies
A
with gastrointestinal dysfunctions. Moreover, the and ASD
transmission of the “C allele” might be enhanced
in families with co-occurring ASD and gastroin- Food intolerances and allergic diseases are com-
testinal conditions [60]. mon presentations in ASD patients [87–89].
MET signaling system alteration can lead to ASD shares with sufferers from IgE and non-­
brain and gastrointestinal dysfunctions, and IgE-­mediated allergic reactions some neuro-­
would explain the pathophysiology of ASD and psychiatric symptoms and mood disorders, such
GI comorbidities [60]. as anxiety, hyperactivity, irritability, tics, sleep
disturbance, incoordination, and learning dis-
abilities that ameliorate after anti-allergic treat-
Immune Dysregulation ments. Allergic diseases are associated with pain
and Neuroinflammation in ASD and discomfort that exacerbate behavioral symp-
toms in ASD patients [90, 91].
There is strong evidence of an association Idiopathic ASD patients frequently show
between immune dysfunction with ASD develop- increased mast cell activation in many body
ment and behavioral symptoms severity, because organs, or “mastocytosis” [92]. Mast cell stimu-
there are critical interactions between CNS and lation may be triggered by stress, environment,
the immune system [79, 80]. Patients with ASD immune reaction, and toxics impacting on brain
often exhibit alterations in cytokine levels, lym- areas associated with behavior and language
phocytes T helper type 2 (Th2), abnormal [93]. Mast cells release pro-inflammatory mole-
immune cell function, mast cell activation, and cules and histamine that stimulate the hypothala-
the presence of autoantibodies, supporting the mus–pituitary–adrenal and sympathetic axes,
association between chronic inflammation and influencing behavior and cognition [94–96].
immune dysregulation [7, 79]. Allergic diseases potentially impact on behav-
General alterations of immune mechanisms ioral symptoms and cognitive activity in ASD
trigger chronic inflammation and immune dys- children. Thus, treatment of allergies may
regulation in CNS, leading to a neuroinflamma- improve symptoms such as anxiety, hyperactiv-
tory response [79–81]. ity, and irritability, ameliorating ASD behavior
Several in-vivo and postmortem studies have [20, 97].
found chronic inflammatory processes in multi-
ple areas of the brain, highlighting the correlation
between immune dysfunction with behavioral  on-celiac Food Sensitivity
N
and cognitive impairments in ASD patients [8, and ASD
82, 83]. Moreover, some studies have shown
immune system dysregulation during a critical Non-celiac gluten sensitivity (NCGS) is a het-
period of development in children with ASD erogeneous condition regarded as a distinct
[79]. These findings suggested that inflammation clinical entity, unrelated to celiac disease, and
plays an important role in the pathogenesis of whose symptoms are triggered by gluten inges-
ASD, and lead to research to elucidate innate tion in the absence of celiac-specific antibodies
immunity pathways associated with neuronal and of classical celiac villous atrophy [98, 99].
activity in ASD [82, 84]. NCGS is an immune reaction against gluten in
The improvement of immunological altera- subjects not affected with celiac disease (CD)
tions in patients with ASD can alleviate some or wheat allergy (WA). It does not involve
33  Metabolic Association Between the Gut–Brain Axis in Autism Spectrum Disorders 471

autoimmune m ­ echanisms. In fact, this entity is correlate with specific aspects of ASD symptoms
characterized by intestinal and extra-intestinal [107–110].
symptoms triggered by the ingestion of gluten- Opioid-active peptides have shown high affin-
containing food [99, 100]. ity for the same brain tissues as other related
There is strong evidence of comorbidity in compounds, such as endorphins or morphine
ASD with intestinal pathology, and susceptibility [110, 111]. Thus, opioid peptides may cause
to suffer from allergies [101, 102]. IgA antibod- excitatory and inhibitory actions in CNS, modu-
ies to gluten and casein were found in ASD lating presynaptic release of neurotransmitters.
patients, suggesting a possible mechanism for the This is associated with the excitatory/inhibitory
increased intestinal permeability and for some of imbalance hypothesis (neuronal homeostasis) in
the symptoms in ASD patients. The presence of ASD [112]. These peptides are involved directly
IgG antibodies to gluten and casein is another or indirectly, firing neural action potentials, and/
indication that the foreign peptides reach the or releasing intracellular calcium [113–116].
blood stream intact and in sufficient amounts to Opioid peptides can inhibit glutamatergic (excit-
stimulate a vigorous IgG response, and are there- atory) signaling at spinal and supraspinal level, or
fore available for uptake by the brain [6, 103]. disinhibit GABAergic signaling (inhibition)
Several studies have described clinical, serologi- [116]. Opioid-receptor binding is involved in cel-
cal, and histological characteristics with eosino- lular proliferation, migration, and differentiation
phil infiltration in the duodenal and colon mucosa during CNS development [117, 118].
in NCGS [103]. Prominent mucosal eosinophil There are three main classes of opioid recep-
infiltration and increased prevalence of blood tors—mu, delta, and kappa—which belong to the
eosinophilia have been found in a high percent- G protein-coupled family. These opioid receptors
age of children with ASD. These features are exhibit different functions and binding character-
improved in children following a gluten-free diet istics. A specific opioid peptide is able to bind
[104, 105]. with more than one type of receptor [119, 120].
Identification and specific medical treatment The binding of opioid peptides to these receptors
of GI pathologies in patients with ASD could leads to analgesia and euphoric response [119].
diminish discomfort, thereby improving behavior Beta casomorphine-7 (BCM-7) interacts with
problems [105]. opioid receptors and affects brain regions includ-
ing the nucleus accumbens, caudate–putamen,
ventral tegmental and median raphe nucleus, and
 euroanatomy and Brain
N orbitofrontal, prefrontal, parietal, temporal,
Chemistry in ASD occipital, and entorhinal cortex. Most of these
brain areas have been found to be altered in
Magnetic resonance images and brain scan stud- ASD. Some of these brain regions are integrated
ies in ASD patients have shown alterations in by dopaminergic, serotoninergic, and GABA-­
brain anatomy and white matter connections in ergic systems, suggesting that BCM-7 may inter-
the frontal lobe, the arcuate bundle, temporal cor- fere with all of these pathways. BCM-7 may
tices, amygdala, and the anterior cingulate cor- disturb cortical association or functional connec-
tex. Moreover, post-mortem studies of ASD tions, and could be implicated in emotions and
patient’s brains have shown abnormal number of motivated behavior, social adaptation, hallucina-
Purkinje cells in cerebellum, as well as dendritic tions, and delusions, typical manifestations of
arborization in hippocampus [106]. These brain patients with ASD [28, 112, 121].
regions are involved in language and social inter- In brief, ASD behavior results from abnormal
action skills, attention control, empathy, motiva- interactions between the endogenous opioid sys-
tion, emotions, error-monitoring, pain perception, tem and various pathways, together with anatom-
behavioral adaptation to a changing environment, ical alterations in the CNS. Biochemical,
and consciousness dissociation. All these facts molecular, neurophysiological, and n­ euroimaging
472 M.A. Delgado et al.

studies should provide further insights into the 11. Pellicano E. Do autistic symptoms persist across
time? Evidence of substantial change in symptom-
pathogenesis of ASD. Due to etiologic and phe-
atology over a 3-year period in cognitively able chil-
notypic heterogeneity of ASD, individualized dren with autism. Am J Intellect Dev Disabil.
diagnosis and prognostic predictions should pro- 2012;17(2):156–66.
vide effective personalized therapies in ASD 12. Fein D, Barton M, Eigsti IM, Kelley E, Naigles L,
Schultz RT, Stevens M, Helt M, Orinstein A,
patients [109, 122, 123].
Rosenthal M, Troyb E, Tyson K. Optimal outcome in
individuals with a history of autism. J Child Psychol
Psychiatry. 2013;54(2):195–205.
References 13. Eriksson MA, Westerlund J, Hedvall Å, Åmark P,
Gillberg C, Fernell E. Medical conditions affect the
outcome of early intervention in preschool children
1. Lord C, Risi S, Lambrecht L, Cook Jr EH, Leventhal
with autism spectrum disorders. Eur Child Adolesc
BL, DiLavore PC, Pickles A, Rutter M. The autism
Psychiatry. 2013;22(1):23–33.
diagnostic observation schedule-generic: a standard
14. Dohan FC. Cereals and schizophrenia data and
measure of social and communication deficits asso-
hypothesis. Acta Psychiatr Scand. 1966;42:125–52.
ciated with the spectrum of autism. J Autism Dev
15. Reichelt KL, Hole K, Hamberger A, Saelid G,
Disord. 2000;30(3):205–23.
Edmminson PD, Braestrup CB, Lingjaerde O, Ledall
2. Christensen DL, Baio J, Van Naarden BK, Bilder D,
P, Orbeck H. Biological active peptide containing
Charles J, Constantino JN, Daniels J, Durkin MS,
fractions in schizophrenia and childhood autism.
Fitzgerald RT, Kurzius-Spencer M, Lee LC, Pettygrove S,
Adv Biochem Psychopharmacol. 1981;28:627–43.
Robinson C, Schulz E, Wells C, Wingate MS, Zahorodny
16. Travé Rodriguez AL, Barreiro Marin P, Gálvez
W, Yeargin-Allsopp M. Centers for disease control and pre-
Borrero IM, del Olmo R-NF, Díaz ÁA. Association
vention (CDC). Prevalence and characteristics of autism
between autism and schizophrenia. J Nerv Ment Dis.
spectrum disorder among children aged 8 years-­Autism
1994;182(8):478–9.
and Developmental isabilities onitoring Network, 11 sites,
17. Gardner MLG. Absorption of intact proteins and
United States, 2012. MMWR Surveill Summ. 2016;65(3):
peptides. In: Johnson LR, editor. Physiology of the
1–23.
gastro-intestinal tract. New York: Raven Press; 1994.
3. Volkmar F, Chawarska K, Klin A. Autism in infancy and
p. 1795–820.
early childhood. Annu Rev Psychol. 2005;56:315–36.
18. D’Eufemia P, Celli M, Finocchiaro R, Pacifico L,
4. Bailey A, Le Couteur A, Gottesman I, Bolton P,
Viozzi L, Zaccagnini M, Cardi E, Giardini
Simonoff E, Yuzda E, Rutter M. Autism as a strongly
O. Abnormal intestinal permeability in children with
genetic disorder: evidence from a British twin study.
autism. Acta Paediatr. 1996;85:1076–9.
Psychol Med. 1995;25:63–77.
19. de Magistris L, Familiari V, Pascotto A, Sapone A,
5. Grafodatskaya D, Chung B, Szatmari P, Weksberg
Frolli A, Iardino P, Carteni M, De Rosa M,
R. Autism spectrum disorders and epigenetics. J Am
Francavilla R, Riegler G, Militerni R, Bravaccio
Acad Child Adolesc Psychiatry. 2010;49:794–809.
C. Alterations of the intestinal barrier in patients
6. Cade R, Private FM, Rowland N, Sun Z, Zele V,
with autism spectrum disorders and in their first-­
Wagemaker H, Edelstein C. Autism and schizophrenia:
degree relatives. J Pediatr Gastroenterol Nutr.
intestinal disorders. Nutr Neurosci. 2000;3:57–72.
2010;51(4):418–24.
7. Molloy CA, Morrow AL, Meinzen-Derr J, Schleifer
20. Goodwin MS, Cowen MA, Goodwin TC. Malabsorption
K, Dienger K, Manning-Courtney P, Altaye M,
and cerebral dysfunction. A multivariate and compara-
Wills-Karp M. Elevated cytokine levels in children
tive study of autistic children. J Autism Child Schizophr.
with autism spectrum disorder. J Neuroimmunol.
1971;1:48–62.
2006;172(1):198–205.
21. Coleman M. Calcium studies and their relationship
8. Morgan JT, Chana G, Pardo CA, Achim C,
to coeliac disease in autistic patients. In: Colema M,
Semendeferi K, Buckwalter J, Courchesne E, Everall
editor. The autistic syndromes. Amsterdam: North
IP. Microglial activation and increased microglial
Holland Publishing Corp; 1976. p. 197–205.
density observed in the dorsolateral prefrontal cortex
22. Buie T, Fuchs 3rd GJ, Furuta GT, Kooros K, Levy J,
in autism. Biol Psychiatry. 2010;68(4):368–76.
Lewis JD, Wershil BK, Winter H. Recommendations
9. Williams BL, Hornig M, Parekh T, Lipkin
for evaluation and treatment of common gastrointes-
WI. Application of novel PCR based methods for
tinal problems in children with ASDs. Pediatrics.
detection, quantitation, and phylogenetic character-
2010;125(Suppl 1):S19–29.
ization of Sutterella species in intestinal biopsy sam-
23. Gorrindo P, Williams KC, Lee EB, Walker LS,
ples from children with autism and gastrointestinal
McGrew SG, Levitt P. Gastrointestinal dysfunction
disturbances. MBio. 2012;3(1):e00261–11.
in autism: parental report, clinical evaluation, and
10. Chauhan A, Audhya T, Chauhan V. Brain region-­
associated factors. Autism Res. 2012;5(2):101–8.
specific glutathione redox imbalance in autism.
24. McElhanon BO, McCracken C, Karpen S, Sharp
Neurochem Res. 2012;37(8):1681–9.
WG. Gastrointestinal symptoms in autism spectrum
33  Metabolic Association Between the Gut–Brain Axis in Autism Spectrum Disorders 473

disorder: a meta-analysis. Pediatrics. 2014;133(5):​ CL, Ledbetter DH, Nelson SF, Cantor RM,
872–83. Geschwind DH. Linkage, association, and gene-­
25. Kanner L. Follow-up study of eleven autistic chil- expression analyses identify CNTNAP2 as an
dren originally reported in 1943. J Autism Child autism-susceptibility gene. Am J Hum Genet.
Schizophr. 1971;1(2):119–45. 2008;82(1):150–9.
26. Adams JB, Audhya T, McDonough-Means S, Rubin 41. Clement JP, Aceti M, Creson TK, Ozkan ED, Shi Y,
RA, Quig D, Geis E, Gehn E, Loresto M, Mitchell J, Reish NJ, Almonte AG, Miller BH, Wiltgen BJ,
Atwood S, Barnhouse S, Lee W. Effect of a vitamin/ Miller CA, Xu X, Rumbaugh G. Pathogenic
mineral supplement on children and adults with SYNGAP1 mutations impair cognitive development
autism. BMC Pediatr. 2011;11:111. by disrupting maturation of dendritic spine synapses.
27. Sun Z, Cade JR, Fregly MJ, Privette RM. Cell. 2012;151(4):709–23.
β-Casomorphin induces fos-like immunoreactivity 42. Durand CM, Betancur C, Boeckers TM, Bockmann
in discrete brain regions relevant to schizophrenia J, Chaste P, Fauchereau F, Nygren G, Rastam M,
and autism. Autism. 1999;3(1):67–83. Gillberg IC, Anckarsäter H, Sponheim E, Goubran-­
28. Sun Z, Cade R. A peptide found in schizophrenia Botros H, Delorme R, Chabane N, Mouren-Simeoni
and autism causes behavioral changes in rats. MC, de Mas P, Bieth E, Rogé B, Héron D, Burglen
Autism. 1999;3(1):85–95. L, Gillberg C, Leboyer M, Bourgeron T. Mutations
29. Panksepp J. A neurochemical theory of autism. in the gene encoding the synaptic scaffolding protein
Trends Neurosci. 1979;2:174–7. SHANK3 are associated with autism spectrum dis-
30. Reichelt KL, Knivsberg AM, Nodland M, Lind orders. Nat Genet. 2007;39(1):25–7.
G. Nature and consequence of hyperpeptiduria and 43. Peñagarikano O, Abrahams BS, Herman EI, Winden
bovine casomorphine found in autistic Syndromes. KD, Gdalyahu A, Dong H, Sonnenblick LI, Gruver R,
Brain Dysfunction. 1994;7:71–85. Almajano J, Bragin A, Golshani P, Trachtenberg JT,
31. Reichelt KL, Knivsberg AM. Can the pathophysiology Peles E, Geschwind DH. Absence of CNTNAP2 leads
of autism be explained by the nature of the discovered to epilepsy, neuronal migration abnormalities, and core
urine peptides? Nutr Neurosci. 2003;6:19–28. autism-related deficits. Cell. 2011;147(1):235–46.
32. Reichelt KL, Tveiten D, Knivsberg AM, Brønstad 44. Piggot J, Shirinyan D, Shemmassian S, Vazirian S,
G. Peptides: role in autism with emphasis on exor- Alarcón M. Neural systems approaches to the neuro-
phins. Microb Ecol Health Dis. 2012;24:23. genetics of autism spectrum disorders. Neuroscience.
33. Millward C, Ferriter M, Calver S, Connell-Jones 2009;164(1):247–56.
G. Gluten- and casein-free diets for autistic spectrum 45. Tabuchi K, Blundell J, Etherton MR, Hammer RE,
disorder. Cochrane Database Syst Rev. 2008;16(2):​ Liu X, Powell CM, Südhof TC. A neuroligin-3 muta-
CD003498. tion implicated in autism increases inhibitory synap-
34. Reichelt KL, Ekrem J, Scott H. Gluten, milk proteins tic transmission in mice. Science. 2007;318(5847):​
and autism: dietary intervention effects on behavior 71–6.
and peptide secretion. J Appl Nutr. 1990;42(1):​1–11. 46. Yashiro K, Riday TT, Condon KH, Roberts AC,
35. Lucarelli S, Frediani T, Zingoni AM, Ferruzzi F, Bernardo DR, Prakash R, Weinberg RJ, Ehlers MD,
Giardini O, Quintieri F, Barbato M, D’Eufemia P, Philpot BD. Ube3a is required for experience-­
Cardi E. Food allergy and infantile autism. dependent maturation of the neocortex. Nat
Panminerva Med. 1995;37(3):137–41. Neurosci. 2009;12(6):777–83.
36. Whiteley P, Rodgersa J. Gluten-free diet as an inter- 47. Ebert DH, Greenberg ME. Activity-dependent neu-
vention for autism and associated spectrum disor- ronal signalling and autism spectrum disorder.
ders: preliminary findings. Autism. 1999;3(1):45. Nature. 2013;493(7432):327–37.
37. Whiteley P, Shattock P. Biochemical aspects in 48. Rubenstein JL, Merzenich MM. Model of autism:
autism spectrum disorders: updating the opioid-­ increased ratio of excitation/inhibition in key neural
excess theory and presenting new opportunities for systems. Genes Brain Behav. 2003;2(5):255–67.
biomedical intervention. Expert Opin Ther Targets. 49. Bradke F, Dotti CG. Establishment of neuronal
2002;6(2):175–83. polarity: lessons from cultured hippocampal neu-
38. Knivsberg AM, Reichelt KL, Høien T, Nødland rons. Curr Opin Neurobiol. 2000;10(5):574–81.
M. A randomised, controlled study of dietary inter- 50. Mueller BK. Growth cone guidance: first steps
vention in autistic syndromes. Nutr Neurosci. towards a deeper understanding. Annu Rev Neurosci.
2002;5(4):251–61. 1999;22:351–88.
39. Pedersen L, Parlar S, Kvist K, Whiteley P, Shattock 51. Qui S, Aldinger KA, Levitt P. Modeling of autism
P. Data mining the ScanBrit study of a gluten- and genetic variations in mice: focusing on synaptic and
casein-free dietary intervention for children with microcircuit dysfunctions. Dev Neurosci. 2012;​
autism spectrum disorders: behavioural and psycho- 34(2–3):88–100.
metric measures of dietary response. Nutr Neurosci. 52. IMGSAC. A full genome screen for autism with evi-
2014;17(5):207–13. dence for linkage to a region on chromosome 7q.
40. Alarcón M, Abrahams BS, Stone JL, Duvall JA, International molecular genetic study of autism con-
Perederiy JV, Bomar JM, Sebat J, Wigler M, Martin sortium. Hum Mol Genet. 1998;7(3):571–8.
474 M.A. Delgado et al.

53. IMGSAC. Further characterization of the autism 66. Birchmeier C, Gherardi E. Developmental roles of
susceptibility locus AUTS1 on chromosome 7q. HGF/SF and its receptor, the c-Met tyrosine kinase.
Hum Mol Genet. 2001;10(9):973–82. Trends Cell Biol. 1998;8:404–10.
54. Zhang H, Liu X, Zhang C, Mundo E, Macciardi F, 67. Yang XM, Park M. Expression of the MET/hepato-
Grayson DR, Guidotti AR, Holden JJ. Reelin gene cyte growth factor/scatter factor receptor and its
alleles and susceptibility to autism spectrum disor- ligand during differentiation of murine P19 embryo-
ders. Mol Psychiatry. 2002;7:1012–7. nal carcinoma cells. Dev Biol. 1993;157:308–20.
55. Yonan AL, Alarcón M, Cheng R, Magnusson PKE, 68. Leraci A, Forni PE, Ponzetto C. Viable hypomorphic
Spence SJ, Palmer AA, Grunn A, Juo SHH, signaling mutant of the Met receptor reveals a role
Terwilliger JD, Liu J, Cantor RM, Geschwind DH, for hepatocyte growth factor in postnatal cerebellar
Gilliam TC. A genomewide screen of 345 families development. Proc Natl Acad Sci U S A. 2002;99:​
for autism-susceptibility loci. Am J Hum Genet. 15200–5.
2003;73(4):886–97. 69. Judson MC, Bergman MY, Campbell DB, Eagleson
56. Persico AM, Napolioni V. Autism genetics. Behav KL, Levitt P. Dynamic gene and protein expression
Brain Res. 2013;15:95–112. patterns of the autism-associated met receptor tyro-
57. Cooper CS, Park M, Blair DG, Tainsky MA, Huebner sine kinase in the developing mouse forebrain.
K, Croce CM, Vande Woude GF. Molecular cloning J Comp Neurol. 2009;513(5):511–31.
of a new transforming gene from a chemically trans- 70. Judson MC, Eagleson KL, Levitt P. A new synaptic
formed human cell line. Nature. 1984;311(5981):​ player leading to autism risk: met receptor tyrosine
29–33. kinase. J Neurodev Disord. 2011;3(3):282–92.
58. Naldini L, Weidner KM, Vigna E, Gaudino G, 71. Beilmann M, Odenthal M, Jung W, Vande Woude
Bardelli A, Ponzetto C, Narsimhan RP, Hartmann G, GF, Dienes HP, Schirmacher P. Neoexpression of the
Zarnegar R, Michalopoulos GK, Birchmeierl W, c-met/hepatocyte growth factor-scatter factor recep-
Comoglio PM. Scatter factor and hepatocyte growth tor gene in activated monocytes. Blood. 1997;90(11):​
factor are indistinguishable ligands for the MET 4450–8.
receptor. EMBO J. 1991;10(10):2867–78. 72. Tahara Y, Ido A, Yamamoto S, Miyata Y, Uto H, Hori
59. Peng Y, Huentelman M, Smith C, Qiu S. MET recep- T, Hayashi K, Tsubouchi H. Hepatocyte growth fac-
tor tyrosine kinase as an autism genetic risk factor. tor facilitates colonic mucosal repair in experimental
Int Rev Neurobiol. 2013;113:135–65. ulcerative colitis in rats. J Pharmacol Exp Ther.
60. Campbell DB, Buie TM, Winter H, Bauman M, 2003;307(1):146–15.
Sutcliffe JS, Perrin JM, Levitt P. Distinct genetic risk 73. Ido A, Numata M, Kodama M, Tsubouchi H. Mucosal
based on association of MET in families with co-­ repair and growth factors: recombinant human hepa-
occurring autism and gastrointestinal conditions. tocyte growth factor as an innovative therapy for
Pediatrics. 2009;123(3):1018–24. inflammatory bowel disease. J Gastroenterol.
61. Campbell DB. When linkage signal for autism MET 2005;40(10):925–31.
candidate gene. Eur J Hum Genet. 2009;17(6):​ 74. Okunishi K, Dohi M, Nakagome K, Tanaka R, Mizuno
699–700. S, Matsumoto K, Miyazaki J, Nakamura T, Yamamoto
62. Jackson PB, Boccuto L, Skinner C, Collins JS, Neri K. A novel role of hepatocyte growth factor as an
G, Gurrieri F, Schwartz CE. Further evidence that immune regulator through suppressing dendritic cell
the rs1858830 C variant in the promoter region of function. J Immunol. 2005;175(7):​4745–53.
the MET gene is associated with autistic disorder. 75. Rudie JD, Hernandez LM, Brown JA, Beck-Pancer
Autism Res. 2009;2(4):232–6. D, Colich NL, Gorrindo P, Thompson PM,
63. Sousa I, Clark TG, Toma C, Kobayashi K, Choma Geschwind DH, Bookheimer SY, Levitt P, Dapretto
M, Holt R, Sykes NH, Lamb JA, Bailey AJ, Battaglia M. Autism-associated promoter variant in MET
A, Maestrini E, Monaco AP. MET and autism sus- impacts functional and structural brain networks.
ceptibility: family and case-control studies. Eur Neuron. 2012;75(5):904–15.
J Hum Genet. 2009;17(6):749–58. 76. Campbell DB, Sutcliffe JS, Ebert PJ, Militerni R,
64. Thanseem I, Nakamura K, Miyachi T, Toyota T, Bravaccio C, Trillo S, Elia M, Schneider C, Melmed
Yamada S, Tsujii M, Tsuchiya KJ, Anitha A, R, Sacco R, Persico AM, Levitt P. A genetic variant
Iwayama Y, Yamada K, Hattori E, Matsuzaki H, that disrupts MET transcription is associated with
Matsumoto K, Iwata Y, Suzuki K, Suda S, Kawai M, autism. Proc Natl Acad Sci U S A. 2006;​
Sugihara G, Takebayashi K, Takei N, Ichikawa H, 103(45):16834–9.
Sugiyama T, Yoshikawa T, Mori N. Further evidence 77. Quigley EM, Hurley D. Autism and the gastrointes-
for the role of MET in autism susceptibility. Neurosci tinal tract. Am J Gastroenterol. 2000;95(9):2154–6.
Res. 2010;68(2):137–41. 78. Levy SE, Souders MC, Ittenbach RF, Giarelli E,
65. Campbell DB, D’Oronzio R, Garbett K, Ebert PJ, Mulberg AE, Pinto-Martin JA. Relationship of
Mirnics K, Levitt P. Persico. Disruption of cerebral dietary intake to gastrointestinal symptoms in chil-
cortex MET signaling in autism spectrum disorder. dren with autistic spectrum disorders. Biol
AM. Ann Neurol. 2007;62(3):243–50. Psychiatry. 2007;61(4):492–7.
33  Metabolic Association Between the Gut–Brain Axis in Autism Spectrum Disorders 475

79. Heuer L, Ashwood P, Schauer J, Goines P, Krakowiak 92. Angelidou A, Alysandratos KD, Asadi S, Zhang B,
P, Hertz-Picciotto I, Hansen R, Croen LA, Pessah Francis K, Vasiadi M, Kalogeromitros D, Theoharides
IN, Van de Water J. Reduced levels of immunoglobu- TC. Brief report: “allergic symptoms” in children
lin in children with autism correlates with behavioral with autism spectrum disorders. More than meets the
symptoms. Autism Res. 2008;1(5):275–83. eye? J Autism Dev Disord. 2011;41(11):1579–85.
80. Enstrom A, Krakowiak P, Onore C, Pessah IN, 93. Theoharides TC, Enakuaa S, Sismanopoulos N,
Hertz-Picciotto I, Hansen RL, Van de Water JA, Asadi S, Papadimas EC, Angelidou A, Alysandratos
Ashwood P. Increased IgG4 levels in children with KD. Contribution of stress to asthma worsening
autism disorder. Brain Behav Immun. 2009;23(3):​ through mast cell activation. Ann Allergy Asthma
389–95. Immunol. 2012;109(1):14–9.
81. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen 94. Scaccianoce S, Lombardo K, Nicolai R, Affricano
R, Pessah I, Van de Water J. Elevated plasma cyto- D, Angelucci L. Studies on the involvement of hista-
kines in autism spectrum disorders provide evidence mine in the hypothalamic–pituitary–adrenal axis
of immune dysfunction and are associated with activation induced by nerve growth factor. Life Sci.
impaired behavioral outcome. Brain, Behav Immu. 2000;67(26):3143–52.
2011;25(1):40–5. 95. Kalogeromitros D, Syrigou EK, Makris M,
82. Li X, Chauhan A, Sheikh AM, Patil S, Chauhan V, Li Kempuraj D, Stavrianeas NG, Vasiadi M,
XM, Ji L, Brown T, Malik M. Elevated immune Theoharides TC. Nasal provocation of patients with
response in the brain of autistic patients. allergic rhinitis and the hypothalamic–pituitary–
J Neuroimmunol. 2009;207(1):111–6. adrenal axis. Ann Allergy Asthma Immunol.
83. Wei H, Zou H, Sheikh AM, Malik M, Dobkin C, 2007;98(3):269–73.
Brown WT, Li X. IL-6 is increased in the cerebellum 96. Liezmann C, Klapp B, Peters EM. Stress, atopy and
of autistic brain and alters neural cell adhesion, migra- allergy: a re-evaluation from a psychoneuroimmuno-
tion and synaptic formation. J Neuroinflammation. logic persepective. Dermatoendocrinol. 2011;3(1):​
2011;8(1):52. 37–40.
84. Gupta S, Ellis SE, Ashar FN, Moes A, Bader JS, 97. Jyonouchi H. Autism spectrum disorders and
Zhan J, West AB, Arking DE. Transcriptome analy- allergy: observation from a pediatric allergy/immu-
sis reveals dysregulation of innate immune response nology clinic. Expert Rev Clin Immunol. 2010;6(3):​
genes and neuronal activity-dependent genes in 397–411.
autism. Nat Commun. 2014;5:5748. 98. Catassi C, Bai JC, Bonaz B, Bouma G, Calabrò A,
85. Boris M, Kaiser CC, Goldblatt A, Elice MW, Carroccio A, Castillejo G, Ciacci C, Cristofori F,
Edelson SM, Adams JB, Feinstein DL. Effect of pio- Dolinsek J, Francavilla R, Elli L, Green P, Holtmeier
glitazone treatment on behavioral symptoms in W, Koehler P, Koletzko S, Meinhold C, Sanders D,
autistic children. J Neuroinflammation. 2007;4:3. Schumann M, Schuppan D, Ullrich R, Vécsei A,
86. Sharma A, Gokulchandran N, Chopra G, Kulkarni P, Volta U, Zevallos V, Sapone A, Fasano A. Non-
Lohia M, Badhe P, Jacob VC. Administration of celiac gluten sensitivity: the new frontier of gluten-
autologous bone marrow-derived mononuclear cells related disorders. Nutrients. 2013;5(10):3839–53.
in children with incurable neurological disorders and 99. Fasano A, Sapone A, Zevallos V, Schuppan
injury is safe and improves their quality of life. Cell D. Nonceliac gluten sensitivity. Gastroenterology.
Transplant. 2012;21(Supplement 1):S79–90. 2015;148(6):1195–204.
87. Schmitt J, Romanos M, Pfennig A, Leopold K, 100. Sapone A, Bai JC, Ciacci C, Dolinsek J, Green PH,
Meurer M. Psychiatric comorbidity in adult eczema. Hadjivassiliou M, Kaukinen K, Rostami K, Sanders
Br J Dermatol. 2009;161(4):878–83. DS, Schumann M, Ullrich R, Villalta D, Volta U,
88. Kohane IS, McMurry A, Weber G, MacFadden D, Catassi C, Fasano A. Spectrum of gluten-related
Rappaport L, Kunkel L, Bickel J, Wattanasin N, ­disorders: consensus on new nomenclature and clas-
Spence S, Murphy S, Churchill S. The co-morbidity sification. BMC Med. 2012;10:13.
burden of children and young adults with autism 101. Sandler RH, Finegold SM, Bolte ER, Buchanan CP,
spectrum disorders. PLoS One. 2012;7(4):e33224. Maxwell AP, Väisänen ML, Nelson MN, Wexler
89. Fasmer OB, Riise T, Eagan TM, Lund A, Dilsaver HM. Short-term benefit from oral vancomycin treat-
SC, Hundal O, Oedegaard KJ. Comorbidity of ment of regressive-onset autism. J Child Neurol.
asthma with ADHD. J Atten Disord. 2011;15(7):​ 2000;15(7):429–35.
564–71. 102. Schieve LA, Gonzalez V, Boulet SL, Visser SN, Rice
90. Millman M, Campbell MB, Wright KL, Johnston CE, Van Naarden BK, Boyle CA. Concurrent medi-
A. Allergy and learning disabilities in children. Ann cal conditions and health care use and needs among
Allergy. 1976;36(3):149–60. children with learning and behavioral developmental
91. Price CE, Rona RJ, Chinn S. Associations of exces- disabilities, National Health Interview Survey, 2006-­
sive irritability with common illnesses and food 2010. Res Dev Disabil. 2011;33(2):467–76.
intolerance. Paediatr Perinat Epidemiol. 1990;4(2):​ 103. Carroccio A, Mansueto P, Iacono G, Soresi M,
156–60. D’Alcamo A, Cavataio F, Brusca I, Florena AM,
476 M.A. Delgado et al.

Ambrosiano G, Seidita A, Pirrone G, Rini GB. Non-­ functioning autism and above average IQ. Mol
celiac wheat sensitivity diagnosed by double-blind Psychiatry. 2014;19(12):1251.
placebo-controlled challenge: exploring a new clini- 113. Siggins GR, Henriksen SJ, Chavkin C, Gruol
cal entity. Am J Gastroenterol. 2012;107(12):​ D. Opioid peptides and epileptogenesis in the limbic
1898–906. system: cellular mechanisms. Adv Neurol. 1986;​
104. Ashwood P, Anthony A, Pellicer AA, Torrente F, 44:501–12.
Walker-Smith JA, Wakefield AJ. Intestinal lympho- 114. Wagner R, DeLeo JA, Coombs DW, Willenbring S,
cyte populations in children with regressive autism: Fromm C. Spinal dynorphin immunoreactivity
evidence for extensive mucosal immunopathology. increases bilaterally in a neuropathic pain model.
J ClinImmunol. 2003;23(6):504–17. Brain Res. 1993;629(2):323–6.
105. Chen B, Girgis S, El-Matary W. Childhood autism and 115. Simmons ML, Chavkin C. Endogenous opioid regu-
eosinophilic colitis. Digestion. 2010;81(2):​127–9. lation of hippocampal function. Int Rev Neurobiol.
106. Courchesne E, Campbell K, Solso S. Brain growth 1996;39:145–96.
across the life span in autism: age-specific changes 116. Henriksen G, Willoch F. Imaging of opioid receptors
in anatomical pathology. Brain Res. 2011;1380:​ in the central nervous system. Brain. 2008;131(Pt
138–45. 5):1171–96.
107. Courchesne E, Pierce K, Schumann CM, Redcay E, 117. Zagon IS, Gibo DM, McLaughlin PJ. Adult and
Buckwalter JA, Kennedy DP, Morgan J. Mapping developing human cerebella exhibit different profiles
early brain development in autism. Neuron. of opioid binding sites. Brain Res. 1990;523:62–8.
2007;56(2):399–413. 118. Zagon IS, McLaughlin PJ. Opioid growth factor
108. Amaral DG, Schumann CM, Nordahl CW. Neuroanatomy receptor in the developing nervous system: labora-
of autism. Trends Neurosci. 2008;31(3):137–45. tory findings. In: Zagon IS, McLaughlin PJ, editors.
109. Catani M, Dell’Acqua F, Budisavljevic S, Howells Receptors and the developing nervous system,
H, Thiebaut de Schotten M, Froudist-Walsh S, Growth Factors and Hormones, vol. L. London:
D’Anna L, Thompson A, Sandrone S, Bullmore ET, Chapman and Hall; 1993. p. 39–62.
Suckling J, Baron-Cohen S, Lombardo MV, 119. Froehlich JC. Opioid peptides. Alcohol Health Res
Wheelwright SJ, Chakrabarti B, Lai MC, Ruigrok World. 1997;21(2):132–6.
AN, Leemans A, Ecker C, Consortium MA, Craig 120. Teschemacher H. Opioid receptor ligands derived
MC, Murphy DG. Frontal networks in adults with from food proteins. Curr Pharm Des. 2003;9(16):​
autism spectrum disorder. Brain. 2016;139(Pt 2):​ 1331–44.
616–30. 121. Doty RW. Schizophrenia: a disease of interhemi-
110. Zioudrou C, Streaty RA, Klee WA. Opioid peptides spheric processes at forebrain and brainstem levels?
derived from food proteins The exorphins. J Biol Behav Brain Res. 1989;34(1–2):1–33.
Chem. 1979;254(7):2446–9. 122. Sher L. Autistic disorder and the endogenous opioid
111. Huebner FR, Lieberman KW, Rubino RP, Wall system. Med Hypotheses. 1997;48(5):413–4.
JS. Demonstration of high opioid-like activity in iso- 123. Lajonchere C, Jones N, Coury DL, Perrin
lated peptides from wheat gluten hydrolysates. JM. Leadership in health care, research, and quality
Peptides. 1984;5(6):1139–47. improvement for children and adolescents with
112. van Elst LT, Maier S, Fangmeier T, Endres D, autism spectrum disorders: autism treatment net-
Mueller GT, Nickel K, Ebert D, Lange T, Hennig J, work and autism intervention research network on
Biscaldi M, Riedel A, Perlov E. Magnetic reso- physical health. Pediatrics. 2012;130(Suppl 2):​
nance spectroscopy comparing adults with high S62–8.

View publication stats

Das könnte Ihnen auch gefallen