Processed Review ASD
Processed Review ASD
Introduction
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental, biologically
based condition with an estimated prevalence of approximately 1 in 44 people [1]
that impacts all areas of child development — from behaviour, problem solving
abilities and self-care skills, to complex social communication ability, language, and
executive functioning skills. The range of symptoms and severity of Autism
Spectrum Disorder (ASD) vary greatly from child to child, and clinical
manifestations depend on the individual’s age, cognitive and language abilities, and
co-occurring conditions.
The last revision of the Diagnostic and Statistical Manual (DSM-5) defines Autism
Spectrum Disorder (ASD) as impairments in two main domains:
Proposed DSM-5 Autism Spectrum Disorder (ASD) criteria include three severity
classifications: Level 1 (“Requiring support”), Level 2 (“Requiring substantial
support”), and Level 3 (“Requiring very substantial support”) [5]. Prescribers often
describe or identify level 1 as mild Autism Spectrum Disorder (ASD), level 2 as
moderate Autism Spectrum Disorder (ASD), and level 3 as the most severe form of
Autism Spectrum Disorder (ASD) [6,7].
2.Epidemiology:
Autism Spectrum Disorder (ASD) occurs more often in boys than girls, with a 4:1
male-to-female ratio. [12] The reported prevalence rates of autism and its related
disorders have been increasing worldwide over the past decades, from
approximately 4 per 10 000 to 6 per 1000 children. [13-17] Globally, autism is
estimated to affect 24.8 million people as of 2015.[18] In the 2000s, the number of
people affected was estimated at 1–2 per 1, 000 people worldwide.[19]
➢ Sensory:
➢ Communication:
➢ Social skills:
➢ Play:
4.Causes of autism:
The exact cause of Autism Spectrum Disorder (ASD) is unknown. The most current
research demonstrates that there’s no single cause. Some of the suspected risk
factors for autism include:
▪ Autistic Disorder: (also called "classic" autism) This is what most people think
of when hearing the word "autism." People with autistic disorder usually have
significant language delays, social and communication challenges, and unusual
behaviors and interests. Many people with autistic disorder also have intellectual
disability.
▪ Asperger Syndrome: People with Asperger syndrome usually have some milder
symptoms of autistic disorder. They might have social challenges and unusual
behaviors and interests. However, they typically do not have problems with
language or intellectual disability.
5.DIAGNOSIS:
Diagnosis is based on behavior, not cause or mechanism.[27,28] Under the DSM-5,
autism is characterized by persistent deficits in social communication and
interaction across multiple contexts, as well as restricted, repetitive patterns of
behavior, interests, or activities. These deficits are present in early childhood,
typically before age three, and lead to clinically significant functional impairment.
[29] Sample symptoms include lack of social or emotional reciprocity, stereotyped
and repetitive use of language or idiosyncratic language, and persistent
preoccupation with unusual objects. Several diagnostic instruments are available.
Two are commonly used in autism research: the Autism Diagnostic Interview-
Revised (ADI-R) is a semi structured parent interview, and the Autism Diagnostic
Observation Schedule (ADOS)[30] uses observation and interaction with the child.
The Childhood Autism Rating Scale (CARS) is used widely in clinical environments
to assess severity of autism based on observation of children. [31] The Diagnostic
interview for social and communication disorders (DISCO) may also be used. [32]
Although the symptoms of autism and Autism Spectrum Disorder (ASD) begin early
in childhood, they are sometimes missed; years later, adults may seek diagnoses to
help them or their friends and family understand themselves, to help their
employers make adjustments, or in some locations to claim disability living
allowances or other benefits. Girls are often diagnosed later than boys. [33]
6.Pathophysiology:
➢ Autism mechanism can be divided into two areas: the pathophysiology of brain
structures and processes associated with autism, and the neuropsychological
linkages between brain structures and behaviours.[34] The behaviours appear to
have multiple pathophysiologies.[35] How autism occurs is not well understood.
➢ All known teratogens (agents that cause birth defects) related to the risk of
autism appear to act during the first eight weeks from conception, and though this
does not exclude the possibility that autism can be initiated or affected later, there is
strong evidence that autism arises very early in development. [39]
➢ Autism affects the amygdala, cerebellum, and many other parts of the brain.[40]
1. Zinc:
Approximately 10% of total brain Zn resides in synaptic vesicles of
glutamatergic neurons.
Zinc plays a role in regulating GABAergic inhibition, reducing seizure
susceptibility, and maintaining anxiolytic effects.
Zn deficiency has been shown to impair GABAergic function [45].
2. Copper:
Copper is a strong inhibitor of GABA-evoked responses, particularly affecting
Purkinje cells.
By interacting with Zn and the GABAA receptor complex, copper modulates
synaptic transmission [45].
Additionally, other NMDA receptor modulators are under investigation for their
therapeutic potential in Autism Spectrum Disorder (ASD). For instance, d-
cycloserine, a partial agonist of NMDA receptors, has demonstrated improvements
in social interactions and reductions in repetitive behaviors in animal models of
Autism Spectrum Disorder (ASD) [63–65]. However, further studies are necessary
to confirm whether these effects translate to human populations.
Findings in animal studies align with human research, showing that oxytocin is well-
tolerated and can significantly improve social behaviors in children with Autism
Spectrum Disorder (ASD) [75]. A double-blind, randomized, placebo-controlled trial
involving intranasal OXT administered over four weeks to 40 adult Autism
Spectrum Disorder (ASD) patients showed no treatment-specific improvements but
suggested positive trends in secondary outcomes like repetitive behaviors and
avoidance. This study recommended further research on multi-dose regimens to
evaluate long-term efficacy [76].
Based on these findings, the study suggests exploring extended multiple-dose IN-
OXT treatment regimens to promote neural changes in Autism Spectrum Disorder
(ASD) patients that persist beyond those achieved with four-week treatments [77].
In one study, 30 children with Autism Spectrum Disorder (ASD) were divided into
experimental (L-carnitine-supplemented) and placebo-controlled groups. The
experimental group showed significant improvements in CARS scores, with notable
differences in both free and total carnitine levels, though no direct link between
these changes was established. The study suggests a six-month L-carnitine
supplementation regimen to improve autism severity but calls for further research
to support this recommendation [86].
Studies have shown that butyrate treatment can improve Autism Spectrum Disorder
(ASD)-related behaviors, including regulating social behavior in autistic mice [92],
acting on transporters and receptors to correct gut-brain axis abnormalities [95],
and suppressing Histone Deacetylase activity to improve immune response [91].
Research has shown that individuals with Autism Spectrum Disorder (ASD) often
have vitamin B12 deficiency [100-102]. Low B12 levels result in elevated
homocysteine and reduced levels of S-Adenosylmethionine (SAM), a critical co-
substrate involved in methyl group transfers, which affects DNA and histone
methylation throughout the body, including in the brain. Depletion of SAM leads to a
decrease in GSH production, impairing the antioxidant defense system.
Folinic acid (vitamin B9), a folate analog, can pass through the blood-brain barrier
even in the presence of folate receptor (FRα) autoantibodies, which are commonly
found in children with Autism Spectrum Disorder (ASD) and their mothers
[104,105]. These autoantibodies block folate activity, and folinic acid has been
shown to prevent behavioral deficits in rats. In humans, folate supplementation
during preconception and gestational periods may help prevent Autism Spectrum
Disorder (ASD) by enabling the methylation of B12, activating the methionine cycle,
and producing GSH.
Studies have suggested that administering low-dose folic acid orally alongside
subcutaneously injected methyl-cobalamin can increase blood plasma levels of
glutathione, which may enhance antioxidant capacity and reduce oxidative stress in
a subset of children with autism [103,106].
Given these positive findings, several studies have examined whether restoring gut
microbiota in Autism Spectrum Disorder (ASD) patients could improve
neurobehavioral symptoms. A 12-week randomized, double-blind, controlled trial
showed some improvements in GI symptoms but no changes in adaptive or
repetitive behaviors [121]. A second randomized, double-blind, placebo-controlled
study reported contrasting results when testing prebiotic monotherapy and
combination therapy. Monotherapy showed no GI improvements, while
combination therapy, along with gluten and casein-free diets, resulted in reduced
anti-sociability scores [122]. Another study with 40 Egyptian children with Autism
Spectrum Disorder (ASD) (aged 2-5) found that after three months of daily probiotic
supplementation, Bifidobacterium and Lactobacillus levels in stool samples
significantly increased. Additionally, 80% of the children exhibited reduced anxiety
and improved sleep patterns [123].
A crossover-design pilot study investigated the effects of bovine colostrum product
(BCP) supplementation in children with Autism Spectrum Disorder (ASD) and
chronic GI symptoms, using both BCP alone and a combination of BCP and
Bifidobacterium (B.). The study included a washout period between the different
regimens [124,125]. The researchers observed significant improvements in GI
symptoms and stool quality, along with notable reductions in aberrant behaviors.
However, due to several limitations of the study, such as the lack of a control group,
the short study duration, and the continuation of concurrent Autism Spectrum
Disorder (ASD) treatments, the authors suggest that further, more rigorous studies
are necessary to verify the reliability of these findings.
Propionic acid (PPA), along with other short-chain fatty acids produced in the gut, is
found in elevated amounts in individuals with autism, and it is associated with
behavioral impairments due to its ability to easily cross the blood-brain barrier
[126]. In rats, PPA administration causes abnormal neural cell organization and
induces several electrophysiological, behavioral, and neuropathological changes
characteristic of Autism Spectrum Disorder (ASD) [127]. The harmful effects of PPA
are primarily linked to its concentration in the body. PPA is produced by gut
bacteria, particularly Clostridia, which generate PPA and exotoxins [128].
Interestingly, children with Autism Spectrum Disorder (ASD) exhibit differences in
gut microbiota compared to neurotypical individuals, with a decrease in
Bacteroidetes and an increase in Candida [117, 129, 130]. This imbalance disrupts
the cycle of PPA production and degradation, leading to elevated PPA levels that
contribute to behavioral deficits.
The updated version of this theory suggests that children with Autism Spectrum
Disorder (ASD), often having a "leaky gut," may metabolize gluten and casein
incompletely. This allows these peptides to pass through a compromised intestinal
barrier, enter the bloodstream, and penetrate the blood-brain barrier, affecting
brain function and neurotransmission [139]. The benefits of a GFCF diet are thus
thought to stem from both the "opioid excess theory" and its updated version.
Children with celiac disease, who are sensitive to gluten, exhibit an immune
response where their intestinal T cells react to gluten, producing interferon-γ,
which impairs gluten digestion and damages the intestinal lining [140,141]. Both
gluten and casein are also common allergens, triggering immune responses that
produce IgA and IgG antibodies [142].
The GFCF diet as an Autism Spectrum Disorder (ASD) treatment option has been
debated. Some studies report improvements in Autism Spectrum Disorder (ASD)
behaviors in children with comorbid gastrointestinal (GI) symptoms when following
the GFCF diet, as observed by their parents. However, other studies find no
statistically significant differences between experimental and control groups [143-
145]. Some researchers criticize the GFCF diet findings as methodologically flawed,
recommending its use only if gluten or casein intolerance or allergy is diagnosed
[146].
In one case-control study, participants were divided into three groups [150]. The
first group followed a modified version of the ketogenic diet called the Modified
Atkins Diet (MAD), which is less restrictive than the standard KD. The second group
adhered to the gluten-free/casein-free (GFCF) diet, and the third group served as
the control. All participants were assessed using the Childhood Autism Rating Scale
(CARS) and the Autism Treatment Evaluation Checklist (ATEC). The study found
that both MAD and GFCF diets led to a reduction in CARS and ATEC scores,
indicating improvements in autism-related behaviors. Notably, MAD participants
showed significant improvements in speech, social interaction, and cognition as
measured by the ATEC. The study concluded that both diets showed beneficial
effects, although these improvements were more pronounced in different
behavioral areas depending on the diet [150].
A six-month pilot study with 30 participants aged 4 to 10 years used the John
Radcliffe keto diet [151]. Of the 18 participants who completed the study, varying
levels of psychosomatic improvements were observed. The researchers
hypothesized that autistic children have impaired glucose oxidation and that ketone
bodies could serve as an alternative energy source for their brains. The study
acknowledged several limitations, including the heterogeneity of the participants'
biochemical responses, difficulties for some children (especially those with severe
Autism Spectrum Disorder (ASD)) in adhering to the diet, and uncertainty about the
optimal duration and method of applying the diet.
Metallothioneins (MTs) are proteins that bind to metals to prevent their harmful
effects, but toxic metals like mercury (Hg) can displace zinc (Zn) and copper (Cu)
from MTs, potentially causing negative health effects. Chelation therapy increases
the excretion of essential trace elements like Cu and Zn, which can lead to
deficiencies, so monitoring these levels is critical. Excess Cu in the body can
interfere with Zn absorption, affecting MT gene transcription and the elimination of
toxic metals, emphasizing the importance of careful monitoring of metal toxicity and
the patient's health status [155].
Chelation therapy should only be conducted under medical supervision, after
assessing the potential risks and benefits. A review of available data identified
serious adverse events associated with chelation therapy in Autism Spectrum
Disorder (ASD), including hypocalcemia, renal impairment, and even deaths, leading
to the conclusion that the risks currently outweigh any proven benefits [157].
14. ANTI-INFLAMMATORIES
14.1. Non-steroidal Anti-inflammatories (NSAIDs)
Among all the vitamins linked to Autism Spectrum Disorder (ASD) pathology,
Vitamin D has the strongest and most extensively researched correlation. Several
studies have found a connection between Vitamin D deficiency in pregnant mothers
and an increased likelihood of their children developing Autism Spectrum Disorder
(ASD) [159, 160]. This led to research exploring how low Vitamin D levels might
contribute to Autism Spectrum Disorder (ASD) and how supplementation might
alleviate related symptoms. Vitamin D's anti-inflammatory effects are seen in its
ability to counteract inflammation induced by Lipopolysaccharide (LPS) by
inhibiting MAPK pathways and the production of inflammatory molecules [161].
Additionally, Vitamin D has a synergistic effect with serotonin. Calcitriol activates
Tryptophan Hydroxylase 2 and suppresses Tryptophan Hydroxylase 1 transcription,
which increases serotonin in the brain, leading to prosocial behaviors. However, this
effect is absent in children with autism who exhibit antisocial behaviors [162]. A
randomized controlled trial demonstrated that Vitamin D supplementation led to
better retention of its precursor, 25(OH)D, and resulted in significantly improved
scores across various behavioral assessments after four months of supplementation
[163].
Zinc is an essential trace element critical for maintaining optimal brain function
[167, 168]. A deficiency in zinc can lead to neuropsychological changes such as
emotional instability, irritability, and depression [169, 170]. Zinc is also vital for
cognitive performance and plays a role in glutamatergic transmission, influencing
both short-term and long-term mental effects [45]. Zinc acts by blocking NMDA
receptors and serving as a signal factor in various cellular processes, and its
deficiency may impair neurotransmission [171]. Moreover, zinc acts as a co-
transmitter with glutamate, helping to prevent excitotoxicity and providing
essential nutrients for enzymes involved in synaptic functions related to learning
and memory processes [45].
Copper toxicity can have a significant impact on the brain, leading to symptoms such
as depression, irritability, anxiety, nervousness, as well as learning and behavioral
issues [45, 170]. Copper serves as a cofactor for dopamine-β-hydroxylase (DBH), an
enzyme that converts dopamine into norepinephrine [172]. Excess copper can
elevate norepinephrine levels, which have been observed in individuals with Autism
Spectrum Disorder (ASD). Additionally, excess copper inhibits the enzyme
hydroxytryptophan decarboxylase, which results in reduced serotonin production.
Hypercupremia, or copper excess, may be linked to depression. Furthermore, an
overabundance of Cu/Zn-dependent superoxide dismutase can heighten oxidative
stress, causing redox imbalances through reactions with hydrogen peroxide (H₂O₂)
and peroxynitrite [45]
Zinc (Zn) and copper (Cu) are essential trace elements with an inverse relationship,
and their levels in the body are crucial for various biological processes. Cytokines
play a role in regulating this balance by facilitating Zn uptake and the production of
ceruloplasmin in the liver. In neurotypical individuals, the normal Zn-to-Cu ratio is
approximately 1:1 [45]. The plasma zinc/serum copper ratio can be used to quickly
assess the functional state of metallothioneins (MTs). Studies have shown that
children with Autism Spectrum Disorder (ASD) tend to have lower Zn/Cu ratios
compared to neurotypical children, suggesting either a deficiency in Zn or an
accumulation of toxic metals that antagonize Zn [45, 169, 170]. Mercury toxicity, in
particular, may significantly disrupt MT function in children with Autism Spectrum
Disorder (ASD), potentially reflected in the altered Zn/Cu ratio [45].
Toxic metals such as cadmium (Cd) and mercury (Hg) can have opposing effects on
Zn and Cu metabolism. MT induction in the liver may influence Cu excretion more
significantly than its mobilization into the bloodstream, while Zn's mobilization to
the blood is more affected. Increased MT induction due to oxidative stress may also
lead to the retention of toxic metals like Cd and Hg in organs such as the liver and
kidneys, explaining the higher levels of these metals found in Autism Spectrum
Disorder (ASD) patients. Once accumulated, these metals may further impair Zn
metabolism, worsening the patient's condition [45].
Patients with Autism Spectrum Disorder (ASD) may have a reduced tolerance to
toxic metal exposure. Enhanced MT induction due to oxidative stress may also
disrupt Cu excretion through the bile, leading to Cu accumulation in the liver and
potential toxicity. This buildup could interfere with Zn metabolism and reduce
tolerance to additional toxic metal exposure. The opposing effects of MT induction
on Cu and Zn metabolism may worsen both Zn deficiency and Cu toxicity
simultaneously in Autism Spectrum Disorder (ASD) patients [45].
15.2. Selenium
Research suggests that disruptions in selenium (Se) metabolism may play a role in
the development of Autism Spectrum Disorder (ASD) [173, 174]. Oxidative stress,
which is commonly observed in Autism Spectrum Disorder (ASD), is linked to
mitochondrial dysfunction, immune dysfunction, and inflammation [78, 79, 174].
Selenium has been shown to help alleviate these conditions, prompting several
studies to explore its potential role in managing Autism Spectrum Disorder (ASD)
[175, 176]. However, the exact mechanisms remain unclear, as numerous peer-
reviewed studies have reported altered selenoprotein expression in Autism
Spectrum Disorder (ASD), while others have found no significant changes [175].
Selenium may offer protection against the harmful effects of mercury [177, 178] and
may also protect against heavy metal toxicity, which has been hypothesized as a
contributing factor in Autism Spectrum Disorder (ASD) development [179]. The
complex interaction between selenium and mercury may have implications for
Autism Spectrum Disorder (ASD) and other neurological disorders, though further
research is needed to better understand this relationship and its clinical
significance.
Alterations in the gut microbiota have also been associated with Autism Spectrum
Disorder (ASD), and selenium deficiency has been shown to cause changes in
bacterial populations that promote lipopolysaccharide (LPS) overproduction and
translocation. Selenium may influence LPS-induced inflammation by modulating
p38 MAPK and NF-κB signaling pathways to protect against endotoxemia [175].
Although the evidence regarding selenium’s role in Autism Spectrum Disorder
(ASD) is limited and sometimes contradictory, it is an essential micronutrient that
may provide benefits for alleviating some Autism Spectrum Disorder (ASD)
symptoms. More research is required to identify the optimal dosage and duration
for selenium supplementation and to clarify its role in Autism Spectrum Disorder
(ASD) treatment.
Calcium (Ca) and magnesium (Mg) are essential minerals that are vital for various
bodily functions, including neurological development and function [180].
Imbalances or deficiencies in these minerals have been implicated in the
pathogenesis of Autism Spectrum Disorder (ASD) [181].
Calcium is the most abundant mineral in the body, playing essential roles in muscle
contraction, blood clotting, nerve function, and regulating neuronal communication,
which is crucial for proper brain function [182]. Studies suggest that calcium
dysregulation may contribute to Autism Spectrum Disorder (ASD) development,
with research showing that children with Autism Spectrum Disorder (ASD) have
lower serum Ca levels compared to neurotypical children [184]. Calcium
dysregulation is also linked to oxidative stress, inflammation, and mitochondrial
dysfunction, which are common in individuals with Autism Spectrum Disorder
(ASD) [183]. Calcium channel blockers, which regulate Ca influx into cells, have been
shown to improve social behavior in Autism Spectrum Disorder (ASD) mouse
models.
Magnesium is another crucial mineral involved in muscle and nerve function, energy
production, and protein synthesis. It plays a key role in regulating calcium levels in
the body [185], and its dysregulation has also been associated with oxidative stress,
inflammation, and mitochondrial dysfunction in Autism Spectrum Disorder (ASD)
[186]. Some studies have indicated that children with Autism Spectrum Disorder
(ASD) have lower serum Mg levels compared to neurotypical children [187, 188].
The interaction between calcium and magnesium is critical for proper body
function. Magnesium regulates calcium levels by activating vitamin D, which is
necessary for calcium absorption in the intestines [189]. Magnesium also controls
the influx of calcium into cells, essential for neurological function, and helps inhibit
glutamate release, a neurotransmitter that can lead to excessive calcium influx and
oxidative stress [190]. Studies have shown that children with Autism Spectrum
Disorder (ASD) have a higher calcium-to-magnesium ratio compared to
neurotypical children, and this ratio correlates with autism severity. Additionally,
children with both Autism Spectrum Disorder (ASD) and ADHD exhibit more
significant changes in magnesium levels in their hair and urine compared to
neurotypical children [188]. Magnesium may help protect against the harmful
effects of calcium dysregulation and has anti-inflammatory properties that could
help mitigate mitochondrial dysfunction, commonly observed in children with
Autism Spectrum Disorder (ASD).
The role of calcium and magnesium in Autism Spectrum Disorder (ASD) is complex,
and further research is needed to clarify their exact mechanisms. However, evidence
suggests that dysregulation of these minerals may contribute to Autism Spectrum
Disorder (ASD) pathogenesis, and supplementation with calcium and magnesium
may alleviate some Autism Spectrum Disorder (ASD) symptoms [192]. The optimal
doses and duration for supplementation still require further investigation.
16. Vitamins
Vitamin supplementation is a common adjunct therapy for individuals with Autism
Spectrum Disorder (ASD) due to the links between certain vitamins and Autism
Spectrum Disorder (ASD) pathology [193-195]. Vitamins A, C, and E are known to
enhance antioxidant capabilities, while vitamin D plays roles in anti-inflammatory
processes and serotonin regulation [162, 196, 197]. Vitamin B1 is involved in
energy regulation [198], and vitamin B6 helps balance excitation and inhibition in
the brain through neurotransmitter synthesis, particularly in regulating GABA [199,
200]. Vitamins B9 and B12 contribute to methylation processes [201].
Individuals with Autism Spectrum Disorder (ASD) often experience metabolic and
nutritional abnormalities, including issues with sulfation, methylation, glutathione
redox imbalances, oxidative stress, and mitochondrial dysfunction [202-204].
Vitamin supplementation may support these physiological processes. A non-
randomized double-blind study involving 16 children with Autism Spectrum
Disorder (ASD), who received high doses of B6 and magnesium, found significant
behavioral improvements [205]. However, the role of B6 supplementation in Autism
Spectrum Disorder (ASD) remains unclear. Vitamin D supplementation may also
benefit individuals with Autism Spectrum Disorder (ASD) due to its immune-
modulating and inflammation-reducing properties [206].
The optimal dosages and duration of vitamin supplementation for individuals with
Autism Spectrum Disorder (ASD) are still unclear. Moreover, vitamin
supplementation may not be effective for all individuals with Autism Spectrum
Disorder (ASD) and should be tailored on a case-by-case basis [194].
17. Tryptophan
Tryptophan, an essential amino acid and precursor to serotonin, may benefit
individuals with Autism Spectrum Disorder (ASD) by improving social behavior,
reducing repetitive behaviors, and increasing cognitive flexibility [207, 208]. While
its exact mechanism in Autism Spectrum Disorder (ASD) is not fully understood, it
may enhance serotonin levels in the brain and exert anti-inflammatory effects.
However, tryptophan supplementation should be approached cautiously due to
potential side effects and the risk of serotonin syndrome when combined with
certain medications. While tryptophan may be deficient in some individuals with
Autism Spectrum Disorder (ASD), supplementation with B vitamins and magnesium
may influence tryptophan metabolism [207]. More research is necessary to better
understand the benefits and risks of tryptophan supplementation in Autism
Spectrum Disorder (ASD).
18. Immunotherapy
Individuals with Autism Spectrum Disorder (ASD) often exhibit immune system
dysregulation, which includes alterations in T cells, B cells, monocytes, natural killer
cells, dendritic cells, and elevated cytokine levels leading to neuroinflammation.
Immune dysfunction and inflammation are key aspects of Autism Spectrum
Disorder (ASD) diagnosis and treatment [209]. Understanding immune
dysregulation and inflammation in Autism Spectrum Disorder (ASD) can
significantly improve diagnostic and therapeutic approaches. Immunotherapy, such
as intravenous immunoglobulin (IVIG) injections, has shown promise in improving
Autism Spectrum Disorder (ASD) symptoms [210]. However, further research is
needed to identify additional immune biomarkers and explore the long-term risks
and effects of such treatments.
Reference:-
1) Maenner MJ, Shaw KA, Bakian AV, Bilder DA, Durkin MS, Esler A, et al.
Prevalence and characteristics of autism spectrum disorder among children aged 8
years — autism and developmental disabilities monitoring network, 11 sites,
United States, 2018. Morbidity and mortality weekly report Surveillance
summaries (Washington, DC : 2002). 2021;70(11):1–16.
2) Johnson J, Spitzer R, Williams J. Diagnostic and Statistical Manual of Mental
Disorders-IV TR. Washington, DC: American Psychiatric Association; 2000.
3) Hyman SL, Levy SE, Myers SM. Identification, evaluation, and management of
children with autism spectrum disorder. Paediatrics. 2020;145(1).
4) Association D-AP. Diagnostic and statistical manual of mental disorders.
Arlington: American Psychiatric Publishing. 2013.
5) Weitlauf AS, Gotham KO, Vehorn AC, Warren ZE (2014) Brief report: DSM-5
“levels of support:” a comment on discrepant conceptualizations of severity in
ASD. J Autism Dev Disord 44(2):471–476
6) The Autism Cafe. AUTISM SEVERITY LEVELS: FROM MILD TO SEVERE
2017 [Available from: https://theautismcafe.com/autism-severity-levels-mild-to-
severe/
7) PsychCentral Levels of Autism: Understanding the Different Types of ASD 2019
[Available from: https://psychcentral.com/pro/child-therapist/2019/11/levels-of-
autism-understanding-the-different-types-of-asd
8) Kolvin I. Studies in childhood psychoses: I. Diagnostic criteria and classification.
Brit J Psychiatry.1971; 118:381–4. [PubMed]
9) Klin A. Asperger syndrome: an update. Rev Bras Psiquiatr. 2003; 25:103–9.
[PubMed]
10) Al-Beltagi M (2021) Autism medical comorbidities. World J Clin Pediatr
10(3):15–28
11) World Health Organisation. Autism 2022 [Available from:
https://www.who.int/newsroom/factsheets/detail/autismspectrumdisorders?
msclkid=1ad10a2ab8c311eca388b42ff641e1c3
12) Autism South Africa. What is Autism 2020 [Available from:
https://aut2know.co.za/?msclkid=d3e8069ab8bc11ec826c587a8f 7e508a
Fombonne E, Zakarian R, Bennett A, Meng L, McLean Heywood D. Pervasive
evelopmental disorders in Montreal, Quebec, Canada: prevalence and links with
immunizations. Pediatrics. 2006; 118:e139–50. Available from:
http://pediatrics.aappublications.org/cgi/reprint/118/1/e139 [lastcited on 2009 Jun
19] [PubMed}.
13) Chakrabarti S, Fombonne E. Pervasive developmental disorders in preschool
children. JAMA.2001; 285:3093–9. [PubMed]
14) Chakrabarti S, Fombonne E. Pervasive developmental disorders in preschool
children: confirmation of high prevalence. Am J Psychiatry. 2005; 162:1133–41.
[PubMed]
15) Centers for Disease Control and Prevention. Mental health in the United States:
parental report of diagnosed autism in children aged 4-17 years, United States,
2003-2004. MMWR Morb Mortal WklyRep. 2006; 55:481–6. [PubMed]
16) Bertrand J, Mars A, Boyle C, Bove F, Yeargin-Allsop M, DeCoufle P. Prevalence
of autism in a United States population: the Brick Township, New Jersey,
investigation. Pediatr. 2001; 108:1155–61. [PubMed]
17) Yeargin-Allsopp M, Rice C, Karapurkar T, Doernberg N, Boyle C, Murphy C.
Prevalence of autismin a US metropolitan area. JAMA. 2003; 289:49–55.
[PubMed]
18) GBD 2015 Disease and Injury Incidence and Prevalence, Collaborators. (8
October 2016). "Global, regional, and national incidence, prevalence, and years
lived with disability for 310 diseases and injuries, 1990–2015: a systematic
analysis for the Global Burden of Disease Study 2015". Lancet. 388 (10053):
1545–602. doi:10.1016/S0140-6736(16)31678-6. PMC 5055577. PMID
27733282.
19) Newschaffer CJ, Croen LA, Daniels J, Giarelli E, Grether JK, Levy SE, Mandell
DS, Miller LA, Pinto-Martin J, Reaven J, Reynolds AM, Rice CE, Schendel D,
Windham GC (2007). "The epidemiology of autism spectrum disorders". Annual
Review of Public Health. 28: 235–58. doi:
10.1146/annurev.publhealth.28.021406.144007. PMID 17367287
20) Landa RJ (2008). "Diagnosis of autism spectrum disorders in the first 3 years of
life". Nat Clin Pract Neurol. 4 (3): 138–47. doi:10.1038/ncpneuro0731. PMID
18253102.
21) Abrahams BS, Geschwind DH (May 2008). "Advances in autism genetics: on the
threshold of a new neurobiology". Nature Reviews. Genetics. 9 (5): 341– PMC
2756414. PMID 18414403.
22) Buxbaum JD (2009). "Multiple rare variants in the etiology of autism spectrum
disorders". Dialogues in Clinical Neuroscience. 11 (1): 35–43. PMC 3181906.
PMID 19432386.
23) Lyall K, Schmidt RJ, Hertz-Picciotto I (April 2014). "Maternal lifestyle and
environmental risk factors for autism spectrum disorders". Int J Epidemiol. 43 (2):
443–PMC 3997376. PMID 24518932.
24) American Psychiatric Association. Diagnostic and Statistical Manual of Mental
Disorders. 4th ed.Arlington, VA: American Psychiatric Publishing, Inc; 2000. pp.
69–84.
25) World Health Organization. International Statistical Classification of Diseases
and Related HealthProblems, 10th Revision. World Health Organization; 2007
Available from: http://www.who.int/classifications/apps/icd/icd10°nline/ [last
cited on 2009 Mar 19]
26) Mattila ML, Kielinen M, Jussila K, Linna SL, Bloigu R, Ebeling H, Moilanen I.
An epidemiological and diagnostic study of Asperger syndrome according to four
sets of diagnostic criteria. J Am Acad Child Adolesc Psychiatry. 2007; 46:636–
46. [PubMed]
27) London E (2007). "The role of the neurobiologist in redefining the diagnosis of
autism". Brain Pathol. 17 (4): 408–11. doi:10.1111/j.1750-3639.2007.00103.x.
PMID 17919126.
28) Baird G, Cass H, Slonims V (2003). "Diagnosis of autism". BMJ. 327 (7413):
488–93. doi:10.1136/bmj.327.7413.488. PMC 188387. PMID 12946972.
29) Autism Spectrum Disorder, 299.00 (F84.0). In: American Psychiatric association.
Diagnostic and Statistical Manual of Mental Disorders, Fifth Edition. American
Psychiatric Publishing; 2013.
30) Gotham K, Risi S, Dawson G, Tager-Flusberg H, Joseph R, Carter A, Hepburn S,
McMahon W, Rodier P, Hyman SL, Sigman M, Rogers S, Landa R, Spence MA,
Osann K, Flodman P, Volkmar F, Hollander E, Buxbaum J, Pickles A, Lord C
(June 2008). "A replication of the Autism Diagnostic Observation Schedule
(ADOS) revised algorithms". Journal of the American Academy of Child and
Adolescent Psychiatry. 47 (6): 642–. PMC 3057666. PMID 18434924.
31) Volkmar FR, Paul R, Pelphrey KA, Rogers SJ, eds. (2014). Handbook of Autism
and Pervasive Developmental Disorders: Volume Two: Assessment,
Interventions, and Policy. 2 (4th ed.). Hoboken, New Jersey: John Wiley & Sons.
p. 301. ISBN 978-1- 118-28220-5. LCCN 2013034363. OCLC 946133861.
Retrieved 1 March 2019 – via Google Books.
32) Kan CC, Buitelaar JK, van der Gaag RJ (June 2008). "[Autism spectrum disorders
in adults]". Nederlands Tijdschrift voor Geneeskunde. 152 (24): 1365–9. PMID
18664213.
33) Why Many Autistic Girls are Overlooked (Report). Child Mind Institute.
Retrieved 13 February 2018.
34) Penn HE (2006). "Neurobiological correlates of autism: a review of recent
research". Child Neuropsychol. 12 (1): 57–79. doi:10.1080/09297040500253546.
PMID 16484102.
35) London E (2007). "The role of the neurobiologist in redefining the diagnosis of
autism". Brain Pathol. 17 (4): 408–11. doi:10.1111/j.1750-3639.2007.00103.x.
PMID 17919126.
36) Wasilewska J, Klukowski M (2015). "Gastrointestinal symptoms and autism
spectrum disorder: links and risks - a possible new overlap syndrome". Paediatric
Health Med Ther(Review). 6: 153– 166. doi:10.2147/PHMT.S85717. PMC
5683266. PMID 29388597
37) Rao M, Gershon MD (September 2016). "The bowel and beyond: the enteric
nervous system in neurological disorders". Nat Rev Gastroenterol Hepatol
(Review). 13 (9): 517–28. doi:10.1038/nrgastro.2016.107. PMC 5005185. PMID
27435372.
38) Betancur C, Sakurai T, Buxbaum JD (2009). "The emerging role of synaptic cell-
adhesion pathways in the pathogenesis of autism spectrum disorders". Trends
Neurosci. 32 (7): 402–12. doi:10.1016/j.tins.2009.04.003. PMID 19541375.
39) Arndt TL, Stodgell CJ, Rodier PM (2005). "The teratology of autism". Int J Dev
Neurosci. 23 (2–3): 189–99. doi:10.1016/j.ijdevneu.2004.11.001. PMID
15749245.
40) Amaral DG, Schumann CM, Nordahl CW (2008). "Neuroanatomy of autism".
Trends Neurosci. 31 (3): 137–45. doi:10.1016/j.tins.2007.12.005.PMID18258309
41) Bourgeron, T. The possible interplay of synaptic and clock genes in autism
spectrum disorders. Cold Spring Harb. Symp. Quant. Biol., 2007, 72(1), 645-654.
http://dx.doi.org/10.1101/sqb.2007.72.020 PMID:18419324
42) Gogolla, N.; LeBlanc, J.J.; Quast, K.B.; Südhof, T.C.; Fagiolini, M.; Hensch, T.K.
Common circuit defect of excitatory-inhibitory balance in mouse models of
autism. J. Neurodev. Disord., 2009, 1(2), 172-181.
http://dx.doi.org/10.1007/s11689-009-9023-x PMID:20664807
43) Cellot, G.; Cherubini, E. GABAergic signaling as therapeutic target for autism
spectrum disorders. Front Pediatr., 2014, 2, 70.
http://dx.doi.org/10.3389/fped.2014.00070 PMID:25072038
44) Nelson, S.B.; Valakh, V. Excitatory/inhibitory balance and circuit homeostasis in
autism spectrum disorders. Neuron, 2015, 87(4), 684-698.
http://dx.doi.org/10.1016/j.neuron.2015.07.033 PMID:26291155
45) Bjorklund, G. The role of zinc and copper in autism spectrum disorders. Acta
Neurobiol. Exp., 2013, 73(2), 225-236. PMID: 23823984
46) Campbell, M.; Rapoport, J.L.; Simpson, G.M. Antipsychotics in children and
adolescents. J. Am. Acad. Child Adolesc. Psychiatry, 1999, 38(5), 537-545.
http://dx.doi.org/10.1097/00004583-199905000-00015 PMID:10230185
47) Chopko, T.C.; Lindsley, C.W. Classics in chemical neuroscience: Risperidone.
ACS Chem. Neurosci., 2018, 9(7), 1520-1529.
http://dx.doi.org/10.1021/acschemneuro.8b00159 PMID:29695153
48) Vardanyan, R. Piperidine-Based Drug Discovery; Elsevier, 2017.
49) Robert, L. Cross-discipline team leader review memo. 2012. Available From:
https://www.accessdata.fda.gov/-
drugsatfda_docs/summary_review/2008/021817se1-001_- SUMR.pdf
50) Kent, J.M.; Kushner, S.; Ning, X.; Karcher, K.; Ness, S.; Aman, M.; Singh, J.;
Hough, D. Risperidone dosing in children and adolescents with autistic disorder:
A double-blind, placebo-controlled study. J. Autism Dev. Disord., 2013, 43(8),
1773-1783. http://dx.doi.org/10.1007/s10803-012-1723-5PMID:23212807
51) Kent, J.M.; Hough, D.; Singh, J.; Karcher, K.; Pandina, G. An open-label
extension study of the safety and efficacy of risperidone in children and
adolescents with autistic disorder. J. Child Adolesc. Psychopharmacol., 2013,
23(10), 676-686. http://dx.doi.org/10.1089/cap.2012.0058PMID:24350813
52) Jesner, O.S.; Aref-Adib, M.; Coren, E. Risperidone for autism spectrum disorder.
Cochrane Libr., 2007, 2010(1),
CD005040.http://dx.doi.org/10.1002/14651858.CD005040.pub2 PMID:17253538
53) West, L.; Waldrop, J. Risperidone use in the treatment of behavioural symptoms
in children with autism. Pediatr. Nurs., 2006, 32(6), 545-549. PMID:17256292
54) Kim, J.-W.; Seung, H.; Kim, K. C.; Gonzales, E. L. T.; Oh, H. A.; Yang, S. M.;
Ko, M. J.; Han, S.-H.; Banerjee, S.; Shin, C. Y. Agmatine rescues autistic
behaviors in the valproic acid-induced animal model of autism.
Neuropharmacology, 2017, 113(Pt A), 71-81.
http://dx.doi.org/10.1016/j.neuropharm.2016.09.014
55) Raasch, W.; Schäfer, U.; Chun, J.; Dominiak, P. Biological significance of
agmatine, an endogenous ligand at imidazoline binding sites. Br. J. Pharmacol.,
2001, 133(6), 755-780. http://dx.doi.org/10.1038/sj.bjp.0704153 PMID:11454649
56) Uzbay, T.; Goktalay, G.; Kayir, H.; Eker, S.S.; Sarandol, A.; Oral, S.;
Buyukuysal, L.; Ulusoy, G.; Kirli, S. Increased plasma agmatine levels in patients
with schizophrenia. J. Psychiatr. Res., 2013, 47(8), 1054-1060.
http://dx.doi.org/10.1016/j.jpsychires.2013.04.004 PMID:23664672
57) Esnafoglu, E.; İrende, İ. Decreased plasma agmatine levels in autistic subjects. J.
Neural Transm., 2018, 125(4), 735-740. http://dx.doi.org/10.1007/s00702-017-
1836-2 PMID:29302750
58) Silverman, J.M.; Brunet, Y.R.; Cascales, E.; Mougous, J.D. Structure and
regulation of the type VI secretion system. Annu. Rev. Microbiol., 2012, 66(1),
453-472. http://dx.doi.org/10.1146/annurev-micro-121809-151619
PMID:22746332
59) Lee, K.; Mills, Z.; Cheung, P.; Cheyne, J.E.; Montgomery, J.M. The role of zinc
and NMDA receptors in autism spectrum disorders. Pharmaceuticals, 2022, 16(1),
1. http://dx.doi.org/10.3390/ph16010001 PMID:36678498
60) Vatankhah Ardestani, S.S.; Karahmadi, M.; Tarrahi, M.J.; Omranifard, V.;
Farzaneh, B. Efficacy of memantine as adjunct therapy for autism spectrum
disorder in children aged 14 years. Adv. Biomed. Res., 2018, 7(1), 131.
http://dx.doi.org/10.4103/abr.abr_100_18 PMID:30320040
61) Soorya, L.V.; Fogg, L.; Ocampo, E.; Printen, M.; Youngkin, S.; Halpern, D.;
Kolevzon, A.; Lee, S.; Grodberg, D.; Anagnostou, E. Neurocognitive outcomes
from memantine: A pilot, double-blind, placebo-controlled trial in children with
autism spectrum disorder. J. Child Adolesc. Psychopharmacol., 2021, 31(7), 475-
484. http://dx.doi.org/10.1089/cap.2021.0010 PMID:34543081
62) Aman, M.G.; Findling, R.L.; Hardan, A.Y.; Hendren, R.L.; Melmed, R.D.;
Kehinde-Nelson, O.; Hsu, H.A.; Trugman, J.M.; Palmer, R.H.; Graham, S.M.;
Gage, A.T.; Perhach, J.L.; Katz, E. Safety and efficacy of memantine in children
with autism: Randomized, placebo-controlled study and open-label extension. J.
Child Adolesc. Psychopharmacol., 2017, 27(5), 403-412.
http://dx.doi.org/10.1089/cap.2015.0146 PMID:26978327
63) Modi, M.E.; Young, L.J. D-cycloserine facilitates socially reinforced learning in
an animal model relevant to autism spectrum disorders. Biol. Psychiatry, 2011,
70(3), 298-304. http://dx.doi.org/10.1016/j.biopsych.2011.01.026PMID:21481844
64) Minshawi, N.F.; Wink, L.K.; Shaffer, R.; Plawecki, M.H.; Posey, D.J.; Liu, H.;
Hurwitz, S.; McDougle, C.J.; Swiezy, N.B.; Erickson, C.A. A randomized,
placebo-controlled trial of d-cycloserine for the enhancement of social skills
training in autism spectrum disorders. Mol. Autism, 2016, 7(1), 2.
http://dx.doi.org/10.1186/s13229-015-0062-8 PMID:26770664
65) Burket, J.A.; Benson, A.D.; Tang, A.H.; Deutsch, S.I. d-- Cycloserine improves
sociability in the BTBR T+ Itpr3tf/J mouse model of autism spectrum disorders
with altered Ras/Raf/ERK1/2 signaling. Brain Res. Bull., 2013, 96, 62-70.
http://dx.doi.org/10.1016/j.brainresbull.2013.05.003 PMID:23685206
66) Zhao, H.; Mao, X.; Zhu, C.; Zou, X.; Peng, F.; Yang, W.; Li, B.; Li, G.; Ge, T.;
Cui, R. GABAergic system dysfunction in autism spectrum disorders. Front. Cell
Dev. Biol., 2022, 9, 781327.
http://dx.doi.org/10.3389/fcell.2021.781327PMID:35198562
67) Braat, S.; D’Hulst, C.; Heulens, I.; De Rubeis, S.; Mientjes, E.; Nelson, D.L.;
Willemsen, R.; Bagni, C.; Van Dam, D.; De Deyn, P.P.; Kooy, R.F. The GABA A
receptor is an FMRP target with therapeutic potential in fragile X syndrome. Cell
Cycle, 2015,14(18),2985-2995.
http://dx.doi.org/10.4161/15384101.2014.989114PMID:25790165
68) Silverman, J.L.; Pride, M.C.; Hayes, J.E.; Puhger, K.R.; Butler-Struben, H.M.;
Baker, S.; Crawley, J.N. GABAB receptor agonist r-baclofen reverses social
deficits and reduces repetitive behavior in two mouse models of autism.
Neuropsychopharmacology, 2015, 40(9), 2228-2239.
http://dx.doi.org/10.1038/npp.2015.66 PMID:25754761
69) Mahdavinasab, S.M.; Saghazadeh, A.; Motamed-Gorji, N.; Vaseghi, S.;
Mohammadi, M.R.; Alichani, R.; Akhondzadeh, S. Baclofen as an adjuvant
therapy for autism: A randomized, double-blind, placebo-controlled trial. Eur.
Child Adolesc. Psychiatry, 2019, 28(12), 1619-1628.
http://dx.doi.org/10.1007/s00787-019-01333-5PMID:30980177
70) Tan, T.; Wang, W.; Xu, H.; Huang, Z.; Wang, Y.T.; Dong, Z. Low-frequency
rTMS ameliorates autistic-like behaviors in rats induced by neonatal isolation
through regulating the synaptic GABA transmission. Front. Cell. Neurosci., 2018,
12, 46. http://dx.doi.org/10.3389/fncel.2018.00046 PMID:29541022
71) Desarkar, P.; Rajji, T.K.; Ameis, S.H.; Blumberger, D.M.; Lai, M.C.; Lunsky, Y.;
Daskalakis, Z.J. Assessing and stabilizing atypical plasticity in autism spectrum
disorder using rTMS: Results from a proof-of-principle study. Clin.
Neurophysiol., 2022, 141, 109-118.
http://dx.doi.org/10.1016/j.clinph.2021.03.046 PMID:34011467
72) Enticott, P.G.; Barlow, K.; Guastella, A.J.; Licari, M.K.; Rogasch, N.C.;
Middeldorp, C.M.; Clark, S.R.; Vallence, A.M.; Boulton, K.A.; Hickie, I.B.;
Whitehouse, A.J.O.; Galletly, C.; Alvares, G.A.; Fujiyama, H.; Heussler, H.;
Craig, J.M.; Kirkovski, M.; Mills, N.T.; Rinehart, N.J.; Donaldson, P.H.; Ford,
T.C.; Caeyenberghs, K.; Albein-Urios, N.; Bekkali, S.; Fitzgerald, P.B. Repetitive
transcranial magnetic stimulation (rTMS) in autism spectrum disorder: Protocol
for a multicentre randomised controlled clinical trial. BMJ Open, 2021, 11(7),
e046830. http://dx.doi.org/10.1136/bmjopen-2020-046830 PMID:34233985
73) Dai, Y.C.; Zhang, H.F.; Schön, M.; Böckers, T.M.; Han, S.P.; Han, J.S.; Zhang,
R. Neonatal oxytocin treatment ameliorates autistic-like behaviors and oxytocin
deficiency in valproic acid-induced rat model of autism. Front. Cell. Neurosci.,
2018, 12, 355. http://dx.doi.org/10.3389/fncel.2018.00355 PMID:30356897
74) Tyzio, R.; Nardou, R.; Ferrari, D.C.; Tsintsadze, T.; Shahrokhi, A.; Eftekhari, S.;
Khalilov, I.; Tsintsadze, V.; Brouchoud, C.; Chazal, G.; Lemonnier, E.;
Lozovaya, N.; Burnashev, N.; Ben-Ari, Y. Oxytocin-mediated GABA inhibition
during delivery attenuates autism pathogenesis in rodent offspring. Science, 2014,
343(6171), 675-679. http://dx.doi.org/10.1126/science.1247190 PMID:24503856
75) Parker, K.J.; Oztan, O.; Libove, R.A.; Sumiyoshi, R.D.; Jackson, L.P.; Karhson,
D.S.; Summers, J.E.; Hinman, K.E.; Motonaga, K.S.; Phillips, J.M.; Carson, D.S.;
Garner, J.P.; Hardan, A.Y. Intranasal oxytocin treatment for social deficits and
biomarkers of response in children with autism. Proc. Natl. Acad. Sci. USA,
2017, 114(30), 8119-8124. http://dx.doi.org/10.1073/pnas.1705521114
PMID:28696286
76) Bernaerts, S.; Boets, B.; Bosmans, G.; Steyaert, J.; Alaerts, K. Behavioral effects
of multiple-dose oxytocin treatment in autism: A randomized, placebo-controlled
trial with long-term follow-up. Mol. Autism, 2020, 11(1), 6.
http://dx.doi.org/10.1186/s13229-020-0313-1 PMID:31969977
77) Bernaerts, S.; Boets, B.; Steyaert, J.; Wenderoth, N.; Alaerts, K. Oxytocin
treatment attenuates amygdala activity in autism: A treatment-mechanism study
with long-term follow-up. Transl. Psychiatry, 2020, 10(1), 383.
http://dx.doi.org/10.1038/s41398-020-01069-w PMID:33159033
78) Yenkoyan, K.; Harutyunyan, H.; Harutyunyan, A. A certain role of SOD/CAT
imbalance in pathogenesis of autism spectrum disorders. Free Radic. Biol. Med.,
2018, 123, 85-95. http://dx.doi.org/10.1016/j.freeradbiomed.2018.05.070
PMID:29782990
79) Harutyunyan, A.A.; Harutyunyan, H.A.; Yenkoyan, K.B. Novel probable glance
at inflammatory scenario development in autistic pathology. Front. Psychiatry,
2021, 12, 788779. http://dx.doi.org/10.3389/fpsyt.2021.788779 PMID:35002805
80) Manivasagam, T. Role of oxidative stress and antioxidants in autism. In:
Personalized Food Intervention and Therapy for Autism Spectrum Disorder
Management. Advances in Neurobiology; Essa, M.; Qoronfleh, M., Eds.;
Springer: Cham, 2020; vol 24, pp. 193-206. http://dx.doi.org/10.1007/978-3-030-
30402-7_7
81) Robea, M.A.; Jijie, R.; Nicoara, M.; Plavan, G.; Ciobica, A.S.; Solcan, C.; Audira,
G.; Hsiao, C.D.; Strungaru, S.A.; Vitamin, C. Vitamin C attenuates oxidative
stress and behavioral abnormalities triggered by fipronil and pyriproxyfen
insecticide chronic exposure on zebrafish juvenile. Antioxidants, 2020, 9(10),
944. http://dx.doi.org/10.3390/antiox9100944 PMID:33019596
82) McGuinness, G.; Kim, Y. Sulforaphane treatment for autism spectrum disorder: A
systematic review. EXCLI J., 2020, 19, 892-903.
http://dx.doi.org/10.17179/excli2020-2487 PMID:33013262
83) Abraham, D.A.; Undela, K.; Narasimhan, U.; Rajanandh, M.G. Effect of L-
Carnosine in children with autism spectrum disorders: A systematic review and
meta-analysis of randomised controlled trials. Amino Acids, 2021, 53(4), 575-
585. http://dx.doi.org/10.1007/s00726-021-02960-6 PMID:33704575
84) Hajizadeh-Zaker, R.; Ghajar, A.; Mesgarpour, B.; Afarideh, M.; Mohammadi,
M.R.; Akhondzadeh, S. LCarnosine as an adjunctive therapy to risperidone in
children with autistic disorder: A randomized, double-blind, placebo-controlled
trial. J. Child Adolesc. Psychopharmacol., 2018, 28(1), 74-81.
85) Demarquoy, C.; Demarquoy, J. Autism and carnitine: A possible link. World J.
Biol. Chem., 2019, 10(1), 7-16. http://dx.doi.org/10.4331/wjbc.v10.i1.7
PMID:30622681
86) Fahmy, S.F.; El-hamamsy, M.H.; Zaki, O.K.; Badary, O.A. l-Carnitine
supplementation improves the behavioral symptoms in autistic children. Res.
Autism Spectr. Disord., 2013, 7(1), 159-166.
http://dx.doi.org/10.1016/j.rasd.2012.07.006
87) Guevara-Campos, J.; González-Guevara, L.; Guevara-- González, J.; Cauli, O.
First case report of primary carnitine deficiency manifested as intellectual
disability and autism spectrum disorder. Brain Sci., 2019, 9(6), 137.
http://dx.doi.org/10.3390/brainsci9060137 PMID:31200524
88) Shakibaei, F.; Jelvani, D. Effect of adding l-carnitine to risperidone on behavioral,
cognitive, social, and physical symptoms in children and adolescents with autism:
A randomized double-blinded placebo-controlled clinical trial. Clin.
Neuropharmacol., 2023, 46(2), 55-59.
http://dx.doi.org/10.1097/WNF.0000000000000544 PMID:36735565
89) Eeckhaut, V.; Van Immerseel, F.; Croubels, S.; De Baere, S.; Haesebrouck, F.;
Ducatelle, R.; Louis, P.; Vandamme, P. Butyrate production in phylogenetically
diverse Firmicutes isolated from the chicken caecum. Microb. Biotechnol., 2011,
4(4), 503-512. http://dx.doi.org/10.1111/j.1751-7915.2010.00244.x
PMID:21375722
90) Hakalehto, E.; Hänninen, O. Gaseous CO 2 signal initiates growth of butyric-
acid-producing Clostridium butyricum in both pure culture and mixed cultures
with Lactobacillus brevis. Can. J. Microbiol., 2012, 58(7), 928-931.
http://dx.doi.org/10.1139/w2012-059 PMID:22697044
91) Liu, S.; Li, E.; Sun, Z.; Fu, D.; Duan, G.; Jiang, M.; Yu, Y.; Mei, L.; Yang, P.;
Tang, Y.; Zheng, P. Altered gut microbiota and short chain fatty acids in Chinese
children with autism spectrum disorder. Sci. Rep., 2019, 9(1), 287.
http://dx.doi.org/10.1038/s41598-018-36430-z PMID:30670726
92) Kratsman, N.; Getselter, D.; Elliott, E. Sodium butyrate attenuates social behavior
deficits and modifies the transcription of inhibitory/excitatory genes in the frontal
cortex of an autism model. Neuropharmacology, 2016, 102, 136-145.
http://dx.doi.org/10.1016/j.neuropharm.2015.11.003 PMID:26577018
93) Stilling, R.M.; van de Wouw, M.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan,
J.F. The neuropharmacology of butyrate: The bread and butter of the microbiota-
gut-brain axis? Neurochem. Int., 2016, 99, 110-132.
http://dx.doi.org/10.1016/j.neuint.2016.06.011 PMID:27346602
94) Barone, R.; Rizzo, R.; Tabbì, G.; Malaguarnera, M.; Frye, R.E.; Bastin, J. Nuclear
peroxisome proliferator-activated receptors (PPARs) as therapeutic targets of
resveratrol for autism spectrum disorder. Int. J. Mol. Sci., 2019, 20(8), 1878.
http://dx.doi.org/10.3390/ijms20081878 PMID:30995737
95) Deckmann, I.; Schwingel, G.B.; Fontes-Dutra, M.; Bambini-Junior, V.; Gottfried,
C. Neuroimmune alterations in autism: A translational analysis focusing on the
animal model of autism induced by prenatal exposure to valproic acid.
Neuroimmunomodulation, 2018, 25(5-6), 285-299.
http://dx.doi.org/10.1159/000492113 PMID:30157484
96) Kumar, P.; Raman, T.; Swain, M.M.; Mishra, R.; Pal, A. Hyperglycemia-induced
oxidative-nitrosative stress induces inflammation and neurodegeneration via
augmented tuberous sclerosis complex-2 (TSC-2) activation in neuronal cells.
Mol. Neurobiol., 2017, 54(1), 238-254. http://dx.doi.org/10.1007/s12035-015-
9667-3 PMID:26738854
97) Das, A.; Durrant, D.; Koka, S.; Salloum, F.N.; Xi, L.; Kukreja, R.C. Mammalian
target of rapamycin (mTOR) inhibition with rapamycin improves cardiac function
in type 2 diabetic mice: Potential role of attenuated oxidative stress and altered
contractile protein expression. J. Biol. Chem., 2014, 289(7), 4145-4160.
http://dx.doi.org/10.1074/jbc.M113.521062 PMID:24371138
98) Kotajima-Murakami, H.; Kobayashi, T.; Kashii, H.; Sato, A.; Hagino, Y.; Tanaka,
M.; Nishito, Y.; Takamatsu, Y.; Uchino, S.; Ikeda, K. Effects of rapamycin on
social interaction deficits and gene expression in mice exposed to valproic acid in
utero. Mol. Brain, 2019, 12(1), 3. http://dx.doi.org/10.1186/s13041-018-0423-2
PMID:30621732
99) Schafer, F.Q.; Buettner, G.R. Redox environment of the cell as viewed through
the redox state of the glutathione disulfide/glutathione couple. Free Radic. Biol.
Med., 2001, 30(11), 1191-1212. http://dx.doi.org/10.1016/S0891-5849(01)00480-
4PMID:11368918
100) Bala, K.A.; Doğan, M.; Mutluer, T.; Kaba, S.; Aslan, O.; Balahoroğlu, R.;
Çokluk, E.; Üstyol, L.; Kocaman, S. Plasma amino acid profile in autism
spectrum disorder (ASD). Eur. Rev. Med. Pharmacol. Sci., 2016, 20(5), 923-929.
PMID:27010152
101) Moretti, P.; Peters, S.U.; del Gaudio, D.; Sahoo, T.; Hyland, K.; Bottiglieri, T.;
Hopkin, R.J.; Peach, E.; Min, S.H.; Goldman, D.; Roa, B.; Bacino, C.A.; Scaglia,
F. Brief report: Autistic symptoms, developmental regression, mental retardation,
epilepsy, and dyskinesias in CNS folate deficiency. J. Autism Dev. Disord., 2008,
38(6), 1170-1177. http://dx.doi.org/10.1007/s10803-007-0492-z PMID:18027081
102) Zhang, Z.; Yu, L.; Li, S.; Liu, J. Association study of polymorphisms in genes
relevant to vitamin B12 and folate metabolism with childhood autism spectrum
disorder in a han chinese population. Med. Sci. Monit., 2018, 24, 370-376.
http://dx.doi.org/10.12659/MSM.905567 PMID:29348398
103) Bertoglio, K.; Jill James, S.; Deprey, L.; Brule, N.; Hendren, R.L. Pilot study of
the effect of methyl B12 treatment on behavioral and biomarker measures in
children with autism. J. Altern. Complement. Med., 2010, 16(5), 555-560.
http://dx.doi.org/10.1089/acm.2009.0177 PMID:20804367
104) Zhang, Y.; Hodgson, N.W.; Trivedi, M.S.; Abdolmaleky, H.M.; Fournier, M.;
Cuenod, M.; Do, K.Q.; Deth, R.C. Decreased brain levels of vitamin B12 in
aging, autism and schizophrenia. PLoS One, 2016, 11(1), e0146797.
http://dx.doi.org/10.1371/journal.pone.0146797 PMID:26799654
105) Frye, R.E.; Slattery, J.C.; Quadros, E.V. Folate metabolism abnormalities in
autism: Potential biomarkers. Biomarkers Med., 2017, 11(8), 687-699.
http://dx.doi.org/10.2217/bmm-2017-0109 PMID:28770615
106) James, S.J.; Melnyk, S.; Fuchs, G.; Reid, T.; Jernigan, S.; Pavliv, O.; Hubanks,
A.; Gaylor, D.W. Efficacy of methylcobalamin and folinic acid treatment on
glutathione redox status in children with autism. Am. J. Clin. Nutr., 2009, 89(1),
425-430. http://dx.doi.org/10.3945/ajcn.2008.26615 PMID:19056591
107) An, S.; Feng, X.; Dai, Y.; Bo, H.; Wang, X.; Li, M.; Woo, J.Z.; Liang, X.; Guo,
C.; Liu, C.X.; Wei, L. Development and evaluation of a speech-generating AAC
mobile app for minimally verbal children with autism spectrum disorder in
Mainland China. Mol. Autism, 2017, 8(1), 52. http://dx.doi.org/10.1186/s13229-
017-0165-5 PMID:29026509
108) Sun, C.; Zou, M.; Zhao, D.; Xia, W.; Wu, L. Efficacy of folic acid
supplementation in autistic children participating in structured teaching: An open-
label trial. Nutrients, 2016, 8(6), 337.
http://dx.doi.org/10.3390/nu8060337PMID:27338456
109) Castro, K.; Klein, L.S.; Baronio, D.; Gottfried, C.; Riesgo, R.; Perry, I.S. Folic
acid and autism: What do we know? Nutr. Neurosci., 2016, 19(7), 310-317.
http://dx.doi.org/10.1179/1476830514Y.0000000142PMID:25087906
110) Kang, D.W.; Adams, J.B.; Gregory, A.C.; Borody, T.; Chittick, L.; Fasano, A.;
Khoruts, A.; Geis, E.; Maldonado, J.; McDonough-Means, S.; Pollard, E.L.;
Roux, S.; Sadowsky, M.J.; Lipson, K.S.; Sullivan, M.B.; Caporaso, J.G.;
Krajmalnik-Brown, R. Microbiota transfer therapy alters gut ecosystem and
improves gastrointestinal and autism symptoms: An open-label study.
Microbiome, 2017, 5(1), 10. http://dx.doi.org/10.1186/s40168-016-0225-7
PMID:28122648
111) Gondalia, S.V.; Palombo, E.A.; Knowles, S.R.; Cox, S.B.; Meyer, D.; Austin,
D.W. Molecular characterisation of gastrointestinal microbiota of children with
autism (with and without gastrointestinal dysfunction) and their neurotypical
siblings. Autism Res., 2012, 5(6), 419-427. http://dx.doi.org/10.1002/aur.1253
PMID:22997101
112) Chaidez, V.; Hansen, R.L.; Hertz-Picciotto, I. Gastrointestinal problems in
children with autism, developmental delays or typical development. J. Autism
Dev. Disord., 2014, 44(5), 1117-1127. http://dx.doi.org/10.1007/s10803-013-
1973-x PMID:24193577
113) Santocchi, E.; Guiducci, L.; Fulceri, F.; Billeci, L.; Buzzigoli, E.; Apicella, F.;
Calderoni, S.; Grossi, E.; Morales, M.A.; Muratori, F. Gut to brain interaction in
autism spectrum disorders: A randomized controlled trial on the role of probiotics
on clinical, biochemical and neurophysiological parameters. BMC Psychiatry,
2016, 16(1), 183. http://dx.doi.org/10.1186/s12888-016-0887-5 PMID:27260271
114) Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut
microbiota on brain and behaviour. Nat. Rev. Neurosci., 2012, 13(10), 701-712.
http://dx.doi.org/10.1038/nrn3346 PMID:22968153
115) McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal
symptoms in autism spectrum disorder: A meta-analysis. Pediatrics, 2014, 133(5),
872-883. http://dx.doi.org/10.1542/peds.2013-3995 PMID:24777214
116) Rao, G.M. Effects of prebiotics, probiotics intervention in children with autism
spectrum disorder: A systematic review. Biomedicine, 2020, 20, 119-122.
117) Strati, F.; Cavalieri, D.; Albanese, D.; De Felice, C.; Donati, C.; Hayek, J.;
Jousson, O.; Leoncini, S.; Renzi, D.; Calabrò, A.; De Filippo, C. New evidences
on the altered gut microbiota in autism spectrum disorders. Microbiome, 2017,
5(1), 24. http://dx.doi.org/10.1186/s40168-017-0242-1 PMID:28222761
118) Dhakal, R.; Bajpai, V.K.; Baek, K.H. Production of gaba (γ - aminobutyric acid)
by microorganisms: A review. Braz. J. Microbiol., 2012, 43(4), 1230-1241.
http://dx.doi.org/10.1590/S1517-83822012000400001 PMID:24031948
119) El-Ansary, A.; Bacha, A.B.; Bjørklund, G.; Al-Orf, N.; Bhat, R.S.; Moubayed, N.;
Abed, K. Probiotic treatment reduces the autistic-like excitation/inhibition
imbalance in juvenile hamsters induced by orally administered propionic acid and
clindamycin. Metab. Brain Dis., 2018, 33(4), 1155-1164.
http://dx.doi.org/10.1007/s11011-018-0212-8 PMID:29582256
120) Ait-Belgnaoui, A.; Colom, A.; Braniste, V.; Ramalho, L.; Marrot, A.; Cartier, C.;
Houdeau, E.; Theodorou, V.; Tompkins, T. Probiotic gut effect prevents the
chronic psychological stress-induced brain activity abnormality in mice.
Neurogastroenterol. Motil., 2014, 26(4), 510-520.
http://dx.doi.org/10.1111/nmo.12295 PMID:24372793
121) Sanctuary, M.R.; Kain, J.N.; Angkustsiri, K.; German, J.B. Dietary considerations
in autism spectrum disorders: The potential role of protein digestion and
microbial putrefaction in the gut-brain axis. Front. Nutr., 2018, 5, 40.
http://dx.doi.org/10.3389/fnut.2018.00040 PMID:29868601
122) Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejía, J.L.;
Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A prebiotic
intervention study in children with autism spectrum disorders (ASDs).
Microbiome, 2018, 6(1), 133. http://dx.doi.org/10.1186/s40168-018-0523-3
PMID:30071894
123) Meguid, N.A.; Mawgoud, Y.I.A.; Bjørklund, G.; Mehanne, N.S.; Anwar, M.;
Effat, B.A.E.K.; Chirumbolo, S.; Elrahman, M.M.A. Molecular characterization
of probiotics and their influence on children with autism spectrum disorder. Mol.
Neurobiol., 2022, 59(11), 6896-6902. http://dx.doi.org/10.1007/s12035-022-
02963-8 PMID:36050597
124) MacFabe, D.F. Short-chain fatty acid fermentation products of the gut
microbiome: Implications in autism spectrum disorders. Microb. Ecol. Health
Dis., 2012, 23(0) http://dx.doi.org/10.3402/mehd.v23i0.19260 PMID:23990817
125) Sanctuary, M.R.; Kain, J.N.; Chen, S.Y.; Kalanetra, K.; Lemay, D.G.; Rose, D.R.;
Yang, H.T.; Tancredi, D.J.; German, J.B.; Slupsky, C.M.; Ashwood, P.; Mills,
D.A.; Smilowitz, J.T.; Angkustsiri, K. Pilot study of probiotic/- colostrum
supplementation on gut function in children with autism and gastrointestinal
symptoms. PLoS One, 2019, 14(1), e0210064.
http://dx.doi.org/10.1371/journal.pone.0210064 PMID:30625189
126) Witters, P.; Debbold, E.; Crivelly, K.; Vande Kerckhove, K.; Corthouts, K.;
Debbold, B.; Andersson, H.; Vannieuwenborg, L.; Geuens, S.; Baumgartner, M.;
Kozicz, T.; Settles, L.; Morava, E. Autism in patients with propionic acidemia.
Mol. Genet. Metab., 2016, 119(4), 317-321.
http://dx.doi.org/10.1016/j.ymgme.2016.10.009 PMID:27825584
127) Choi, J.; Lee, S.; Won, J.; Jin, Y.; Hong, Y.; Hur, T.Y.; Kim, J.H.; Lee, S.R.;
Hong, Y. Pathophysiological and neurobehavioral characteristics of a propionic
acid-mediated autism-like rat model. PLoS One, 2018, 13(2), e0192925.
http://dx.doi.org/10.1371/journal.pone.0192925 PMID:29447237
128) Frye, R.E.; Rose, S.; Slattery, J.; MacFabe, D.F. Gastrointestinal dysfunction in
autism spectrum disorder: The role of the mitochondria and the enteric
microbiome. Microb. Ecol. Health Dis., 2015, 26(0), 27458.
http://dx.doi.org/10.3402/mehd.v26.27458 PMID:25956238
129) Williams, B.L.; Hornig, M.; Buie, T.; Bauman, M.L.; Cho Paik, M.; Wick, I.;
Bennett, A.; Jabado, O.; Hirschberg, D.L.; Lipkin, W.I. Impaired carbohydrate
digestion and transport and mucosal dysbiosis in the intestines of children with
autism and gastrointestinal disturbances. PLoS One, 2011, 6(9), e24585.
http://dx.doi.org/10.1371/journal.pone.0024585 PMID:21949732
130) Tomova, A.; Husarova, V.; Lakatosova, S.; Bakos, J.; Vlkova, B.; Babinska, K.;
Ostatnikova, D. Gastrointestinal microbiota in children with autism in Slovakia.
Physiol. Behav., 2015, 138, 179-187.
http://dx.doi.org/10.1016/j.physbeh.2014.10.033 PMID:25446201
131) Hogan, S.; O’Gara, J.P.; O’Neill, E. Novel treatment of Staphylococcus aureus
device-related infections using fibrinolytic agents. Antimicrob. Agents
Chemother., 2018, 62(2), e02008-17. http://dx.doi.org/10.1128/AAC.02008-17
PMID:29203484
132) Zapotoczna, M.; McCarthy, H.; Rudkin, J.K.; O’Gara, J.P.; O’Neill, E. An
essential role for coagulase in Staphylococcus aureus biofilm development reveals
new therapeutic possibilities for device-related infections. J. Infect. Dis., 2015,
212(12), 1883-1893. http://dx.doi.org/10.1093/infdis/jiv319 PMID:26044292
133) Chang, Y.; Gu, W.; McLandsborough, L. Low concentration of
ethylenediaminetetraacetic acid (EDTA) affects biofilm formation of Listeria
monocytogenes by inhibiting its initial adherence. Food Microbiol., 2012, 29(1),
10-17. http://dx.doi.org/10.1016/j.fm.2011.07.009 PMID:22029913
134) Miyazaki, Y.; Yokota, H.; Takahashi, H.; Fukuda, M.; Kawakami, H.; Kamiya,
S.; Hanawa, T. Effect of probiotic bacterial strains of Lactobacillus,
Bifidobacterium, and Enterococcus on Enteroaggregative Escherichia coli. J.
Infect. Chemother., 2010, 16(1), 10-18. http://dx.doi.org/10.1007/s10156-009-
0007-2 PMID:20054601
135) Panksepp, J. A neurochemical theory of autism. Trends Neurosci., 1979, 2, 174-
177. http://dx.doi.org/10.1016/0166-2236(79)90071-7
136) Gillberg, C.; Terenius, L.; Lönnerholm, G. Endorphin activity in childhood
psychosis. Spinal fluid levels in 24 cases. Arch. Gen. Psychiatry, 1985, 42(8),
780-783. http://dx.doi.org/10.1001/archpsyc.1985.01790310042005
PMID:4015322
137) Guareschi Cazzullo, A.; Musetti, M.C.; Musetti, L.; Bajo, S.; Sacerdote, P.;
Panerai, A. β-Endorphin levels in peripheral blood mononuclear cells and long-
term naltrexone treatment in autistic children. Eur. Neuropsychopharmacol.,
1999, 9(4), 361-366. http://dx.doi.org/10.1016/S0924-977X(99)00010-3
PMID:10422898
138) Zioudrou, C.; Streaty, R.A.; Klee, W.A. Opioid peptides derived from food
proteins. The exorphins. J. Biol. Chem., 1979, 254(7), 2446-2449.
http://dx.doi.org/10.1016/S0021-9258(17)30243-0 PMID:372181
139) Whiteley, P.; Shattock, P. Biochemical aspects in autism spectrum disorders:
Updating the opioid-excess theory and presenting new opportunities for
biomedical intervention. Expert Opin. Ther. Targets, 2002, 6(2), 175-183.
http://dx.doi.org/10.1517/14728222.6.2.175 PMID:12223079
140) Camarca, A.; Anderson, R.P.; Mamone, G.; Fierro, O.; Facchiano, A.; Costantini,
S.; Zanzi, D.; Sidney, J.; Auricchio, S.; Sette, A.; Troncone, R.; Gianfrani, C.
Intestinal T cell responses to gluten peptides are largely heterogeneous:
Implications for a peptide-based therapy in celiac disease. J. Immunol., 2009,
182(7), 4158-4166. http://dx.doi.org/10.4049/jimmunol.0803181 PMID:19299713
141) Catassi, C.; Fasano, A. Celiac disease. Curr. Opin. Gastroenterol., 2008, 24(6),
687-691. http://dx.doi.org/10.1097/MOG.0b013e32830edc1e PMID:19122516
142) Ghalichi, F.; Ghaemmaghami, J.; Malek, A.; Ostadrahimi, A. Effect of gluten free
diet on gastrointestinal and behavioral indices for children with autism spectrum
disorders: A randomized clinical trial. World J. Pediatr., 2016, 12(4), 436-442.
http://dx.doi.org/10.1007/s12519-016-0040-z PMID:27286693
143) Pennesi, C.M.; Klein, L.C. Effectiveness of the glutenfree, casein-free diet for
children diagnosed with autism spectrum disorder: Based on parental report. Nutr.
Neurosci., 2012, 15(2), 85-91.
http://dx.doi.org/10.1179/1476830512Y.0000000003 PMID:22564339
144) Johnson, C.R.; Handen, B.L.; Zimmer, M.; Sacco, K.; Turner, K. Effects of gluten
free / casein free diet in young children with autism: A pilot study. J. Dev. Phys.
Disabil., 2011, 23(3), 213-225. http://dx.doi.org/10.1007/s10882-010-9217-x
145) Elder, J.H.; Shankar, M.; Shuster, J.; Theriaque, D.; Burns, S.; Sherrill, L. The
gluten-free, casein-free diet in autism: Results of a preliminary double blind
clinical trial. J. Autism Dev. Disord., 2006, 36(3), 413-420.
http://dx.doi.org/10.1007/s10803-006-0079-0 PMID:16555138
146) Lange, K.W.; Hauser, J.; Reissmann, A. Gluten-free and casein-free diets in the
therapy of autism. Curr. Opin. Clin. Nutr. Metab. Care, 2015, 18(6), 572-575.
http://dx.doi.org/10.1097/MCO.0000000000000228 PMID:26418822
147) Desai, A.; Sequeira, J.M.; Quadros, E.V. Prevention of behavioral deficits in rats
exposed to folate receptor antibodies: Implication in autism. Mol. Psychiatry,
2017, 22(9), 1291-1297. http://dx.doi.org/10.1038/mp.2016.153 PMID:27646260
148) Castro, K.; Baronio, D.; Perry, I.S.; Riesgo, R.S.; Gottfried, C. The effect of
ketogenic diet in an animal model of autism induced by prenatal exposure to
valproic acid. Nutr. Neurosci., 2017, 20(6), 343-350.
http://dx.doi.org/10.1080/1028415X.2015.1133029 PMID:26856821
149) Spilioti, M.; Evangeliou, A.E.; Tramma, D.; Theodoridou, Z.; Metaxas, S.;
Michailidi, E.; Bonti, E.; Frysira, H.; Haidopoulou, A.; Asprangathou, D.;
Tsalkidis, A.J.; Kardaras, P.; Wevers, R.A.; Jakobs, C.; Gibson, K.M. Evidence
for treatable inborn errors of metabolism in a cohort of 187 Greek patients with
autism spectrum disorder (ASD). Front. Hum. Neurosci., 2013, 7, 858.
http://dx.doi.org/10.3389/fnhum.2013.00858 PMID:24399946
150) El-Rashidy, O.; El-Baz, F.; El-Gendy, Y.; Khalaf, R.; Reda, D.; Saad, K.
Ketogenic diet versus gluten free casein free diet in autistic children: A case-
control study. Metab. Brain Dis., 2017, 32(6), 1935-1941.
http://dx.doi.org/10.1007/s11011-017-0088-z PMID:28808808
151) Evangeliou, A.; Vlachonikolis, I.; Mihailidou, H.; Spilioti, M.; Skarpalezou, A.;
Makaronas, N.; Prokopiou, A.; Christodoulou, P.; Liapi-Adamidou, G.; Helidonis,
E.; Sbyrakis, S.; Smeitink, J. Application of a ketogenic diet in children with
autistic behavior: Pilot study. J. Child Neurol., 2003, 18(2), 113-118.
http://dx.doi.org/10.1177/08830738030180020501 PMID:12693778
152) Hartman, R.E.; Patel, D. Dietary approaches to the management of autism
spectrum disorders. Advances in Neurobiology; Springer, 2020, Vol. 24, pp. 547-
571. http://dx.doi.org/10.1007/978-3-030-30402-7_19
153) Nurchi, V.M.; Buha Djordjevic, A.; Crisponi, G.; Alexander, J.; Bjørklund, G.;
Aaseth, J. Arsenic toxicity: Molecular targets and therapeutic agents.
Biomolecules, 2020, 10(2), 235. http://dx.doi.org/10.3390/biom10020235
PMID:32033229
154) Bjørklund, G.; Crisponi, G.; Nurchi, V.M.; Cappai, R.; Buha Djordjevic, A.;
Aaseth, J. A review on coordination properties of thiol-containing chelating
agents towards mercury, cadmium, and lead. Molecules, 2019, 24(18), 3247.
http://dx.doi.org/10.3390/molecules24183247 PMID:31489907
155) Bjørklund, G.; Mutter, J.; Aaseth, J. Metal chelators and neurotoxicity: Lead,
mercury, and arsenic. Arch. Toxicol., 2017, 91(12), 3787-3797.
http://dx.doi.org/10.1007/s00204-017-2100-0 PMID:29063135
156) Yassa, H.A. Autism: A form of lead and mercury toxicity. Environ. Toxicol.
Pharmacol., 2014, 38(3), 1016-1024. http://dx.doi.org/10.1016/j.etap.2014.10.005
PMID:25461563
157) James, S.; Stevenson, S.W.; Silove, N.; Williams, K. Chelation for autism
spectrum disorder (ASD). Cochrane Libr., 2015, 2016(10), CD010766.
http://dx.doi.org/10.1002/14651858.CD010766.pub2 PMID:26114777
158) T Schultz, S.; G Gould, G. Acetaminophen use for fever in children associated
with autism spectrum disorder. Autism Open Access, 2016, 6(2), 170.
http://dx.doi.org/10.4172/2165-7890.1000170 PMID:27695658
159) Wang, T.; Shan, L.; Du, L.; Feng, J.; Xu, Z.; Staal, W.G.; Jia, F. Serum
concentration of 25-hydroxyvitamin D in autism spectrum disorder: A systematic
review and meta-analysis. Eur. Child Adolesc. Psychiatry, 2016, 25(4), 341-350.
http://dx.doi.org/10.1007/s00787-015-0786-1 PMID:26514973
160) Fernell, E.; Bejerot, S.; Westerlund, J.; Miniscalco, C.; Simila, H.; Eyles, D.;
Gillberg, C.; Humble, M.B. Autism spectrum disorder and low vitamin D at birth:
A sibling control study. Mol. Autism, 2015, 6(1), 3.
http://dx.doi.org/10.1186/2040-2392-6-3 PMID:25874075
161) Huang, Y.N.; Ho, Y.J.; Lai, C.C.; Chiu, C.T.; Wang, J.Y. 1,25-Dihydroxyvitamin
D3 attenuates endotoxin-induced production of inflammatory mediators by
inhibiting MAPK activation in primary cortical neuron-glia cultures. J.
Neuroinflammation, 2015, 12(1), 147. http://dx.doi.org/10.1186/s12974-015-
0370-0 PMID:26259787
162) Patrick, R.P.; Ames, B.N.; Vitamin, D. Vitamin D hormone regulates serotonin
synthesis. Part 1: Relevance for autism. FASEB J., 2014, 28(6), 2398-2413.
http://dx.doi.org/10.1096/fj.13-246546 PMID:24558199
163) Saad, K.; Abdel-Rahman, A.A.; Elserogy, Y.M.; Al-Atram, A.A.; El-Houfey,
A.A.; Othman, H.A.K.; Bjørklund, G.; Jia, F.; Urbina, M.A.; Abo-Elela, M.G.M.;
Ahmad, F.A.; Abd El-Baseer, K.A.; Ahmed, A.E.; Abdel-Salam, A.M. Retracted:
Randomized controlled trial of vitamin D supplementation in children with autism
spectrum disorder. J. Child Psychol. Psychiatry, 2018, 59(1), 20-29.
http://dx.doi.org/10.1111/jcpp.12652 PMID:27868194
164) Kostiukow, A.; Samborski, W. The effectiveness of hyperbaric oxygen therapy
(HBOT) in children with autism spectrum disorders. Pol. Merkur. Lekarski, 2020,
48(283), 15-18.
165) Rossignol, D.A.; Rossignol, L.W.; Smith, S.; Schneider, C.; Logerquist, S.;
Usman, A.; Neubrander, J.; Madren, E.M.; Hintz, G.; Grushkin, B.; Mumper,
E.A. Hyperbaric treatment for children with autism: A multicenter, randomized,
double-blind, controlled trial. BMC Pediatr., 2009, 9(1), 21.
http://dx.doi.org/10.1186/1471-2431-9-21 PMID:19284641
166) Sakulchit, T.; Ladish, C.; Goldman, R.D. Hyperbaric oxygen therapy for children
with autism spectrum disorder. Can. Fam. Physician, 2017, 63(6), 446-448.
PMID:28615394
167) Choi, S.; Hong, D.K.; Choi, B.Y.; Suh, S.W. Zinc in the brain: Friend or foe? Int.
J. Mol. Sci., 2020, 21(23), 8941. http://dx.doi.org/10.3390/ijms21238941
PMID:33255662
168) Bitanihirwe, B.K.Y.; Cunningham, M.G. Zinc: The brain’s dark horse. Synapse,
2009, 63(11), 1029-1049. http://dx.doi.org/10.1002/syn.20683 PMID:19623531
169) Cope, E.C.; Levenson, C.W. Role of zinc in the development and treatment of
mood disorders. Curr. Opin. Clin. Nutr. Metab. Care, 2010, 13(6), 685-689.
http://dx.doi.org/10.1097/MCO.0b013e32833df61a PMID:20689416
170) Russo, A. J. Decreased zinc and increased copper in individuals with anxiety.
Nutr Metab Insights, 2011, 4, 1-5. http://dx.doi.org/10.4137/NMI.S6349
171) Krall, R.; Gale, J.R.; Ross, M.M.; Tzounopoulos, T.; Aizenman, E. Intracellular
zinc signaling influences NMDA receptor function by enhancing the interaction
of ZnT1 with GluN2A. Neurosci. Lett., 2022, 790, 136896.
http://dx.doi.org/10.1016/j.neulet.2022.136896 PMID:36202195
172) Miyata, S.; Nagata, H.; Yamao, S.; Nakamura, S.; Kameyama, M. Dopamine-β-
hydroxylase activities in serum and cerebrospinal fluid of aged and demented
patients. J. Neurol. Sci., 1984, 63(3), 403-409. http://dx.doi.org/10.1016/0022-
510X(84)90163-1PMID:6726279
173) Skalny, A.V.; Simashkova, N.V.; Klyushnik, T.P.; Grabeklis, A.R.; Radysh, I.V.;
Skalnaya, M.G.; Nikonorov, A.A.; Tinkov, A.A. Assessment of serum trace
elements and electrolytes in children with childhood and atypical autism. J. Trace
Elem. Med. Biol., 2017, 43, 9-14. http://dx.doi.org/10.1016/j.jtemb.2016.09.009
PMID:27707611
174) Wu, H.; Zhao, G.; Liu, S.; Zhang, Q.; Wang, P.; Cao, Y.; Wu, L. Supplementation
with selenium attenuates autism-- like behaviors and improves oxidative stress,
inflammation and related gene expression in an autism disease model. J. Nutr.
Biochem., 2022, 107, 109034. http://dx.doi.org/10.1016/j.jnutbio.2022.109034
PMID:35500829
175) Skalny, A.V.; Skalnaya, M.G.; Bjørklund, G.; Gritsenko, V.A.; Aaseth, J.;
Tinkov, A.A. Selenium and autism spectrum disorder. In: Selenium. Molecular
and Integrative Toxicology; Springer: Cham, 2018; pp. 193-210.
http://dx.doi.org/10.1007/978-3-319-95390-8_10
176) Raymond, L.J.; Deth, R.C.; Ralston, N.V.C. Potential role of selenoenzymes and
antioxidant metabolism in relation to autism etiology and pathology. Autism Res.
Treat., 2014, 2014, 1-15. http://dx.doi.org/10.1155/2014/164938 PMID:24734177
177) Bjørklund, G.; Aaseth, J.; Ajsuvakova, O.P.; Nikonorov, A.A.; Skalny, A.V.;
Skalnaya, M.G.; Tinkov, A.A. Molecular interaction between mercury and
selenium in neurotoxicity. Coord. Chem. Rev., 2017, 332, 30-37.
http://dx.doi.org/10.1016/j.ccr.2016.10.009
178) Bjørklund, G. Selenium as an antidote in the treatment of mercury intoxication.
Biometals, 2015, 28(4), 605-614. http://dx.doi.org/10.1007/s10534-015-9857-5
PMID:25947386
179) El-Ansary, A.; Bjørklund, G.; Tinkov, A.A.; Skalny, A.V.; Al Dera, H.
Relationship between selenium, lead, and mercury in red blood cells of Saudi
autistic children. Metab. Brain Dis., 2017, 32(4), 1073-1080.
http://dx.doi.org/10.1007/s11011-017-9996-1 PMID:28326463
180) Kirkland, A.; Sarlo, G.; Holton, K. The role of magnesium in neurological
disorders. Nutrients, 2018, 10(6), 730. http://dx.doi.org/10.3390/nu10060730
PMID:29882776
181) Yasuda, H.; Tsutsui, T. Assessment of infantile mineral imbalances in autism
spectrum disorders (ASDs). Int. J. Environ. Res. Public Health, 2013, 10(11),
6027-6043. http://dx.doi.org/10.3390/ijerph10116027 PMID:24284360
182) Beto, J.A. The role of calcium in human aging. Clin. Nutr. Res., 2015, 4(1), 1-8.
http://dx.doi.org/10.7762/cnr.2015.4.1.1 PMID:25713787
183) Nguyen, R.L.; Medvedeva, Y.V.; Ayyagari, T.E.; Schmunk, G.; Gargus, J.J.
Intracellular calcium dysregulation in autism spectrum disorder: An analysis of
converging organelle signaling pathways. Biochim. Biophys. Acta Mol. Cell Res.,
2018, 1865(11), 1718-1732. http://dx.doi.org/10.1016/j.bbamcr.2018.08.003
PMID: 30992134
184) Chen, L.; Shi, X.J.; Liu, H.; Mao, X.; Gui, L.N.; Wang, H.; Cheng, Y. Oxidative
stress marker aberrations in children with autism spectrum disorder: A systematic
review and meta-analysis of 87 studies (N = 9109). Transl. Psychiatry, 2021,
11(1), 15. http://dx.doi.org/10.1038/s41398-020-01135-3 PMID: 33414386
185) Vetter, T.; Lohse, M.J. Magnesium and the parathyroid. Curr. Opin. Nephrol.
Hypertens., 2002, 11(4), 403-410. http://dx.doi.org/10.1097/00041552-
200207000-00006 PMID: 12105390
186) Pangrazzi, L.; Balasco, L.; Bozzi, Y. Oxidative stress and immune system
dysfunction in autism spectrum disorders. Int. J. Mol. Sci., 2020, 21(9), 3293.
http://dx.doi.org/10.3390/ijms21093293 PMID: 32384730
187) Saghazadeh, A.; Ahangari, N.; Hendi, K.; Saleh, F.; Rezaei, N. Status of essential
elements in autism spectrum disorder: Systematic review and meta-analysis. Rev.
Neurosci., 2017, 28(7), 783-809. http://dx.doi.org/10.1515/revneuro-2017-0015
PMID: 28665792
188) Skalny, A.V.; Mazaletskaya, A.L.; Ajsuvakova, O.P.; Bjørklund, G.; Skalnaya,
M.G.; Chernova, L.N.; Skalny, A.A.; Tinkov, A.A. Magnesium status in children
with attention-deficit/hyperactivity disorder and/or autism spectrum disorder. J.
Korean Acad. Child Adolesc. Psychiatry, 2020, 31(1), 41-45.
http://dx.doi.org/10.5765/jkacap.190036 PMID: 32612412
189) Uwitonze, A.M.; Razzaque, M.S. Role of magnesium in vitamin D activation and
function. J. Am. Osteopath. Assoc., 2018, 118(3), 181-189.
http://dx.doi.org/10.7556/jaoa.2018.037 PMID: 29480918
190) Muir, K.W. Magnesium in stroke treatment. Postgrad. Med. J., 2002, 78(925),
641-645. http://dx.doi.org/10.1136/pmj.78.925.641 PMID: 12496316
191) Schmunk, G.; Gargus, J.J. Channelopathy pathogenesis in autism spectrum
disorders. Front. Genet., 2013, 4, 222. http://dx.doi.org/10.3389/fgene.2013.00222
PMID: 24204377
192) Martineau, J.; Barthelemy, C.; Garreau, B.; Lelord, G. Vitamin B6, magnesium,
and combined B6-Mg: Therapeutic effects in childhood autism. Biol. Psychiatry,
1985, 20(5), 467-478. http://dx.doi.org/10.1016/0006-3223(85)90019-8 PMID:
3886023
193) Bjørklund, G.; Waly, M.I.; Al-Farsi, Y.; Saad, K.; Dadar, M.; Rahman, M.M.;
Elhoufey, A.; Chirumbolo, S.; Jóźwik-Pruska, J.; Kałużna-Czaplińska, J. The role
of vitamins in autism spectrum disorder: What do we know? J. Mol. Neurosci.,
2019, 67(3), 373-387. http://dx.doi.org/10.1007/s12031-018-1237-5 PMID:
30607900
194) Saad, K.; Abdel-rahman, A.A.; Elserogy, Y.M.; Al-Atram, A.A.; Cannell, J.J.;
Bjørklund, G.; Abdel-Reheim, M.K.; Othman, H.A.K.; El-Houfey, A.A.; Abd El-
Aziz, N.H.R.; Abd El-Baseer, K.A.; Ahmed, A.E.; Ali, A.M. Vitamin D status in
autism spectrum disorders and the efficacy of vitamin D supplementation in
autistic children. Nutr. Neurosci., 2016, 19(8), 346-351.
http://dx.doi.org/10.1179/1476830515Y.0000000019 PMID: 25876214
195) Chirumbolo, S.; Bjørklund, G.; Sboarina, A.; Vella, A. The role of vitamin D in
the immune system as a pro-survival molecule. Clin. Ther., 2017, 39(5), 894-916.
http://dx.doi.org/10.1016/j.clinthera.2017.03.021 PMID:28438353
196) Patrick, R.P.; Ames, B.N. Vitamin D and the omega-3 fatty acids control
serotonin synthesis and action, part 2: Relevance for ADHD, bipolar disorder,
schizophrenia, and impulsive behavior. FASEB J., 2015, 29(6), 2207-2222.
http://dx.doi.org/10.1096/fj.14-268342 PMID: 25713056
197) Cui, X.; Eyles, D.W. Vitamin D and the central nervous system: Causative and
preventative mechanisms in brain disorders. Nutrients, 2022, 14(20), 4353.
http://dx.doi.org/10.3390/nu14204353 PMID: 36297037
198) Zastre, J.A.; Sweet, R.L.; Hanberry, B.S.; Ye, S. Linking vitamin B1 with cancer
cell metabolism. Cancer Metab., 2013, 1(1), 16. http://dx.doi.org/10.1186/2049-
3002-1-16 PMID: 24280319
199) Jung, H.Y.; Kwon, H.J.; Kim, W.; Nam, S.M.; Kim, J.W.; Hahn, K.R.; Yoo,
D.Y.; Yoon, Y.S.; Choi, S.Y.; Kim, D.W.; Hwang, I.K. Role of pyridoxine in
GABA synthesis and degradation in the hippocampus. Tissue Cell, 2019, 61, 72-
78. http://dx.doi.org/10.1016/j.tice.2019.09.005 PMID: 31759410
200) Stover, P.J.; Field, M.S. Vitamin B-6. Adv. Nutr., 2015, 6(1), 132-133.
http://dx.doi.org/10.3945/an.113.005207 PMID: 25593152 Scott, J.M. Folate and
vitamin B 12
201) Proc. Nutr. Soc., 1999, 58(2), 441-448.
http://dx.doi.org/10.1017/S0029665199000580 PMID: 10466189
202) Bjørklund, G.; Doşa, M.D.; Maes, M.; Dadar, M.; Frye, R.E.; Peana, M.;
Chirumbolo, S. The impact of glutathione metabolism in autism spectrum
disorder. Pharmacol. Res., 2021, 166, 105437.
http://dx.doi.org/10.1016/j.phrs.2021.105437 PMID: 33493659
203) Bjørklund, G.; Tinkov, A.A.; Hosnedlová, B.; Kizek, R.; Ajsuvakova, O.P.;
Chirumbolo, S.; Skalnaya, M.G.; Peana, M.; Dadar, M.; El-Ansary, A.; Qasem,
H.; Adams, J.B.; Aaseth, J.; Skalny, A.V. The role of glutathione redox imbalance
in autism spectrum disorder: A review. Free Radic. Biol. Med., 2020, 160, 149-
162. http://dx.doi.org/10.1016/j.freeradbiomed.2020.07.017 PMID: 32743
204) Bjørklund, G.; Meguid, N.A.; El-Bana, M.A.; Tinkov, A.A.; Saad, K.; Dadar, M.;
Hemimi, M.; Skalny, A.V.; Hosnedlová, B.; Kizek, R.; Osredkar, J.; Urbina,
M.A.; Fabjan, T.; El-Houfey, A.A.; Kałużna-Czaplińska, J.; Gątarek, P.;
Chirumbolo, S. Oxidative stress in autism spectrum disorder. Mol. Neurobiol.,
2020, 57(5), 2314-2332. http://dx.doi.org/10.1007/s12035-019-01742-2 PMID:
32026227
205) Rimland, B.; Callaway, E.; Dreyfus, P. The effect of high doses of vitamin B6 on
autistic children: A double- blind crossover study. Am. J. Psychiatry, 1978,
135(4), 472-475. http://dx.doi.org/10.1176/ajp.135.4.472 PMID: 345827
206) El-Ansary, A.; Cannell, J.J.; Bjørklund, G.; Bhat, R.S.; Al Dbass, A.M.; Alfawaz,
H.A.; Chirumbolo, S.; Al-Ayadhi, L. In the search for reliable biomarkers for the
early diagnosis of autism spectrum disorder: The role of vitamin D. Metab. Brain
Dis., 2018, 33(3), 917-931. http://dx.doi.org/10.1007/s11011-018-0199-1 PMID:
29497932
207) Kałużna-Czaplińska, J.; Gątarek, P.; Chirumbolo, S.; Chartrand, M.S.; Bjørklund,
G. How important is tryptophan in human health? Crit. Rev. Food Sci. Nutr.,
2019, 59(1), 72-88. http://dx.doi.org/10.1080/10408398.2017.1357534 PMID:
28799778
208) Kałużna-Czaplińska, J.; Jóźwik-Pruska, J.; Chirumbolo, S.; Bjørklund, G.
Tryptophan status in autism spectrum disorder and the influence of
supplementation on its level. Metab. Brain Dis., 2017, 32(5), 1585-1593.
http://dx.doi.org/10.1007/s11011-017-0045-x PMID: 28608247
209) Bjørklund, G.; Saad, K.; Chirumbolo, S.; Kern, J.K.; Geier, D.A.; Geier, M.R.;
Urbina, M.A. Immune dysfunction and neuroinflammation in autism spectrum
disorder. Acta Neurobiol. Exp., 2016, 76(4), 257-268.
http://dx.doi.org/10.21307/ane-2017-025 PMID: 28094817
210) Connery, K.; Tippett, M.; Delhey, L.M.; Rose, S.; Slattery, J.C.; Kahler, S.G.;
Hahn, J.; Kruger, U.; Cunningham, M.W.; Shimasaki, C.; Frye, R.E. Intravenous
immunoglobulin for the treatment of autoimmune encephalopathy in children
with autism. Transl. Psychiatry, 2018, 8(1), 148.
http://dx.doi.org/10.1038/s41398-018-0214-7 PMID: 30097568