Volume: 0 | Issue: 000 | View as PDF
  • OPEN ACCESS

Gut Microbiome in Autistic Spectrum Disorders

  • Viktoria Pencheva* ,
  • Ivan Pavlov,
  • Diliyana Dobrikova,
  • Ekaterina Aleksandrova and
  • Aleksandar Kolevski
 Author information
Nature Cell and Science   2024;2(3):141-150

doi: 10.61474/ncs.2024.00004

Abstract

The microorganisms that inhabit different parts of the human body are referred to as the microbiota, while the microbiome refers to the genes they carry. The human microbiome is the focus of numerous studies, most of which highlight the importance of the trillions of bacteria inhabiting various parts of the body in maintaining the health of the macroorganism. Changes in the microbiome have been observed in various conditions, with particular interest in those associated with neurological diseases such as Parkinson’s disease, Alzheimer’s disease, and autism. This review examines the role of the gut microbiome in autism, specifically the currently established differences between individuals with autism and neurotypical individuals, the possible mechanisms by which microorganisms may influence neurological function, and potential therapeutic approaches.

Keywords

Autism, Autistic spectrum disorders, Gut-Brain axis, Gut microbiome, Microbiome, Probiotics

Introduction

Autism spectrum disorders (ASD) encompass a wide range of neurodevelopmental conditions characterized by heterogeneous cognitive, behavioral, and communication impairments that typically manifest in early childhood.1 Common features of autism include impaired communication skills, social withdrawal, and a repetitive or restrictive pattern of behavior, interests, and activities. These diagnostic traits usually appear early in the development of the patient. Additional behaviors often reported include picky eating habits, increased aggression, and anxiety.2 Regressive autism or late-onset autism refers to a subgroup of patients with initially normal development, who gradually lose acquired communication or social interaction skills.3 The diverse phenotypic manifestations of ASD suggest that multiple factors contribute to its etiology. While both genetic and environmental influences are implicated, most cases of ASD are isolated, with the precise cause remaining unknown. To date, over 100 genes have been identified as putatively associated with ASD.4 However, many genetic variants are linked with heterogeneous phenotypes, making it difficult to identify the molecular mechanisms responsible for specific impairments.5 A notable characteristic of individuals with ASD is abnormal eating habits, which may lead to vitamin, mineral, and fatty acid deficiencies.6 Additionally, gastrointestinal (GI) disorders are more commonly observed in autistic children,7 with symptoms such as constipation, diarrhea, or bloating reported at higher rates compared to neurotypical children.8 These findings suggest a relationship between the GI tract and the brain in individuals with ASD. Gut microbiota is thought to play a key role in this relationship by influencing the central nervous system through various mechanisms.9 Further evidence supporting the importance of gut microbiota in ASD is the observed reduction in behavioral and GI symptoms when autistic children are treated with probiotics and/or antibiotics.10 However, the relationships among the altered eating habits of these children, more frequent GI disorders, changes in the gut microbiome, and their correlation with the severity of autistic manifestations are not yet fully understood.

This review aims to highlight the main findings regarding changes in the gut microbiome in ASD, the factors that influence it, and its potential role in the pathogenesis of ASD. A better understanding of these mechanisms and how to modulate them may lead to new approaches for preventing and treating these conditions.

Factors influencing the microbial composition in children with autism

According to a meta-analysis, children born by cesarean section have a 23% higher risk of developing ASD compared to those born via vaginal delivery.11 Another multinational study, which included 5 million births from Norway, Sweden, Denmark, Finland, and Western Australia, similarly reported a higher risk of ASD in children born by cesarean section. However, this study did not establish a relationship between the gestational age at the time of delivery and the risk of developing ASD.12 Premature birth, mode of delivery, and breastfeeding influence the microbial composition of the newborn’s gut (Fig. 1). Babies born vaginally and breastfed tend to have healthier microbiota, with higher levels of beneficial bacteria such as Bifidobacterium, and lower levels of pathogenic bacteria, including Clostridium difficile and Escherichia coli.13 Vaginally delivered infants have a microbiota resembling their mother’s vaginal microbiota, dominated by Lactobacillus, Prevotella and Snethia spp. In contrast, infants born by cesarean section exhibit a microbiota that resembles the mother’s skin, with the dominance of Staphylococcus, Corynebacterium, Propionibacterium spp., Escherichia coli, and Clostridium difficile.14

The impact of the mode of delivery on the gut microbiome in infants
Fig. 1  The impact of the mode of delivery on the gut microbiome in infants

Regardless of the mode of delivery, evidence suggests that changes in the vaginal microbiome during pregnancy may contribute to the development of ASD in offspring.15 One proposed mechanism for this interaction is maternal immune activation, which is thought to affect fetal neurodevelopment. Maternal immune activation refers to the activation of the maternal immune system during pregnancy, leading to the production of cytokines and other proinflammatory molecules that can cross the placenta and influence fetal brain development. Dysbiosis, or an imbalance in the microbiome, may trigger both local and systemic inflammation or predispose the mother to infections, further stimulating the immune system.16 In vaginally delivered children, the composition of the maternal vaginal microbiome is crucial, as it provides the newborn with its first colonizers. Vaginal dysbiosis, such as a reduction in Lactobacillus species, may negatively affect early neurodevelopment. Factors such as maternal stress or antibiotic use during pregnancy can disrupt the maternal vaginal microbiome, leading to dysbiosis and potentially affecting fetal brain development.17

Another factor contributing to microbial dysbiosis is the early and excessive use of antibiotics. This can affect the gut-brain axis by causing epigenetic modifications that impact genes associated with ASD, potentially contributing to the development of the disorder.18 Madore et al. also observed a higher risk of ASD in children who were born prematurely, with very low birth weight, by cesarean section, or who experienced prolonged hospitalization. Additionally, children treated with antibiotics for extended periods or whose mothers had infections during pregnancy were found to have an increased risk of developing ASD.19

GI disorders in children with autism

In addition to dysbiosis, GI symptoms are four times more prevalent in children with ASD compared to the general population.20 These children experience a wide range of GI symptoms, including constipation, diarrhea, bloating, abdominal pain, reflux, vomiting, meteorism and foul-smelling stools. Food allergies are also more common in this population. Children with autism are less likely to consume foods high in glutamic acid, serine, choline, phenylalanine, leucine, tyrosine, valine, and histidine, all of which play a role in neurotransmitter biosynthesis.21

A study comparing 230 preschool children revealed that those with autism suffered significantly more from GI problems than healthy controls did.20 ASD patients with GI symptoms were reported to experience more anxiety and other somatic complaints than those without GI symptoms. In addition, GI disorders in children with autism are associated with an increase in tantrums, aggressive behavior, and sleep disturbances, which further exacerbate behavioral symptoms compared to autistic individuals without GI symptoms.22 A review by Ding et al. suggested that behaviors such as aggression, self-injury, or sleep disturbances in children with autism may be manifestations of abdominal discomfort.23

According to Naviaux, children with autism exhibit altered metabolism and decreased absorption of disaccharides in the gut epithelium.24 The sodium-dependent glucose co-transporter (SGLT1) and glucose transporter 2 (GLUT2) actively transport glucose, galactose, and fructose across the lumen and basolateral membranes of enterocytes. Children with autism have been reported to have significantly reduced mRNA levels of both hexose transporters (SGLT1 and GLUT2) in the ileum. As a result of malabsorption in the small intestine, increased amounts of mono- and disaccharides enter the large intestine, promoting the growth of bacteria that ferment these sugars, thereby altering the microbial composition in the GI tract. The presence of excess sugars in the colon can lead to osmotic diarrhea or serve as substrates for gas production, further contributing to GI distress.25

Changes in the microbiome in children with autism

Significantly increased bacterial species in children with autism include Akkermansia muciniphila, Anaerofilum, Barnesiella intestinihominis, Clostridium spp., Dorea spp., Enterobacteriaceae, Roseburia spp., Parasutterella excrementihominis, and Turicibacter spp. At the same time, the abundances of Bifidobacterium, Fusobacterium, Oscillospira, Sporobacter, Streptococcus, and Subdoligranulum are significantly reduced. The genera Collinsella spp. are also underrepresented, except for Collinsella aerofaciens, as well as Enterococcus spp., Lactobacillus, Lactococcus, and Staphylococcus.3 Kang et al.26 compared the gut flora of twenty autistic children with GI problems to that of twenty neurotypical children. They found significantly lower bacterial diversity in children with autism, which correlated with the severity of their GI symptoms.

A common observation is that Bifidobacterium spp. are less prevalent in the intestines of children with autism, whereas Clostridia spp. are more abundant. A simulation by Weston et al. revealed a two-way relationship between the anti-inflammatory genera Bifidobacterium and the pro-inflammatory Clostridia and Desulfovibrio. Bifidobacterium is inhibited by lysozyme and the growth of Desulfovibrio.27 To some extent, Desulfovibrio thrives on metabolites produced by Bifidobacterium. On the other hand, Clostridia growth is suppressed by both lysozyme and the presence of Bifidobacterium. The authors suggest that an increase in Clostridia in the gut, especially when Bifidobacterium levels are low, may be linked to a higher risk of developing ASD.

Other microorganisms reported to be relevant in children with autism include Candida spp. Several studies have shown a higher amount of Candida spp., particularly Candida albicans, in autistic children.28,29 Altered microbial diversity in autistic populations may favor fungal growth. For example, Lactobacillus spp. stimulate the immune system to produce IL-22. IL-17 and IL-22 together inhibit the overgrowth of Candida spp. Once Candida spp. become established in the gut, they prevent recolonization by commensal microbes. In a dysbiotic environment—often observed in the autistic population—Candida proliferates and produces ammonia and toxins, which are reported to exacerbate autistic behavior. Candida spp. may also cause malabsorption of minerals and carbohydrates, potentially playing a role in the pathophysiology of ASD.

The possible impact of some changes in microbiome composition is represented in Table 1.3,13,30–37

Table 1

Gut microbiome changes in children with autistic spectrum disorders (ASD) and the possible impact.

ChangeGenus/SpeciesPossible impactReferences
IncreasedClostridium sppExcess propionic acid production; Toxin production; Altered tryptophan metabolism leading to reduced serotonin levels; Dysregulation of dopamine signaling; Disruption in the secretion of Gamma-Aminobutyric Acid (GABA) and glutamate; Elevated histamine level; Disruption of blood-brain barrierDe Angelis (2013)3; Abdelli (2019)30
EnterobacteriaceaeIncreased lipopolysaccharides (LPS) production leading to inflammation; Decreased serotonin productionXu (2019)31
Desulfovibrio sppIncreased hydrogen sulfide production which disrupts gut barrier function, alters gut motility and may lead to gut inflammationFinegold (2011)32
Candida sppStimulation of immune system leading to inflammation; Production of toxic metabolites such as ethanol and acetaldehyde which may increase neuroinflammationHerman (2022)33
DecreasedBifidobacterium sppLower levels of neurotransmitters GABA and serotonin; Impaired carbohydrate fermentation; Reduced Treg activity; Deficiency of B-vitaminsNiu (2019)34; Ha (2021)35; Srikantha (2019)13
Bacteroides sppLower levels of short chain fatty acids (SCFA); Altered immune function; Reduced synthesis of neurotransmitters – serotonin; Impaired carbohydrate metabolismNiu (2019)34; Ha (2021)35; Xu (2019)31
Lactobacillus sppImmune system dysregulation; Decreased Gamma-Aminobutyric Acid (GABA); Reduced serotonin levels; Reduced SCFATaniya (2022)36
Akkermansia muciniphilaDisruption of mucus layer production which can lead to increased gut permeability and gut inflammationWang (2011)37

Possible mechanisms and the gut-brain axis

The gut-brain axis is defined as the complex bidirectional communication system between the central nervous system and the enteric nervous system.38 Research is being conducted on the gut-brain axis (GBA) and its relationship with various neurological disorders, including autism. Over the past decade, studies on GBA-modulating factors have revealed the central role of the gut microbiome—the trillions of microbes that colonize the gut—in regulating neuroimmune networks, modifying neural networks, and communicating directly with the brain.39 Alterations in the gut microbiome and the subsequent dysregulation of the GBA are thought to contribute to the pathogenesis of neurodevelopmental disorders, including autism. However, the precise mechanisms and the extent to which the microbiome influences these dynamics are still unclear (Fig. 2).

Gut-brain axis and microbiota interactions.
Fig. 2  Gut-brain axis and microbiota interactions.

HPA, Hypothalamic-Pituitary-Adrenal; SCFA, short chain fatty acids; GABA, Gamma-Aminobutyric Acid.

Disturbances in the bidirectional communication between the gut and the brain can lead to various conditions, such as inflammatory bowel syndrome, and various chronic neurological conditions, including Parkinson’s disease, ASD, chronic pain and mood changes.40 The gut microbiome can affect the central nervous system (CNS) through multiple mechanisms, primarily by modulating the immune system through various cytokines and the release of metabolites, including neurotransmitters, by gut bacteria.13 Children with autism more frequently exhibit increased intestinal permeability. This may lead to greater interactions between the mucosal immune system and the lipopolysaccharides (LPS) of gut bacteria, inducing the production of cytokines such as IL-1β, IL-6, IL-8, and TNF-α. Elevated levels of immune modulators in plasma have been associated with neurodevelopmental disorders in children with autism.41,42 A study by Zurita et al. demonstrated a correlation between differences in the gut microbiome of children with autism and neurotypical children, with significantly elevated serum levels of TGF-β (thrombocyte-growth factor-β). No significant changes in the serum levels of IL-6 were noted.43 The bacterial genera most strongly associated with differences in cytokine profiles were Bacteroides, Prevotella, and Bifidobacterium.44 The central role of the immune system in mediating communication between the gut microbiome and the brain, as well as other peripheral systems, was further highlighted by Jacobson et al.39

During their metabolic processes, bacteria can release various substances that affect the CNS. For example, short chain fatty acids(SCFAs) such as acetic acid, propionic acid, and butyric acid are released. In children with autism, increased levels of acetate and propionate and decreased levels of butyrate are commonly observed. These changes may be related to neurodevelopment by directly affecting CNS physiology through increased epigenetic changes and/or mitochondrial dysfunctions.45,46 Elevated levels of another metabolic product, p-cresol, primarily released by Clostridium difficile and Bifidobacterium spp., are also associated with behavioral deterioration in children with autism.47 By inhibiting dopamine-β-hydroxylase, p-cresol can influence dopamine metabolism in the brain.48 A simulation study analyzing enzyme production involved in glutamate metabolism across different microbiomes revealed that these enzymes were less represented in the autistic microbiome compared to healthy microbiome. Glutamate is an important metabolite for neurological development. On one hand, it is a component of the peptide glutathione, which acts as an antioxidant to reduce oxidative stress in cells. On the other hand, glutamate is an excitatory neurotransmitter, and an imbalance in its activation and repression within the CNS may contribute to the development of ASD.49 The microbial population in the colon may also be related to other molecules that serve as neurotransmitters in the CNS. For instance, Lactobacillus spp. and Bifidobacterium spp. can produce Gamma-Aminobutyric Acid (GABA), a major inhibitory neurotransmitter which is found in increased concentrations in children with autism.50 Particularly important in the gut-brain axis is the neurotransmitter serotonin. Some species, such as Escherichia, Enterococcus, and Candida, can directly synthesize serotonin, while others, such as Clostridium spp., and Lactobacillus spp., may be involved in regulating its secretion.50,51 In children with autism, hyperserotoninemia is observed along with reduced levels of serotonin in the brain.52 Elevated 3-(3-hydroxyphenyl)-3-hydroxypropionic acid, 3-hydroxyphenylacetic acid, and 3-hydroxyhippuric acid in children with autism suggest disruptions in phenylalanine metabolism. These metabolites are associated with an abundance of Clostridia spp., which may exacerbate autistic behavior.53

One of the main pathways for gut-brain interactions is the vagus nerve (n. vagus). It is composed of approximately 20% efferent fibers, which regulate gastrointestinal, lung, and heart functions, and approximately 80% afferent fibers, which are responsible for transmitting visceral and somatic sensations.54 The vagus nerve has anti-inflammatory functions, interacting with the hypothalamic-pituitary-adrenal axis, the splenic sympathetic anti-inflammatory pathway, and the cholinergic anti-inflammatory pathway. These interactions lead to reduced inflammation in the gut and brain, enhanced immune regulation, stress regulation, neurodevelopment, and the maintenance of cognitive functions.55 Impaired anti-inflammatory functions of the vagus nerve may contribute to low-grade inflammation in the gut and brain, as reported in children with ASD.5 In turn, chronic gut inflammation may lead to vagus dysfunctions, such as disruption of the cholinergic anti-inflammatory pathway, altered vagal signaling, vagal hypersensitivity, reduced vagal tone, or neuroinflammation.56 Gut microbiota metabolites, such as SCFAs, play a key role in maintaining gut barrier function and possess immune-modulatory activity, which reduces inflammation. SCFAs, particularly butyrate, can enhance vagal tone, thereby reducing symptoms such as anxiety and depression.57 Gut microbiota can also directly affect brain functions, influencing mood and behavior via the vagus nerve, with microbiota metabolites such as SCFAs and neurotransmitters such as GABA, serotonin, and dopamine acting as mediators.58 Although the direct mechanisms are not yet fully understood, increasing evidence suggests a link between dysbiosis, autistic manifestations, and the vagus nerve as a critical component of these interactions.

Although the mechanisms underlying the gut microbiome-brain connection and its relevance to autism development are not yet fully understood, these studies highlight key pathways that warrant further exploration in future research.

Modulation of the microbiome by diet, probiotics and antibiotics in children with autism

Various factors can influence the formation of the gut microbiome, including diet, vitamin and mineral intake, and the use of prebiotics, probiotics and antibiotics. Following the discovery of the gut-brain connection, numerous studies have explored how modulating the microbiome could affect neurological functions, including in children with autism.

Prebiotics are described as “non-digestible food ingredients that beneficially affect the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the colon, thereby improving host health”.59 In a study by Grimaldi and colleagues, galactooligosaccharides were shown to affect the production of SCFAs, lowering propionate and increasing butyrate, which may benefit children with autism. In addition, this prebiotic can affect brain dysfunction caused by the overproduction of GABA.49

Probiotics are defined as “live microorganisms which, when administered in adequate amounts, confer a health benefit on the host”.60 Various strains and combinations of strains are being investigated for their potential beneficial effects on both GI symptoms and behavior in children with autism. In a study by Shaaban and colleagues, 30 autistic children who consumed probiotics containing Lactobacillus acidophilus, Lactobacillus rhamnosus and Bifidobacteria longum were followed for 3 months. The study revealed an improvement in GI symptoms and a reduction in the severity of autistic symptoms.61 Billeci et al. demonstrated an improvement in the brain function of children with autism following probiotic intake, as measured by beta- and gamma-wave activity through EEG.62 A reduction in frontopolar region power was found in the beta and gamma bands, which are often elevated in autistic children.63 In a case report examining a 12-year-old patient with autism, fewer GI complaints, and an improvement in autistic manifestations were also observed.64

The number of participants in clinical trials on this subject is limited, but various studies are being conducted in animal models to establish the exact mechanisms by which probiotics affect neurodevelopment. The difficulties in studying ASD patients have led to the development of animal models that mimic the clinical characteristics of these patients. Depending on the construction methods, genetic models (Table 2),47 environment-induced models and idiopathic models (Table 3) are distinguished.47,65 These models are based on the understanding that, in addition to genetic factors, environmental factors may also influence ASD development.66 Environmental factors include pesticides, drug consumption, heat, diet, and pollutants.67,68

Table 2

Animal models of Аutistic Spectrum Disorders (ASD) – genetic models47

ModelFunction/DamageAnimals
GENETIC MODELSSyndromic ASD genes
MECP2Starts or inhibits transcription; neuron maturation; regulated by developmentMouse, Rodent, Fish, Invertebrates
FMR1Involves in translation; affects neuronal proliferation and migrationRodent, Fish, Invertebrates
SHANK3Promotes formation, maturation, and stability of dendritic spinesMouse, Rodent, Fish, Invertebrates
TSC1/2Regulates mTORC1 pathway, neuronal differentiation, and Purkinje cell excitabilityRodent
UBE3ARegulates neuronal homeostatic synaptic plasticityRodents
Non-syndromic ASD genes
NLGNsRegulates the formation of hippocampal neurons and post-glutamatergic synapse proteinsRodents
NRXNsEncodes neuronal transmembrane protein; interacts glial cellsRodents
CHD8Controls epigenetic and transcript regulation; affects brain phenotypeRodents, Fish
POGZRegulates neuronal developmentRodents
ANK2Affects axonal branching; regulates postnatal development of excitatory synapsesRodents
MIR137Regulates neuronal gene expression and neurogenesisRodents
Table 3

Animal models of Аutistic Spectrum Disorders (ASD)–Environmental-induced and idiopathic models47

Environmental factors
ENVIROMENTAL-INDUCED MODELS DRUG-INDUCED MODELSVPA (Valproic acid)Affects expression of BDFN mRNA in brain tissueRodents, Fish
PPA (Propionic acid)Reactive astrocyte keratinization of brain tissue; microglia are activated; oxidative stress markers rise; glutathione declinesRodents
BPA (Bisphenol propan)Changes in number of neurons and glia in the medial prefrontal lobeRodents
SevofluraneIncreases the number of apoptotic cells in brain; inhibits the axon development of hippocampal neuronsRodents
ENVIRONMENTAL-INDUCED MODELSMaternal immune activation models (MIA)Abnormal increase of offspring’s brain volumeMonkey, Rodents
Borna disease virus (BDV) modelsAbnormal hippocampal and cerebellar developmentRodents
Gut microbiota modelsRegulation of neuroactive metabolitesMonkey, Rodents, Fish, Invertebrates
Repeated cold temperature stress (RCS) modelsChanges in neurotransmitter and corticosterone levelsRodents
Inbred line
IDIOPATHIC MODELSBTBR T+Itpr3tf/J mouse modelPolymorphisms in the Kmo gene, which encodes urine 3-monooxygenase
Inbred line BALB/cByJ mouse modelReduced corpus callosum volume

In a study by Abuaish et al. using rodent models, alterations in the ratios of Clostridium spp. led to a decrease in Clostridium perfringens and an increase in Clostridium cluster IV, resulting in improved social behavior in these models when treated with Bifidobacterium longum BB536.69 In another study on mouse models of ASD supplemented with Lactobacillus plantarum ST-III, an increase in beneficial Lachnospiraceae spp. and a decrease in Alistipes spp. were observed, which also led to improvements in social behavior.70 In a study by Tabouy et al., mouse models of ASD treated with Lactobacillus reuteri showed increased GABA-receptor gene expression and protein levels of GABRA1 in the hippocampus.71 Furthermore, Lactobacillus reuteri therapy has been found to increase oxytocin levels in the brain, potentially improving brain function and behavior by stimulating the vagus nerve.72 Ingesting Lactobacillus rhamnosus in BTBR mouse models of ASD led to increased levels of beneficial neuroactive compounds such as 5-aminovaleric acid and choline.73 Despite these promising results in animal models, further clinical studies are needed to identify the exact bacterial strains or combinations that can serve as adjunctive therapies for autism in children.

Another approach to modulating the gut microbiome is through antibiotics. Oral vancomycin is often discussed due to its poor absorption in the GI tract, resulting in minimal systemic effects, and its action specifically targeting gram-positive bacteria, including Clostridium spp.74 In a study by Sandler et al.,11 children with ASD who received oral vancomycin therapy were followed up. The results revealed an improvement in GI symptoms and behavior in eight of them.75 However, the effects of this therapy were short-lived, likely due to the survival and subsequent proliferation of Clostridium spp. spores.3 The findings of Sandler et al. are being further explored in animal models, where vancomycin has also been shown to impact autistic behaviors. The effects of vancomycin can be explained by its influence on the gut microbiome, which in turn affects the production of SCFAs and various bacterial metabolites, impacting the mitochondrial dysfunction observed in autism.76 On the other hand, the changes induced by vancomycin may also lead to alterations in the relationship between the intestinal microbiome and the immune system.77

Another therapeutic approach of interest in children with ASD is fecal microbiota transplantation (FMT). FMT involves the transplantation of fecal matter from healthy donors to recipients to restore the healthy balance of the gut microbiota. It is often used to treat conditions related to gut dysbiosis, such as Clostridium difficile infections. While more studies have been conducted in animal models, a few have included FMT in children with ASD. In a systematic review by Zhang et al., five studies were reviewed: two prospective open-label studies, two retrospective observational studies, and a case report. All of them reported improvements in behavioral symptoms and reductions in gastrointestinal symptoms.78 In a clinical trial by Kang et al., 18 children diagnosed with ASD received fecal matter transplantation two weeks after antibiotic treatment and intestinal clearance. Eight weeks after transplantation, improvements in both gastrointestinal and behavioral symptoms were observed.79 Similar findings were reported in another study involving 8 children with ASD, two years after FMT.80 The encouraging results from these studies warrant additional, larger double-blind clinical trials to establish precise guidelines for this treatment.

Limitations

This review highlights the influence of the gut microbiome on the gut-brain axis and its role in ASD, while also presenting some potential therapeutic approaches. However, several limitations must be acknowledged. Тhe potential therapeutic approaches have been studied in different studies with small sample sizes and short-term observations, which may not provide a comprehensive understanding of long-term effects. Larger double-blind placebo-controlled trials are necessary to confirm the effects of these treatments and establish exact guidelines. Additionally, studies on gut microbiome abnormalities have been conducted on a relatively small number of participants, which may affect the reliability of their results. When tracking changes in the microbiome, it is important to consider the specific characteristics of the microbiome in different populations, which are influenced by geographical location, environmental factors, local diet, and traditions. To more accurately identify differences in the microbial profile of children with autism, studies involving larger participant numbers and focused on specific populations are needed. Notably, the mechanisms of interaction between the gut microbiome and the gut-brain axis in ASD are still not fully understood. Our review reflects the main findings to date; however, further research is necessary to clarify these relationships fully and comprehensively.

Conclusions

The role of the gut microbiome in health and disease is garnering increasing interest, particularly in its impact on neurological disorders such as ASD. ASD is socially significant because of its chronic nature and the limited number of effective therapeutic approaches available. Research suggests a connection between the gastrointestinal tract and neurological disorders, with evidence showing more frequent gastrointestinal symptoms in children with autism. Advances in microbiome research have revealed differences in microbial profiles between children with autism and neurotypical children. Factors such as birth method, diet, and the use of antibiotics by either the child or the mother during childbirth or pregnancy can influence the composition of the microbiome. Managing these factors may offer preventative benefits and help reduce the incidence or severity of such conditions in children. The metabolic byproducts of gut bacteria can affect the central nervous system directly or by influencing other metabolic processes, leading to changes at various molecular levels. Understanding the precise mechanisms by which the microbiome influences the gut-brain axis through its metabolites is crucial. Positive outcomes from modulating the gut microbiome via the use of probiotics, antibiotics, or fecal microbiota transplantation present promising new therapeutic options for addressing neurological and gastrointestinal disorders. However, this area of research is still developing, and more studies with larger sample sizes are needed to establish precise guidelines.

Declarations

Acknowledgement

The authors have no individuals or organizations to acknowledge in relation to this article.

Funding

The authors received no specific funding for this work.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as potential conflicts of interest.

Authors’ contributions

Review’s concept and design (VP, AK), acquisition of data (VP, IP, DD, EA), analysis and interpretation of data (VP, IP, DD, EA), drafting the manuscript (VP, IP, DD, EA), critical revision of the manuscript (AK), technical support (AK), and study supervision (AK) were performed. All the authors have approved the final manuscript.

References

  1. Lord C, Brugha TS, Charman T, Cusack J, Dumas G, Frazier T, et al. Autism spectrum disorder. Nat Rev Dis Primers 2020;6(1):5 View Article PubMed/NCBI
  2. Manchia M, Fanos V. Targeting aggression in severe mental illness: The predictive role of genetic, epigenetic, and metabolomic markers. Prog Neuropsychopharmacol Biol Psychiatry 2017;77:32-41 View Article PubMed/NCBI
  3. De Angelis M, Piccolo M, Vannini L, Siragusa S, De Giacomo A, Serrazzanetti DI, et al. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS One 2013;8(10):e76993 View Article PubMed/NCBI
  4. Satterstrom FK, Kosmicki JA, Wang J, Breen MS, De Rubeis S, An JY, et al. Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism. Cell 2020;180(3):568-584.e23 View Article PubMed/NCBI
  5. Iakoucheva LM, Muotri AR, Sebat J. Getting to the Cores of Autism. Cell 2019;178(6):1287-1298 View Article PubMed/NCBI
  6. Berding K, Donovan SM. Microbiome and nutrition in autism spectrum disorder: current knowledge and research needs. Nutr Rev 2016;74(12):723-736 View Article PubMed/NCBI
  7. McElhanon BO, McCracken C, Karpen S, Sharp WG. Gastrointestinal symptoms in autism spectrum disorder: a meta-analysis. Pediatrics 2014;133(5):872-883 View Article PubMed/NCBI
  8. Lefter R, Ciobica A, Timofte D, Stanciu C, Trifan A. A Descriptive Review on the Prevalence of Gastrointestinal Disturbances and Their Multiple Associations in Autism Spectrum Disorder. Medicina (Kaunas) 2019;56(1):11 View Article PubMed/NCBI
  9. Morton JT, Jin DM, Mills RH, Shao Y, Rahman G, McDonald D, et al. Multi-level analysis of the gut-brain axis shows autism spectrum disorder-associated molecular and microbial profiles. Nat Neurosci 2023;26(7):1208-1217 View Article PubMed/NCBI
  10. Lewandowska-Pietruszka Z, Figlerowicz M, Mazur-Melewska K. Microbiota in Autism Spectrum Disorder: A Systematic Review. Int J Mol Sci 2023;24(23):16660 View Article PubMed/NCBI
  11. Curran EA, O’Neill SM, Cryan JF, Kenny LC, Dinan TG, Khashan AS, et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry 2015;56(5):500-508 View Article PubMed/NCBI
  12. Yip BHK, Leonard H, Stock S, Stoltenberg C, Francis RW, Gissler M, et al. Caesarean section and risk of autism across gestational age: a multi-national cohort study of 5 million births. Int J Epidemiol 2017;46(2):429-439 View Article PubMed/NCBI
  13. Srikantha P, Mohajeri MH. The Possible Role of the Microbiota-Gut-Brain-Axis in Autism Spectrum Disorder. Int J Mol Sci 2019;20(9):2115 View Article PubMed/NCBI
  14. Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther 2016;158:52-62 View Article PubMed/NCBI
  15. Hashimoto K. Emerging role of the host microbiome in neuropsychiatric disorders: overview and future directions. Mol Psychiatry 2023;28(9):3625-3637 View Article PubMed/NCBI
  16. Di Simone N, Santamaria Ortiz A, Specchia M, Tersigni C, Villa P, Gasbarrini A, et al. Recent Insights on the Maternal Microbiota: Impact on Pregnancy Outcomes. Front Immunol 2020;11:528202 View Article PubMed/NCBI
  17. Johnson D, Letchumanan V, Thurairajasingam S, Lee LH. A Revolutionizing Approach to Autism Spectrum Disorder Using the Microbiome. Nutrients 2020;12(7):1983 View Article PubMed/NCBI
  18. Eshraghi RS, Deth RC, Mittal R, Aranke M, Kay SS, Moshiree B, et al. Early Disruption of the Microbiome Leading to Decreased Antioxidant Capacity and Epigenetic Changes: Implications for the Rise in Autism. Front Cell Neurosci 2018;12:256 View Article PubMed/NCBI
  19. Madore C, Leyrolle Q, Lacabanne C, Benmamar-Badel A, Joffre C, Nadjar A, et al. Neuroinflammation in Autism: Plausible Role of Maternal Inflammation, Dietary Omega 3, and Microbiota. Neural Plast 2016;2016:3597209 View Article PubMed/NCBI
  20. Fulceri F, Morelli M, Santocchi E, Cena H, Del Bianco T, Narzisi A, et al. Gastrointestinal symptoms and behavioral problems in preschoolers with Autism Spectrum Disorder. Dig Liver Dis 2016;48(3):248-254 View Article PubMed/NCBI
  21. Dalangin R, Kim A, Campbell RE. The Role of Amino Acids in Neurotransmission and Fluorescent Tools for Their Detection. Int J Mol Sci 2020;21(17):6197 View Article PubMed/NCBI
  22. Iovene MR, Bombace F, Maresca R, Sapone A, Iardino P, Picardi A, et al. Intestinal Dysbiosis and Yeast Isolation in Stool of Subjects with Autism Spectrum Disorders. Mycopathologia 2017;182(3-4):349-363 View Article PubMed/NCBI
  23. Ding HT, Taur Y, Walkup JT. Gut Microbiota and Autism: Key Concepts and Findings. J Autism Dev Disord 2017;47(2):480-489 View Article PubMed/NCBI
  24. Naviaux RK. Metabolic features of the cell danger response. Mitochondrion 2014;16:7-17 View Article PubMed/NCBI
  25. Williams BL, Hornig M, Buie T, Bauman ML, Cho Paik M, Wick I, et al. Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances. PLoS One 2011;6(9):e24585 View Article PubMed/NCBI
  26. Kang DW, Park JG, Ilhan ZE, Wallstrom G, Labaer J, Adams JB, et al. Reduced incidence of Prevotella and other fermenters in intestinal microflora of autistic children. PLoS One 2013;8(7):e68322 View Article PubMed/NCBI
  27. Weston B, Fogal B, Cook D, Dhurjati P. An agent-based modeling framework for evaluating hypotheses on risks for developing autism: effects of the gut microbial environment. Med Hypotheses 2015;84(4):395-401 View Article PubMed/NCBI
  28. Strati F, Cavalieri D, Albanese D, De Felice C, Donati C, Hayek J, et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome 2017;5(1):24 View Article PubMed/NCBI
  29. Kantarcioglu AS, Kiraz N, Aydin A. Microbiota-Gut-Brain Axis: Yeast Species Isolated from Stool Samples of Children with Suspected or Diagnosed Autism Spectrum Disorders and In Vitro Susceptibility Against Nystatin and Fluconazole. Mycopathologia 2016;181(1-2):1-7 View Article PubMed/NCBI
  30. Abdelli LS, Samsam A, Naser SA. Propionic Acid Induces Gliosis and Neuro-inflammation through Modulation of PTEN/AKT Pathway in Autism Spectrum Disorder. Sci Rep 2019;9(1):8824 View Article PubMed/NCBI
  31. Xu M, Xu X, Li J, Li F. Association Between Gut Microbiota and Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. Front Psychiatry 2019;10:473 View Article PubMed/NCBI
  32. Finegold SM. Desulfovibrio species are potentially important in regressive autism. Med Hypotheses 2011;77(2):270-274 View Article PubMed/NCBI
  33. Herman A, Herman AP. Could Candida Overgrowth Be Involved in the Pathophysiology of Autism?. J Clin Med 2022;11(2):442 View Article PubMed/NCBI
  34. Niu M, Li Q, Zhang J, Wen F, Dang W, Duan G, et al. Characterization of Intestinal Microbiota and Probiotics Treatment in Children With Autism Spectrum Disorders in China. Front Neurol 2019;10:1084 View Article PubMed/NCBI
  35. Ha S, Oh D, Lee S, Park J, Ahn J, Choi S, et al. Altered Gut Microbiota in Korean Children with Autism Spectrum Disorders. Nutrients 2021;13(10):3300 View Article PubMed/NCBI
  36. Taniya MA, Chung HJ, Al Mamun A, Alam S, Aziz MA, Emon NU, et al. Role of Gut Microbiome in Autism Spectrum Disorder and Its Therapeutic Regulation. Front Cell Infect Microbiol 2022;12:915701 View Article PubMed/NCBI
  37. Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA. Low relative abundances of the mucolytic bacterium Akkermansia muciniphila and Bifidobacterium spp. in feces of children with autism. Appl Environ Microbiol 2011;77(18):6718-6721 View Article PubMed/NCBI
  38. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Ann Gastroenterol 2015;28(2):203-209 PubMed/NCBI
  39. Jacobson A, Yang D, Vella M, Chiu IM. The intestinal neuro-immune axis: crosstalk between neurons, immune cells, and microbes. Mucosal Immunol 2021;14(3):555-565 View Article PubMed/NCBI
  40. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest 2015;125(3):926-938 View Article PubMed/NCBI
  41. Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, et al. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007;55(5):453-462 View Article PubMed/NCBI
  42. Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Pessah I, Van de Water J. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun 2011;25(1):40-45 View Article PubMed/NCBI
  43. Zurita MF, Cárdenas PA, Sandoval ME, Peña MC, Fornasini M, Flores N, et al. Analysis of gut microbiome, nutrition and immune status in autism spectrum disorder: a case-control study in Ecuador. Gut Microbes 2020;11(3):453-464 View Article PubMed/NCBI
  44. Hsiao EY, McBride SW, Hsien S, Sharon G, Hyde ER, McCue T, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013;155(7):1451-1463 View Article PubMed/NCBI
  45. 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 View Article PubMed/NCBI
  46. Reigstad CS, Salmonson CE, Rainey JF, Szurszewski JH, Linden DR, Sonnenburg JL, et al. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J 2015;29(4):1395-1403 View Article PubMed/NCBI
  47. Ming X, Stein TP, Barnes V, Rhodes N, Guo L. Metabolic perturbance in autism spectrum disorders: a metabolomics study. J Proteome Res 2012;11(12):5856-5862 View Article PubMed/NCBI
  48. Clayton TA. Metabolic differences underlying two distinct rat urinary phenotypes, a suggested role for gut microbial metabolism of phenylalanine and a possible connection to autism. FEBS Lett 2012;586(7):956-961 View Article PubMed/NCBI
  49. Grimaldi R, Cela D, Swann JR, Vulevic J, Gibson GR, Tzortzis G, et al. In vitro fermentation of B-GOS: impact on faecal bacterial populations and metabolic activity in autistic and non-autistic children. FEMS Microbiol Ecol 2017;93(2):fiw233 View Article PubMed/NCBI
  50. Dinan TG, Stilling RM, Stanton C, Cryan JF. Collective unconscious: how gut microbes shape human behavior. J Psychiatr Res 2015;63:1-9 View Article PubMed/NCBI
  51. Gabriele S, Sacco R, Persico AM. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol 2014;24(6):919-929 View Article PubMed/NCBI
  52. Chugani DC, Muzik O, Behen M, Rothermel R, Janisse JJ, Lee J, et al. Developmental changes in brain serotonin synthesis capacity in autistic and nonautistic children. Ann Neurol 1999;45(3):287-295 View Article PubMed/NCBI
  53. Li Q, Zhou JM. The microbiota-gut-brain axis and its potential therapeutic role in autism spectrum disorder. Neuroscience 2016;324:131-139 View Article PubMed/NCBI
  54. Bonaz B, Sinniger V, Pellissier S. Therapeutic Potential of Vagus Nerve Stimulation for Inflammatory Bowel Diseases. Front Neurosci 2021;15:650971 View Article PubMed/NCBI
  55. Breit S, Kupferberg A, Rogler G, Hasler G. Vagus Nerve as Modulator of the Brain-Gut Axis in Psychiatric and Inflammatory Disorders. Front Psychiatry 2018;9:44 View Article PubMed/NCBI
  56. Mayer EA, Tillisch K, Gupta A. Gut/brain axis and the microbiota. J Clin Invest 2015;125(3):926-938 View Article PubMed/NCBI
  57. Fattorusso A, Di Genova L, Dell’Isola GB, Mencaroni E, Esposito S. Autism Spectrum Disorders and the Gut Microbiota. Nutrients 2019;11(3):521 View Article PubMed/NCBI
  58. Fülling C, Dinan TG, Cryan JF. Gut Microbe to Brain Signaling: What Happens in Vagus…. Neuron 2019;101(6):998-1002 View Article PubMed/NCBI
  59. Gibson GR, Scott KP, Rastall RA, Tuohy KM, Hotchkiss AT, Dubert-Ferrandon A, et al. Dietary prebiotics: Current status and new definition. Food Sci Technology Bulletin Functional Foods 2010;7(1):1-19 View Article
  60. FAO/WHO. Evaluation of health and nutritional properties of powder milk and live lactic acid bacteria. Geneva: WHO; 2001
  61. Shaaban SY, El Gendy YG, Mehanna NS, El-Senousy WM, El-Feki HSA, Saad K, et al. The role of probiotics in children with autism spectrum disorder: A prospective, open-label study. Nutr Neurosci 2018;21(9):676-681 View Article PubMed/NCBI
  62. Billeci L, Callara AL, Guiducci L, Prosperi M, Morales MA, Calderoni S, et al. A randomized controlled trial into the effects of probiotics on electroencephalography in preschoolers with autism. Autism 2023;27(1):117-132 View Article PubMed/NCBI
  63. Nicotera AG, Hagerman RJ, Catania MV, Buono S, Di Nuovo S, Liprino EM, et al. EEG Abnormalities as a Neurophysiological Biomarker of Severity in Autism Spectrum Disorder: A Pilot Cohort Study. J Autism Dev Disord 2019;49(6):2337-2347 View Article PubMed/NCBI
  64. Grossi E, Melli S, Dunca D, Terruzzi V. Unexpected improvement in core autism spectrum disorder symptoms after long-term treatment with probiotics. SAGE Open Med Case Rep 2016;4:2050313X16666231 View Article PubMed/NCBI
  65. Li Z, Zhu YX, Gu LJ, Cheng Y. Understanding autism spectrum disorders with animal models: applications, insights, and perspectives. Zool Res 2021;42(6):800-824 View Article PubMed/NCBI
  66. Almandil NB, Alkuroud DN, AbdulAzeez S, AlSulaiman A, Elaissari A, Borgio JF. Environmental and Genetic Factors in Autism Spectrum Disorders: Special Emphasis on Data from Arabian Studies. Int J Environ Res Public Health 2019;16(4):658 View Article PubMed/NCBI
  67. He X, Tu Y, Song Y, Yang G, You M. The relationship between pesticide exposure during critical neurodevelopment and autism spectrum disorder: A narrative review. Environ Res 2022;203:111902 View Article PubMed/NCBI
  68. Heidari H, Lawrence DA. An integrative exploration of environmental stressors on the microbiome-gut-brain axis and immune mechanisms promoting neurological disorders. J Toxicol Environ Health B Crit Rev 2024;27(7):233-263 View Article PubMed/NCBI
  69. Abuaish S, Al-Otaibi NM, Abujamel TS, Alzahrani SA, Alotaibi SM, AlShawakir YA, et al. Fecal Transplant and Bifidobacterium Treatments Modulate Gut Clostridium Bacteria and Rescue Social Impairment and Hippocampal BDNF Expression in a Rodent Model of Autism. Brain Sci 2021;11(8):1038 View Article PubMed/NCBI
  70. Guo M, Li R, Wang Y, Ma S, Zhang Y, Li S, et al. Lactobacillus plantarum ST-III modulates abnormal behavior and gut microbiota in a mouse model of autism spectrum disorder. Physiol Behav 2022;257:113965 View Article PubMed/NCBI
  71. Tabouy L, Getselter D, Ziv O, Karpuj M, Tabouy T, Lukic I, et al. Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain Behav Immun 2018;73:310-319 View Article PubMed/NCBI
  72. Sgritta M, Dooling SW, Buffington SA, Momin EN, Francis MB, Britton RA, et al. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019;101(2):246-259.e6 View Article PubMed/NCBI
  73. Pochakom A, Mu C, Rho JM, Tompkins TA, Mayengbam S, Shearer J. Selective Probiotic Treatment Positively Modulates the Microbiota-Gut-Brain Axis in the BTBR Mouse Model of Autism. Brain Sci 2022;12(6):781 View Article PubMed/NCBI
  74. Finegold SM. Therapy and epidemiology of autism—clostridial spores as key elements. Med Hypotheses 2008;70(3):508-511 View Article PubMed/NCBI
  75. Sandler RH, Finegold SM, Bolte ER, Buchanan CP, Maxwell AP, Väisänen ML, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol 2000;15(7):429-435 View Article PubMed/NCBI
  76. Frye RE, Slattery J, MacFabe DF, Allen-Vercoe E, Parker W, Rodakis J, et al. Approaches to studying and manipulating the enteric microbiome to improve autism symptoms. Microb Ecol Health Dis 2015;26:26878 View Article PubMed/NCBI
  77. Bilbo SD, Nevison CD, Parker W. A model for the induction of autism in the ecosystem of the human body: the anatomy of a modern pandemic?. Microb Ecol Health Dis 2015;26:26253 View Article PubMed/NCBI
  78. Zhang J, Zhu G, Wan L, Liang Y, Liu X, Yan H, et al. Effect of fecal microbiota transplantation in children with autism spectrum disorder: A systematic review. Front Psychiatry 2023;14:1123658 View Article PubMed/NCBI
  79. Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome 2017;5(1):10 View Article PubMed/NCBI
  80. Kang DW, Adams JB, Coleman DM, Pollard EL, Maldonado J, McDonough-Means S, et al. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Sci Rep 2019;9(1):5821 View Article PubMed/NCBI
  • Nature Cell and Science
  • 2958-695X

Gut Microbiome in Autistic Spectrum Disorders

Viktoria Pencheva, Ivan Pavlov, Diliyana Dobrikova, Ekaterina Aleksandrova, Aleksandar Kolevski
  • Reset Zoom
  • Download TIFF