The interplay between gut microbiota and autism spectrum disorders: A focus on immunological pathways

https://doi.org/10.1016/j.pnpbp.2020.110091Get rights and content

Highlights

  • Children with ASD represent high rates of gastrointestinal complications.

  • Gut microbiota affects the immune system via various signaling pathways.

  • Gut-brain axis is defined as a bidirectional interaction between gut microbiota and brain.

  • Inappropriate stimulation of the immune system disrupts brain function through different mechanisms.

  • Modifying the microbiota alterations via psychobiotics could reverse the ASD-related behaviors.

Abstract

Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterized by impairments in social and cognitive activities, stereotypical and repetitive behaviors and restricted areas of interest. A remarkable proportion of ASD patients represent immune dysregulation as well as gastrointestinal complications. Hence, a novel concept has recently emerged, addressing the possible intercommunication between the brain, the immune system, the gut and its commensals. Here, we provide an overview of how gut microbes and their metabolites are associated with neurobehavioral features of ASD through various immunologic mechanisms. Moreover, we discuss the potential therapeutic options that could modify these features.

Introduction

It is now well established that there is bidirectional communication between gut microbiota and brain, known as the microbiome-gut-brain axis. Dysregulation in any part of this axis might contribute to the pathogenesis of not only gut-related disorders such as inflammatory bowel disease (IBD), inflammatory bowel syndrome (IBS) and celiac disease but also neuropsychological conditions such as depression, Alzheimer’s disease, schizophrenia and autism spectrum disorders (ASD) (Hsiao et al., 2013; Nemani et al., 2015; Petra et al., 2015; Shen et al., 2020). In support, it has been shown that ASD is accompanied by gastrointestinal (GI) problems such as abdominal pain, diarrhea and constipation (Li et al., 2019). The precise mechanisms linking the gut and the brain are not fully understood yet. However, there is increasing evidence regarding the fundamental role of the immune system in mediating this interaction.

It has been demonstrated that even slight activation of the immune system early in life exerts a significant impact on the central nervous system (CNS) development. For instance, sustained action and proliferation of glial cells, i.e., microglia and astrocytes (Kierdorf and Prinz, 2013) due to various exogenous and endogenous stimuli could lead to pathological changes in the CNS since the activated forms of these cells release pro-inflammatory factors and cause neuroinflammation. In this regard, post-mortem brains of ASD patients represent higher levels of microglia, astrocytes, as well as pro-inflammatory biomarkers, suggesting the possible role of the immune system in ASD pathogenesis (Vuong and Hsiao, 2017).

Recent evidence proposes the necessity of normal gut composition for expression, maturation and proliferation of microglia. In this regard, studies on germ-free rodents with altered commensal population displayed atypical microglia and disturbed activity of the immune system as a result, which were both corrected after recolonizing gut microbial populations (Erny et al., 2015). Importance of gut microbes, their metabolites and mucosal barrier integrity in neurodevelopmental conditions such as ASD has been reflected by the presence of altered composition of gut microbiota and intestinal permeability in autistic children compared to healthy controls (de Magistris et al., 2010; Matta et al., 2019). In addition, Matta and colleagues showed that both microglia characteristics and ASD phenotypes improve following recolonization of the gut with normal microbiota (Matta et al., 2019). This issue has led to the introduction of microbiota-related treatment approaches like probiotics in the management of ASD behavioral symptoms.

Although there is mounting evidence linking ASD with gut microbiota and immune system independently, data of the interaction between these three systems together remains limited. Therefore, in this paper, we review recent evidence regarding immune modulation of the microbiome-gut-brain axis as a possible critical pathway in mediating ASD. First, we review the literature focusing on the ASD-related GI problems, including microbiota alterations, altered intestinal permeability and altered microbial metabolites profile and we propose a number of factors that could result these conditions. Then, we explain the presence of immune dysregulation in ASD and discuss the immunologic signaling pathways that interconnect the gut problems with CNS dysfunction. Finally, all the discussions end with potential clinical advances that could be achieved in the near future as well as a hypothetical model of the system’s biology in ASD.

Section snippets

Gastrointestinal system and ASD

The prevalence of GI symptoms in ASD patients is widely variable, ranging from 9% to as far as 90% (Vuong and Hsiao, 2017). However, it is now considered a fact that ASD patients suffer from significantly higher rates of GI symptoms (e.g., chronic diarrhea and or constipation, abdominal pain, gastroesophageal reflux disease) compared to the healthy persons. Endoscopy examination of the GI tract has revealed several findings in this respect, including but not limited to, patchy inflammation,

Immune dysregulation and ASD

There is growing evidence on the association between ASD and altered activity of the immune system. First, coexistence of ASD and various immune-based disorders (i.e., inflammatory bowel diseases, allergy, diabetes type 1) has been repeatedly addressed in recent years (Onore et al., 2012; Wang et al., 2014). Second, positive family history for autoimmune disorders has been linked with a higher risk of autism (Atladottir et al., 2009; Molloy et al., 2006; Vinet et al., 2015). Third, specific

Microglia

Microglia colonize the brain in the first days of embryo development and contribute to the regulation of synaptic formation (Reemst et al., 2016). These cells are much involved in fine pruning of dendritic spines and trees as well as the gross apoptosis of neurons (Kettenmann et al., 2013). Besides, they also release chemical transmitters such as glutamate and ATP that help in modulating the synaptic activity (Araque and Navarrete, 2010). Hence, the intercommunication between microglia and

The NF-κB signaling pathway

Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) is a transcription factor acting on multiple aspects of innate and adaptive immune responses. NF-κB increases the expression of pro-inflammatory genes, regulates the proliferation and differentiation of a wide range of immune cells and is considered as a mediator linking immunological etiologies to immune cells (Liu et al., 2017). It has been indicated that NF-κB signaling mediates the association between alterations in the

Clinical implications of the gut-brain axis in ASD

A significant proportion of the literature addresses the gut microbiota as an appropriate target to alleviate behavioral disturbances related to the ASD. For instance, psychobiotics were shown to alleviate not only GI problems but also behavioral abnormalities in individuals with ASD. Psychobiotics are in fact a group of probiotics that have the capacity to influence the CNS related functions and behaviors. The effects of these reagents in the ASD course have been investigated by a vast number

Concluding Remarks and Future Perspectives

ASD patients usually show limited response to contemporary treatments since the underlying etiology remains unclear. There are ongoing efforts to resolve the existing uncertainties in the implicated mechanisms. In the current report, we reviewed the recent evidence for the link between gut microbiota, immune system and CNS in ASD. Autism symptoms are not limited to behavioral abnormalities and multiple organs are affected by the disease. There is a complex interaction between these

Funding

The authors received no financial support for the research, authorship, and/or publication of this article.

Declaration of Competing Interest

The authors of this manuscript declare that they have no conflicts of interest.

Acknowledgments

None.

References (149)

  • E. Esnafoglu et al.

    Increased serum zonulin levels as an intestinal permeability marker in autistic subjects

    J. Pediatr.

    (2017)
  • L. Fernandez de Cossio et al.

    Prenatal infection leads to ASD-like behavior and altered synaptic pruning in the mouse offspring

    Brain Behav. Immun.

    (2017)
  • S.M. Finegold et al.

    Pyrosequencing study of fecal microflora of autistic and control children

    Anaerobe

    (2010)
  • R.P. Gaykema et al.

    Brain response to cecal infection with Campylobacter jejuni: analysis with Fos immunohistochemistry

    Brain Behav. Immun.

    (2004)
  • T. Goldmann et al.

    Fine-tuning of type I IFN-signaling in microglia — implications for homeostasis, CNS autoimmunity and interferonopathies

    Curr. Opin. Neurobiol.

    (2016)
  • A. Haghikia et al.

    Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine

    Immunity

    (2016)
  • J.A. Hall et al.

    The role of retinoic acid in tolerance and immunity

    Immunity

    (2011)
  • E.Y. Hsiao et al.

    Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders

    Cell

    (2013)
  • I.I. Ivanov et al.

    Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine

    Cell Host Microbe

    (2008)
  • F.N. Kaufmann et al.

    NLRP3 inflammasome-driven pathways in depression: clinical and preclinical findings

    Brain Behav. Immun.

    (2017)
  • H. Kettenmann et al.

    Microglia: new roles for the synaptic stripper

    Neuron

    (2013)
  • J.-H. Kim et al.

    Implication of intestinal VDR deficiency in inflammatory bowel disease

    Biochim. Biophys. Acta Biomembr.

    (2013)
  • L.S. Liu et al.

    A rat model of chronic gastric sensorimotor dysfunction resulting from transient neonatal gastric irritation

    Gastroenterology

    (2008)
  • R.A. Luna et al.

    Distinct microbiome-neuroimmune signatures correlate with functional abdominal pain in children with autism spectrum disorder

    Cell. Mol. Gastroenterol. Hepatol.

    (2017)
  • M. Lyte et al.

    Induction of anxiety-like behavior in mice during the initial stages of infection with the agent of murine colonic hyperplasia Citrobacter rodentium

    Physiol. Behav.

    (2006)
  • N.V. Malkova et al.

    Maternal immune activation yields offspring displaying mouse versions of the three core symptoms of autism

    Brain Behav. Immun.

    (2012)
  • S.M. Matta et al.

    The influence of neuroinflammation in autism spectrum disorder

    Brain Behav. Immun.

    (2019)
  • M.M. Meeking et al.

    Propionic acid induced behavioural effects of relevance to autism spectrum disorder evaluated in the hole board test with rats

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2020)
  • J.T. Morgan et al.

    Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism

    Biol. Psychiatry

    (2010)
  • J.T. Morgan et al.

    Abnormal microglial-neuronal spatial organization in the dorsolateral prefrontal cortex in autism

    Brain Res.

    (2012)
  • K. Nemani et al.

    Schizophrenia and the gut-brain axis

    Prog. Neuro-Psychopharmacol. Biol. Psychiatry

    (2015)
  • M.A. Odenwald et al.

    Intestinal permeability defects: is it time to treat?

    Clin. Gastroenterol. Hepatol

    (2013)
  • C. Onore et al.

    The role of immune dysfunction in the pathophysiology of autism

    Brain Behav. Immun.

    (2012)
  • J.B. Adams et al.

    Nutritional and metabolic status of children with autism vs. neurotypical children, and the association with autism severity

    Nutr. Metab.

    (2011)
  • C.B. Appleyard et al.

    Pretreatment with the probiotic VSL# 3 delays transition from inflammation to dysplasia in a rat model of colitis-associated cancer

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2011)
  • A. Araque et al.

    Glial cells in neuronal network function

    Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci.

    (2010)
  • N. Arpaia et al.

    Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation

    Nature

    (2013)
  • M. Asadabadi et al.

    Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial

    Psychopharmacology

    (2013)
  • P. Ashwood et al.

    Intestinal lymphocyte populations in children with regressive autism: evidence for extensive mucosal immunopathology

    J. Clin. Immunol.

    (2003)
  • P. Ashwood et al.

    Spontaneous mucosal lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms: mucosal immune activation and reduced counter regulatory interleukin-10

    J. Clin. Immunol.

    (2004)
  • K. Atarashi et al.

    Induction of colonic regulatory T cells by indigenous Clostridium species

    Science (New York, N.Y.)

    (2011)
  • H.O. Atladottir et al.

    Association of family history of autoimmune diseases and autism spectrum disorders

    Pediatrics

    (2009)
  • P. Bercik et al.

    The anxiolytic effect of Bifidobacterium longum NCC3001 involves vagal pathways for gut-brain communication

    Neurogastroenterol. Motil.

    (2011)
  • G. Bjorklund et al.

    Immune dysfunction and neuroinflammation in autism spectrum disorder

    Acta Neurobiol. Exp.

    (2016)
  • J.A. Bravo et al.

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

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

    (2011)
  • A.J. Bruce-Keller et al.

    Maternal obese-type gut microbiota differentially impact cognition, anxiety and compulsive behavior in male and female offspring in mice

    PLoS One

    (2017)
  • D.B. Campbell et al.

    A genetic variant that disrupts MET transcription is associated with autism

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

    (2006)
  • P.D. Cani et al.

    Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability

    Gut

    (2009)
  • F. Carbonero et al.

    Microbial pathways in colonic sulfur metabolism and links with health and disease

    Front. Physiol.

    (2012)
  • J. Croonenberghs et al.

    Increased serum albumin, gamma globulin, immunoglobulin IgG, and IgG2 and IgG4 in autism

    Psychol. Med.

    (2002)
  • Cited by (13)

    • Autism, heparan sulfate and potential interventions

      2022, Experimental Neurology
      Citation Excerpt :

      CK869 acts as an inhibitor by binding to the Arp2/3 complex in a hydrophobic pocket on the Arp3 subunit, which causes allosteric changes, thereby destabilizing the active conformation of Arp2/3. Considering that neuroinflammation has been indicated in the pathology of ASD (Ye et al., 2021; Moradi et al., 2021; Vargas et al., 2005), treatments that target neuroinflammation may be viable therapeutic option. While HSPGs are known to mediate neuroinflammation in various pathological states (O'Callaghan et al., 2018; Collins and Troeberg, 2019), it's not clear how a deficiency in HS may impact neuroinflammation in ASD.

    • Melatonin improves learning and memory of mice with chronic social isolation stress via an interaction between microglia polarization and BDNF/TrkB/CREB signaling pathway

      2021, European Journal of Pharmacology
      Citation Excerpt :

      Recent literature has related these dysfunctions after chronic stress to alterations in the immune system (Lehmann et al., 2018). Resident microglia are the key cellular components of the neuroimmune system that undergo several modifications in response to external and internal stressors (Moradi et al., 2020a). In this manner, one of the major concerns is the function of stimulated M1 microglia, whereby pro-inflammatory activities are provoked.

    • Gut microbiota in psychiatric disorders: Better understanding or more complexity to be resolved?

      2021, Progress in Neuro-Psychopharmacology and Biological Psychiatry
    • Targeting the microbiome-gut-brain axis for improving cognition in schizophrenia and major mood disorders: A narrative review

      2021, Progress in Neuro-Psychopharmacology and Biological Psychiatry
      Citation Excerpt :

      This may also apply to cognition and the MGB axis. Recent studies have revealed a relationship between the cognitive abilities of neurodevelopmental disorders and the MGB axis (Moradi et al., 2020). How gut microbes and their metabolites are associated with cognitive domains in autism, remains still far from definitive.

    View all citing articles on Scopus
    View full text