Next Article in Journal
Antibiotics as Adjunctive Therapy in the Non-Surgical Treatment of Peri-Implantitis: A Systematic Review and Meta-Analysis
Next Article in Special Issue
The Antifungal Antibiotic Filipin as a Diagnostic Tool of Cholesterol Alterations in Lysosomal Storage Diseases and Neurodegenerative Disorders
Previous Article in Journal
Use of a Novel Anti-Infective Noble Metal Alloy-Coated Titanium Orthopedic Nail in Patients with Open Fractures: A Case Series from Malaysia
Previous Article in Special Issue
Antimicrobial Peptides (AMPs) in the Pathogenesis of Alzheimer’s Disease: Implications for Diagnosis and Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exposure to Antibiotics and Neurodevelopmental Disorders: Could Probiotics Modulate the Gut–Brain Axis?

1
Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, 00185 Rome, Italy
2
Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy
*
Author to whom correspondence should be addressed.
First author.
These authors contributed equally to this work.
Antibiotics 2022, 11(12), 1767; https://doi.org/10.3390/antibiotics11121767
Submission received: 4 November 2022 / Revised: 30 November 2022 / Accepted: 5 December 2022 / Published: 7 December 2022
(This article belongs to the Special Issue Antibiotics as Tool to Investigate Cell Functional State)

Abstract

:
In order to develop properly, the brain requires the intricate interconnection of genetic factors and pre-and postnatal environmental events. The gut–brain axis has recently raised considerable interest for its involvement in regulating the development and functioning of the brain. Consequently, alterations in the gut microbiota composition, due to antibiotic administration, could favor the onset of neurodevelopmental disorders. Literature data suggest that the modulation of gut microbiota is often altered in individuals affected by neurodevelopmental disorders. It has been shown in animal studies that metabolites released by an imbalanced gut–brain axis, leads to alterations in brain function and deficits in social behavior. Here, we report the potential effects of antibiotic administration, before and after birth, in relation to the risk of developing neurodevelopmental disorders. We also review the potential role of probiotics in treating gastrointestinal disorders associated with gut dysbiosis after antibiotic administration, and their possible effect in ameliorating neurodevelopmental disorder symptoms.

Graphical Abstract

1. Introduction: Neurodevelopmental Disorders

Brain development is a dynamic process resulting from a constant interplay between genetic and environmental factors [1]. Neurodevelopmental disorders (NDDs) are a group of heterogeneous syndromes with functional impairments in the central nervous system (CNS) caused by the disruption of essential processes during neurodevelopment [2]. Brain dysfunctions during development reflect deficits in social communication, verbal, nonverbal, and social interactions and are characterized by repetitive behaviors and activities [3]. NDDs include autism spectrum disorder (ASD), intellectual disability (ID), attention deficit hyperactivity disorder (ADHD), epilepsy [4], and schizophrenia (SCZ). To date, there has been a worldwide increase in the prevalence rates of NDDs: ID, 0.63%, ADHD, 5–11%, ASD, 0.70–3%, and SCZ 0.32% [5,6,7,8,9,10]. Although NDDs tend to run in families, as suggested by twin and family studies, the inheritance patterns are complex and still under investigation [11]. Genetic alterations including de novo mutations, and rare and common copy number variants [12,13,14,15,16,17,18] have been found to be an emerging source of genetic causality [19]. Despite the huge amount of risk in genes found in the human population, there are overlapping functions that affect a limited number of biological pathways [20]. A relevant and recurrent pathway is represented by the synapse function along with CNS development. Risk genes involved in the synaptic pathway are found expressed in dendrites, axons, and pre- and postsynaptic terminals, playing different roles [21,22]. There are several genetic mutations found in synaptic genes identified in individuals, encoding for proteins including NLGNs, SHANK2, SHANK3, and CNTNAP2, providing a direct link with the synaptic function and the etiology of NDDs [23,24,25,26,27]. Although recent new technologies favor research advancements in the genetic background of NDDs, they cannot provide information about the environmental influences on genetic predispositions [28,29]. It is well established that environmental factors may alter and/or trigger childhood psychiatric conditions [30,31]. A recent rise in the prevalence of NDDs suggests a major involvement of the environment during different critical developmental windows that can be classified into prenatal, perinatal, and postnatal [32] windows. Early-life environmental injurious factors could be represented by premature birth, low birth weight, environmental contaminants, fetal exposure to smoking, alcohol, drugs, or medications during pregnancy [33,34,35]. Antibiotics are a class of drugs to which both mothers and children are exposed during pregnancy. Treatments based on antibiotics have been considered to provide minimal adverse effects on health, although epidemiologic studies have shown that early-life exposure increases the risk of obesity, asthma, and celiac disease in children [36,37,38,39]. Antibiotics have been proven to cause multiple birth defects, when administered to pregnant mothers, and are associated with several diseases in newborns by increasing the risk of congenital malformations [40,41]. Antibiotics affect the brain directly or could have an indirect effect via their actions on the human microbiome, causing long-term consequences [42,43,44].

2. Gut–Brain Axis

The two-way street between gut and brain has been known since ancient Grecian times, when Hippocrates affirmed that “All disease begins in the gut”, but only in 1870 was the existence of a gut–brain axis established [45]. This concept has been extended to the microbiota residing in the gastrointestinal (GI) tract, termed gut microbiota, which have been shown to be a key regulator of the gut–brain axis, and thus the microbiota–gut–brain axis has been proposed [46,47]. The relationship between gut and microbiota shows bidirectional communication with the CNS through the production of neurotransmitters, innervation via the vagus nerve, or activation of the hypothalamic–pituitary–adrenal axis [48]. Multiple factors, including mode of delivery, lifestyle, antibiotic usage, diet, age, gender, and geographic localization affect and modify the profile of the trillions of indigenous microorganisms, including bacteria, fungi, and viruses, that colonize the GI tract from birth throughout the lifespan. Additionally, gradients of pH, oxygen, antimicrobial peptides, and bile salts determine the density and diversity of microbial species along the GI tract of the host. Nowadays, it is clear that neurotransmitters such as glutamate, GABA, serotonin, and dopamine, short-chain fatty acids (SCFAs), cytokines, neuropeptides, endocannabinoids, and hormones are the molecules used for the communication between the brain and the gut through the endocrine, immune, and neural systems in order to maintain homeostatic conditions [47] in our body. Gut microbiota [48,49,50] synthesizes a vast milieu of metabolites that may directly or indirectly impact neuronal activity as well as induce host cells to produce signaling molecules, shaping both local and extraintestinal host functions [51]. Among the neuroactive molecules, SCFAs, including butyrate, acetate, and propionate, are metabolites produced from the bacterial fermentation of nondigestible fibers in the gut [52]. These play several functions in maintaining gut homeostasis, including anti-inflammatory properties. Their effects are mainly mediated by G-protein coupled receptors [53] and can also trigger regulation of gene expression and epigenetic modulation by histone deacetylases, as reported for butyrate [54].
In particular, SCFAs strongly stimulate the release of gut hormones in enteroendocrine cells [55], whereas in immune cells, selected strains might regulate T regulatory cell differentiation [56,57,58,59] and myeloid cell lineages in the bone marrow, and might affect the function of circulating mature granulocytes [59]. It has been shown that short-chain fatty acid (SCFA) levels were altered in ASD patients due to alterations in the gut microbiota composition, and administration of butyrate has been shown to improve ASD symptoms [60]. Moreover, a reduction of SCFA levels can affect both intestinal barrier and blood–brain barrier (BBB) integrity, leading to neuroinflammation with direct implications in brain functions [61]. In immunometabolism, gut microbes promote peripheral immune responses associated with CNS disorders through driving, for instance, inflammatory Th17 cell responses [62]. Long-range effects on CNS-resident immune cell function, can be promoted following the release of bacterial metabolites, such as cytokines, into the bloodstream and lymphatic systems [59]. On the other hand, the loss of intestinal barrier integrity due to gut inflammation can activate innate and adaptive immune cells to release pro-inflammatory cytokines IL-1β, IL-6, and tumor necrosis factor-α (TNFα) into the circulatory system, leading to systemic inflammation. In the last decade, the metabolism of tryptophan is gaining attention in the microbiota–gut–brain axis because this essential amino acid serves as a precursor to a variety of imperative bioactive molecules generated by both the enterochromaffin cells of the host and gut microbes [63]. Moreover, 5-hydroxytryptamine (5-HT), commonly known as serotonin, is the main product of tryptophan conversion and, incidentally, it has been demonstrated that some bacteria found in gut microbiota are able to synthesize 5-HT, affecting thus the plasma levels of this neurotransmitters [64]. It is true that 5-HT has a pivotal role in the regulation of several functions both at the intestinal and central level, such as modulation of mood, memory, and cognition [65]. It has been found that altered levels of 5-HT, mainly synthesized in the gut, are implicated by different diseases, such as irritable bowel syndrome (IBS) as well as humor disorders (i.e., anxiety and depression-like disorders) [66] and ASD [67]. During inflammation or in response to stress, tryptophan metabolism shifts to kynurenine production, leading to the formation of either kynurenic acid or quinolinic acid. The balance between these compounds is crucial in psychiatric and neurological disorders; indeed, kynurenic acid has a neuroprotective role, and quinolinic acid is involved in neurotoxicity. A variety of health benefits for the neurotransmitter of GABA have been shown, including neurostimulation and gut modulation [68,69]. The GABAergic system has an important implication also in the NDDs and neurodegenerative disorders, in terms of alterations of GABA concentrations and GABA receptor expression [70,71]. A body of evidence has associated alterations in the gut–brain axis with drug and antibiotics therapies and, in turn, altered levels of specific neuroactive molecules in different types of NDDs [60,72]. It is now clear that the gut–brain axis plays an active role in the neurodevelopmental processes, including the establishment of BBB, neurogenesis, maturation of microglia, and myelination [73] with long-term health effects (Figure 1).

3. NDD-Associated Gastrointestinal Symptoms

Recently, gut microbiota has assumed considerable importance for its function in influencing brain functions, including social behaviors [74]. Individuals with NDDs show different compositions of the intestinal microbiome in terms of the number and types of species. A balanced and appropriate composition of the gut microbiota confers health benefits; a disruption of this balance can reflect on brain function and behavior by acting through the microbiome–gut–brain axis [46], our “second brain” [45]. The relative abundance of the single species constituting the microbiota and its metabolites are associated with the onset of neurologic and psychiatric disorders [75]. GI disorders (ranging from severe constipation to diarrhea) are frequently observed in individuals with ASD; however, they are estimated to occur with a wide variability, from 2.2% to 96.8% of the ASD population [76]. Despite this considerable heterogeneity, overall, most studies highlight a greater prevalence of GI problems in children with ASD compared with their neurotypical counterparts, suggesting that microbial dysbiosis could contribute to gut symptoms in neurodevelopmental disorder (NDD)-affected individuals [77,78]. Moreover, a compromised gut epithelial barrier (so-called “leaky gut”) has been described in association with GI problems in NDDs [79]. This will enable bacteria and metabolites to cross the GI barrier and trigger aberrant immune responses. Accordingly, elevated levels of inflammatory cytokines in children with ASD have been reported in association with symptom severity [80]. Cytokine levels have also been correlated with ASD-associated bacterial populations (e.g., Clostridiales) and GI symptomatology [81,82]. Differences in microbial composition among ADHD Dutch young patients were found by sequencing 16S rRNA extracted from fecal samples [83,84,85]. A significant decrease in the gut microbial diversity in ADHD children has been reported: an unusually higher level of the family Bacteroidaceae, Neisseriaceae that causes a significant decline in the gut microbial diversity in ADHD patients, differing from a higher level of Prevotellaceae, Catabacteriaceae, and Porphiromonadaceae found in the control group [86]. An altered Bifidobacterium population during early childhood has been correlated with a high risk of developing ADHD [87,88].
Among the NDDs, SCZ is highly heterogeneous with a genetic and epigenetic component. SCZ-affected individuals are characterized by gut microbiota dysbiosis with consequential GI problems like gastroenteritis, colitis, and IBS [89,90,91,92]. Altered species diversity identifies the gut microbiota in SCZ patients [93] causing cognitive and functional anomalies [94]. However, higher levels of oropharyngeal microbial species such as Bifidobacterium dentium, Lactobacillus oris, Veillonella atypica, Dialister invisus, Veillonella dispar, and Streptococcus salivarius were found in SCZ individuals with respect to healthy controls [95,96]. In NDD individuals, there is a significant decrease or complete absence of Bifidobacteria, with respect to the control subjects [97]. A deficit of intestinal Bifidobacteria has been correlated with indigestion, vitamin B12 deficiency, dysregulated immune system, gut inflammation, depression, and anxiety-like behaviors [98,99]. The microbiota can be included among the wide range of environmental factors affecting neurodevelopment. Indeed, microbiota disruption due to antibiotics administration can impact neurogenesis and behavioral deficits [100]. Alteration in the gut microbiome composition has been observed in several NDDs conditions, including depression [101], autism [102], and other conditions [103]. Mice models have been extensively used to understand the contribution of the microbiota on behavior and how gut dysbiosis may contribute to development of NDDs. Germ-free mice colonized with fecal microbiota from children with ASD show increased autistic-like behaviors in comparison to controls [104]. Moreover, specific bacteria groups, such as Clostridiaceae, Lactobacillales, Enterobacteriaceae, and Bacteroides, were found to be enriched in the ASD gut microbiota composition in mice in comparison with the control microbiota. Interestingly, mice that were treated with gut microbiota from ASD patients displayed an alternative splicing in the brain of ASD-related genes [105]. Several mutations have been reported in risk genes for NDDs, genetic mouse models have been generated by introducing the human mutation in the endogenous murine gene, whereas environmental models of ASDs are characterized by the exposure to a specific external influence [106]. Both types of mice models have been considered for studying how the genetic or environmental modification impacts on the gut–brain axis [107]. The R451C mutation, mapping in the postsynaptic protein neuroligin3 (NLGN3), was found in two affected brothers [108] who, beyond their behavioral deficits, displayed GI dysfunction including chronic gut pain, diarrhea, and esophageal regurgitation [109]. The mouse model, expressing the human R451C NLGN3 mutation, displays in vivo small intestinal motility and increased numbers of myenteric neurons in the small intestine, suggesting that the mutation alters enteric nervous system (ENS) function and structure [109]. In addition, R451C NLGN3 mutation alters mucus density and the spatial distribution of bacteria species in the GI tract of mice [110]. Additionally, in the knockout (KO) mouse model for NLGN3, gut dysfunction is characterized by a faster colonic motility and an increased colonic diameter, although GI structure and enteric neuron populations were unaltered [111]. Similarly to the alterations observed in mice models for NLGN3, the deletion of Shank3, a leading ASD candidate gene encoding for a neuronal scaffold protein implicated in the organization of the synapse through several protein–protein interactions among which are neuroligin-1, alters gut function and the microbiome [112,113]. SHANK3αβ KO mice show altered GI morphology and display differences in the composition of fecal microbiota [112]. Mutations in chromodomain helicase DNA binding protein 8 (CHD8) are among the most common de novo mutations associated with ASD in the developing brain [114]. Mutations in CHD8 increase susceptibility to GI issues in affected individuals [115,116]. CHD8 mutations associated with ASDs caused lower intestinal motility when expressed in zebrafish [115]. The CHD8+/−mouse model has a shorter intestine [117], the width of the mucus layer is lower in the small intestine of CHD8+/− mouse, and the number of goblet cells is reduced. Moreover, it was found that the CHD8+/− mouse has a higher bacterial load and microbiota diversity in the colon tract with respect to WT mice [118]. Fragile X syndrome is an NDD caused by a mutation in the X-linked FMR1 gene [119]. In the Fmr1 KO2 mouse, the gut microbiome is altered and associated with different bacterial species population, belonging to genera Akkermansia, Sutterella, Allobaculum, Bifidobacterium, Odoribacter, Turicibacter, Flexispira, Bacteroides, and Oscillospira [120]. A nongenetic, idiopathic ASD mouse model is the black and tan brachyury mouse strain (BTBR), which is characterized by repetitive behavior and social deficits [121]. The BTBR mouse model displays a marked intestinal dysbiosis with respect to the WT strain. The GI profile exhibits an altered gut microbial composition, altered social behavior, increased gut permeability, and colon proinflammatory biomarkers [122]. During pregnancy, alterations of the maternal gut can influence the microbial diversity and immunity in the offspring, predisposing them to the onset of NDD conditions [123,124,125]. This is the case of the maternal immune activation (MIA) mouse model, in which pregnant mice are administered with potent immune activator and generate pups with ASD-like behaviors [126]. MIA mice show decreased intestinal barrier integrity, dysbiosis of the intestinal microbiota and neurodevelopmental abnormalities in the offspring [127]. MIA offspring display an altered serum metabolomic profile, increased gut permeability, and abnormal intestinal cytokine profiles, such as the IL-6 [128], IL-1β, and TNF-α and also the total number of bacteria is significantly reduced in MIA offspring [129].
The influence of the microbiota composition and its effects on gut–brain communication is not fully clear and involves multiple mechanisms and factors. The microbiota colonization is a pivotal event; the gut microbiota changes during pregnancy, and maternal antibiotic administration during lactation influences the milk composition, which can affect the infant gut microbial composition [130]. In other words, the human gut microbiota exhibits distinct and singular metabolic traits characterized by a maternal signature [131]. At present, it is not possible to define a single bacterium as a hallmark of NDDs despite the significant increase, decrease, or complete absence of species found in affected individuals and mouse models of disorders. The increased or decreased abundance of gut microbes can be correlated with disrupted GI mucosal barrier, pathological intestinal conditions, and decreased immune surveillance, due to altered gut metabolite production. It is important to underline that some bacteria species have positive or negative effects on the gut–brain axis outcomes in humans and in mouse models and the same components of the gut microbiota do not have the same effect on different individuals, as suggested by the evidence obtained in both genetic and environmental NDD mouse models; in fact, single bacteria species may be important either for health or disease conditions.

4. Prenatal Antibiotics Exposure and the Risk of ASD

Pregnancy is a crucial period for fetal brain development. Embryonic brain development can be divided into three main stages: the first trimester, which is characterized by the formation of the neural tube and the production of neural progenitor cells and neurons; the second trimester, when neurons migrate to the cortical layer and begin to form synaptic connections; and finally, the third trimester, during which neuronal axons, glia and oligodendrocytes, are integrated into neural circuits [132]. During the prenatal period, the fetal brain is particularly sensitive to the surrounding environmental stimuli impacting on the CNS development [133]. A possible disturbance of the prenatal environment due to drug exposure, such as antibiotic administration, might cause neurodevelopmental alterations and subsequently lead to the onset of NDDs. Antibiotic treatment during pregnancy is continuously increasing as intrapartum prophylaxis; however in utero exposure to treatments may affect the newborn [134,135,136]. The most frequently prescribed antibiotics are “macrolides’’ that include erythromycin, azithromycin, and clarithromycin [137]. A systematic study on the use of macrolides, during the first trimester of pregnancy, showed consistent evidence of an increased risk of abortion and cardiovascular fetus malformation [40]. A large epidemiological work associates maternal infection and the use of antibiotics during pregnancy with an increased risk of developing NDDs [138] and altered brain functions in the offspring. However, it is still unclear how the exposure to antibiotics in utero affects the maturation of the gut microbiome from the fetal to the adult and the development of CNS in children. A long-term follow-up in children whose mothers took part in the “ORACLE II’’ trial of antibiotics, showed an increased prevalence of cerebral palsy associated with antibiotic treatment [138]. Neural tube defects were reported more frequently in children from women exposed, for 12 weeks before conception, to trimethoprim, an antibiotic that blocks the dihydrofolate reductase enzyme responsible for the conversion to folate [139], which is necessary for the closure of the neural tube during development. This is indicating a direct correlation between the treatment of the mothers and the newborn. Longer treatments increased the correlation between antibiotics and ADHD development [140]. Population-based studies showed an association between the maternal use of antibiotics during pregnancy and ASD in children [141,142]. Two main studies involved 96,736 and 780,547 children, respectively. The first study reported a significant increase in the risk of ASD after treatment with antibiotics during pregnancy [142]. In the second one, a strong association between prenatal exposure to antibiotics was correlated with the risk of ASD [141]. The associations between antibiotic exposure and later development of NDDs could reflect the direct effects on the gut–brain axis. Prenatal exposure was associated with a 32–41% (hazard risk ratios (HR) = 1.32–1.41) increased the possibility of sleep disorders, whereas the risk estimates for NDDs increased from 12% to 53%. The analysis has included antibiotics used against airway, urinary tract, skin and, soft tissue infections [143].
To deeply explore the contribution of maternal gut microbiota during pregnancy on newborn brain development, researchers have used animal models treated with antibiotics known to alter the maternal GI tract. Tochitani et al. showed that the administration of antibiotics to pregnant dams, during the embryonic stage (E9–E16), perturbs the maternal composition of the gut microbiota and flora in the offspring at the stage of postnatal period (P24) and this affects social behavior and locomotor activity with respect to control animals [144]. Similar evidence comes from a recent study where C57BL/6J mouse dams were exposed to antibiotic treatment dissolved in drinking water from gestational day 12 through offspring of the postnatal period (P14). Male and female offspring display ASD-like behaviors, including alteration in ultrasonic vocalization production during maternal separation and altered offspring thermoregulation in comparison to age-matched control [145].
During pregnancy, the role of the gut perturbation, due to antibiotic administration, on brain development and CNS dysfunction has been studied on rat models. Females were exposed, during the gestational period, to antibiotics that proved to alter the social behavior of the offspring [146]. In the same study, pups exposed to prenatal antibiotic treatment displayed anxiety-like behavior and greatly reduced social interactions [146]. Voung and colleagues identify that early mid-gestation is a critical period during which the maternal microbiome promotes fetal thalamocortical axonogenesis in the offspring in order to support developmental processes regulating behaviors in adult mice [147]. Taken together, these findings support the influence of maternal stimuli on fetal development; however, the molecular pathways implicated, and the metabolites involved, still remain unclear.

5. Early Antibiotics Exposure and the Risk of ASD

Brain plasticity is the change in neuronal networks in response to various stimuli that can permanently shape the brain [148]. Neuronal plasticity is sensitive to internal and/or external stimuli, and the interaction between genes and the environment are influenced by a variety of factors [149]. Childhood and adolescence are pivotal periods for brain development which include critical events such as neurogenesis, axonal dendritic growth, and synaptogenesis [150]. Antibiotics, often essential for treating infections in an early stage of life, can promote long-lasting adverse effects on brain development [151,152,153,154,155]. The use of antibiotics in children who develop NDDs was shown by several studies [156,157,158]. A higher risk of developing severe mental disorders at an adult age was found in children and adolescents treated with antibiotics, with the most pronounced effects observed by the use of a broad and moderate spectrum of antibiotics [159,160]. Early-life antibiotic exposure has been linked to a lower intelligence quotient and social scores, as well as higher behavioral difficulty scores, suggesting that it may represent a risk factor for ADHD, depression, and anxiety disorders [161]. ADHD is one the most common NDDs, and several studies have demonstrated that the exposure to antibiotics in early life alters the equilibrium in the gut microbiota and contributes to the development of ADHD [162]. The window between 0 and 2 years represents an important period in brain development, wherein changes, modification, and organization of the brain occur more and more rapidly than at any time during childhood [163]. The relationship between antibiotic exposure, in the first 2 years of life, and cognitive deficits was examined in a cohort of 342 children at the age of 11. This study showed that toddlers treated with antibiotics had an increased risk of developing behavioral deficits and depression symptoms during childhood [164]. The association between the use of antibiotics during early life, ADHD, and ASD was also studied in twins from 7 to 12 years old in the Netherlands and in 9-year-old twins from Sweden. In both studies, children that were temporarily exposed to antibiotics between 0 and 2 years (any pharmaceutical formulation, oral or intravenous, defined as parent-reported) resulted in an increased risk of ADHD and ASD; however the importance of the familial environment and the genetics influence in the etiology of NDDs has also to be considered [165]. Children exposed in the first two years of life to the most prescribed antibiotic classes (penicillins, cephalosporins, and macrolides that markedly impact on microbiota composition (see Table 1)) were more likely to develop asthma and allergic rhinitis and atopic dermatitis and ADHD [36]. The effect of postnatal exposure to penicillin was tested on a cohort of 677,403 children that resulted in an increased risk of developing ASD in comparison to untreated children [166]. Children are more susceptible to bacterial infections than adults, and severe infections regarding the nervous system during childhood might also result in the onset of NDDs later in life [167,168,169,170,171]. Infections treated with antibiotic drugs, and infections requiring hospitalizations in particular, were associated with increased risks of SCZ and psychiatric disorders, which may be mediated by effects of infections/inflammation in the brain, alterations of the microbiome, genetics, or other environmental factors. The connection between infections during periods of rapid growth, brain plasticity and diagnosis of NDDs may be explained by microbial metabolites, produced by the enteric bacteria, interfering with normal brain development. During normal conditions, gut metabolites produced by Lactobacillus and Bifidobacterium spp. produce the inhibitory neurotransmitter GABA affecting its activity in the brain. Antibiotic treatment alters the production of metabolites, such as SCFAs or amino acids, that may lead to dysfunction of the epithelial barrier in the intestine and BBB in the brain. Individuals with NDDs were more likely to have experienced severe infections of CNS from age 0–3 years [172]. A study on a Danish population confirms these observations: a wide range of NDDs in relation to previous CNS infections are reported and a significant association (HR 3.29) with developing ID and ASD [173]. Otitis media is one of the most common infections in childhood with high incidence in children with ASD [174]. Wimberley and colleagues found an increased case of ASDs associated with otitis media infection and antibiotics treatment in a study based on a Danish cohort of 780,547 children [141]. The correlation between infections and antibiotic treatment may contribute to a later diagnosis of NDDs [167]. Antibiotics used to treat gastroenteritis caused by Shigella infection, when administered at a young age (5–18 years), increase risks of developing ADHD respective to children who did not [175], confirming that the antibiotic treatment by affecting the human microbiome, plays an important role in developing NDDs [176].
The effects of early exposure to antibiotics on gene expression and behaviors has been widely documented in several in vivo studies using the murine model [177,178,179,180]. Leclercq and colleagues exposed mice of both sexes to a low dose of penicillin in late pregnancy and early postnatal life and showed changes in anxiety-like and aggressive behavior and decreased sociability [181]. They showed antibiotics treatment caused long-lasting changes in mice gut microbiota and altered BBB integrity in the hippocampus brain region [181]. At the molecular level, antibiotics administration altered the expression of molecules involved in memory and learning like the brain-derived neurotrophic factor (BDNF) that is crucial for promoting neuronal survival and synaptic plasticity [182]. In young mice, disruption of gut microbiota by antibiotics, shows deficits in memory retention and leads to a significant reduction of BDNF production in the hippocampus of the adult brain [183]. Levels of BDNF and its receptor, tropomyosin-related kinase B, are downregulated in the hippocampus and are unchanged in the prefrontal cortex of treated animals [182]. Dysbiosis in mice can be obtained by exposing young animals to a cocktail of a broad spectrum of antibiotics that cause gut inflammation, depressive-like symptoms, social behavior and cognitive deficits, along with changes in brain neuronal firing and microglial–glial activation [178]. A recent study shows that perinatal exposure to antibiotics affects cortical development with a long-lasting effect on brain functions in young mice [184]. Perinatal penicillin exposure significantly increased sensorimotor gating and decreased the ability to discriminate between textures in adolescent mice. These behavioral alterations were accompanied by increased spontaneous neuronal activities and a delayed maturation of inhibitory neuronal circuits [184]. An excess use of antibiotics induced neurotoxic effects on mice brain [185]. Amoxicillin administration, at clinical doses, has been reported to induce depression in young rats [186]. Significant changes in gene expression have been observed in both the frontal cortex and amygdala after 10 days exposure to a low dose of penicillin to postnatal mice [187]. Alterations in the microbiota were more extensive in mice that were exposed to antibiotics during the gestation of the dams, confirming the connection and the transfer between the maternal microbiota and the embryos with respect to mice treated after birth [187]. These results provide evidence that early-life antibiotic exposure in humans and mice have effects not only on the gut microbiome but also on gene expression within critical brain structures, which are vulnerable to perinatal insults [188]. Early-life antibiotic exposure causes unexpected consequences on childhood health; however, these findings require further validation.

6. Probiotics and Use of Probiotics in NDDs

In this paragraph, all the names of lactobacilli are cited according to the original species names, preceding the reclassification of Lactobacillus genus that have published in 2020 [191]; for the current nomenclature please see the following link: http://lactotax.embl.de/wuyts/lactotax/ (last updated on 2 September 2021)). According to the current definition, probiotic bacteria, traditionally belonging to Gram-positive taxa, (i.e., Lactobacilli and Bifidobacteria), are “live microorganisms that, when administered in adequate amounts, as part of a food or a supplement, confer a health benefit on the host” [192]. In the last several decades, probiotics have been largely used as adjuvant in the treatment of several diseases, mainly for the maintenance of a healthy gut environment through their impact on gut microbiota composition, interactions with the intestinal epithelium, and finally with the immune system [193]. So far, the role of probiotics in treating GI disorders is well known, especially for the restoration of gut dysbiosis associated with antibiotic administration, but probiotics have shown the potential to have a broad spectrum of health benefits, ranging from digestive to neurodevelopment and neurodegenerative disorders [65]. In this context, due to the crucial role of the gut microbiota in modulating human brain function via the gut–brain axis [194], a particular class of probiotics, defined with the term “psychobiotic” have shown the ability to specifically confer health benefits at the brain level. As conventional probiotics, psychobiotics can directly modulate the gut microbiota composition and functionality. During their transient colonization at the intestinal level, they can contribute to the maintenance of a healthy gut microbiota by producing growth factors that favor the growth of beneficial microbes, for example during antibiotic therapy, by competing for nutrients and/or producing inhibiting molecules that protect from pathogens colonization, and by interacting with the intestinal mucosa and modulating the intestinal immune system [70]. The most speculated mechanism of action by which psychobiotics exert their beneficial effects for mental health is the production and/or stimulation of different types of neuroactive molecules, previously described, directly involved in the two-way microbiota–brain communication [195]. Lactobacilli and Bifidobacteria are involved in the production of GABA with some Lactobacillus spp. and also in the production of acetylcholine, whereas Bacillus species can stimulate the production of dopamine and noradrenaline. Serotonin has been found to be produced by certain Escherichia, Enterococcus, and Streptococcus species [195,196]. Moreover, the production of neurotransmitters has been reported as a species-specific feature in Lactobacillus genus [197]. Intestinal bacteria can also be involved in modulating neurotransmitter levels by regulating the metabolism of neurotransmitter precursors, as the case of increased plasma tryptophan levels by Bifidobacterium infantis [198] or, for example, by indirectly stimulating the serotonergic system through the production of SCFAs [199]. Interestingly, bacteria from food origins have been recently shown to be able to modulate neurotransmitters [200]. For example, Lactobacillus plantarum DR7 improved stress, anxiety, and cognitive functions by stimulating dopamine and serotonin pathways [201,202], whereas the food-associated L. plantarum C29 showed the ability to improve cognitive functions in adults with mild cognitive impairments [203]. Several studies reported the ability of different probiotics species in restoring neurotransmitter levels in diverse neurological diseases. In Table 2, we summarize several pieces of preclinical and clinical evidence of the use of probiotics in NDDs, and we discuss this evidence below. Lactobacillus rhamnosus (JB-1) has been found to reduce stress-induced corticosterone and anxiety- and depression-related behavior in mice by modulating GABA expression in the brain via the vagus nerve [204]. Similar effects have been reported for Lactobacillus helveticus R0052 and B. longum R0175 effects in rats [205]. Amelioration of depression-like behavior via reduction of 5-HT and dopamine levels in the brain of rats have been found after administration of Bifidobacterium infantis 35624 [206]. Intake of Bacteroides fragilis restores normal 5-HT levels in an ASD animal model [207], whereas L. plantarum of PS128 increased the dopamine level in the prefrontal cortex in early-life stress mice [208] and improved many of the behavioral aspects of ASD, such as disruptive and rule-breaking attitudes and hyperactivity/impulsivity in children [208]. A strain of L. plantarum (MTCC1325) was able to restore acetylcholine also in rats affected by neurodegenerative disorders [209]. Probiotic Lactobacillus helveticus NS8 also showed the ability to modulate neurotransmitters, such as BDNF, serotonin, and noradrenaline in the hippocampus of rats as well as to increase circulating antiinflammatory cytokines, leading to improvement of both the intestinal barrier and the BBB, and in turn ameliorating the global inflammation status [210]. Although the entire mechanism by which probiotics ameliorate diverse NDDs symptoms is still unrevealed, a healthy gut microbiota, and in turn, the maintenance of a proper signaling network from ENS to CNS is recognized to be fundamental for proper brain functions; thus, the use of probiotics and related metabolites as an alternative intervention strategy to ameliorate and/or counteract NDDs is emerging (and clinical trials are increasing but still limited). The generation and the use of ASD animal models has shown not only that the microbiota is essential for development of social behavior [211], but also that restoring normal gut microbiota components with probiotics can correct GI permeability defects. In fact, the altered microbial composition and ASD-related abnormalities are linked to reduced intestinal production and toxin absorption [128]. Probiotic administration has been shown to improve ASD symptoms [212], and to prevent somatic symptoms in SCZ [213], and in drug-resistant epilepsy [214]. Intake of fermented food and probiotics, such as Bifidobacterium spp. and Lactobacillus spp., have been shown to ameliorate psychiatric disorder-related behaviors, including anxiety, depression, obsessive-compulsive disorder, and memory skills, as well as to attenuate stress responses [215]. Two randomized control studies by Santocchi et al. [216,217] evaluated the effects of a diet supplemented with a mix of probiotics, called De Simone Formula (labeled as Vivomixxâ in EU) on the main symptoms of ASD in preschool children with and without GI symptoms. The treatment, which involved the administration of eight probiotic strains, has shown significant effects not only in the improvement of GI symptoms but also in multisensory processing and adaptive functions [217]. Recently, Kalenik et al. reported some randomized trials in which different probiotic strains have been administered in ADHD children to evaluate the effect of probiotic supplementation with the occurrence of ADHD symptoms [218], but 4 out of 5 studies have been shown no substantial differences in cognitive and neurodevelopmental outcomes. However, in one study evaluating the impact of early administration of Lactobacillus rhamnosus GG on the development of ADHD in infants, probiotic supplementation showed a preventive effect in reducing the risk of developing ADHD [87]. Interestingly, in 2019 a promising multinational clinical trial has been started to evaluate the effect of the administration of a symbiotic formula (Synbiotic 2000 Forte 400) containing a mixture of different probiotics (Pediococcus pentosaceus, Lactobacillus paracasei subsp. Paracasei, L. plantarum, and Leuconostoc mesenteroides) and four prebiotics (β-glucan, inulin, pectin, and resistant starch) in a cohort of 180 adults with ADHD [219]. To date, clinical studies applying probiotics in ADHD patients are still limited and future studies are needed to achieve sufficient evidence to recommend probiotics administration as beneficial treatment in ADHD. Probiotics supplementation has been applied as an alternative treatment in SCZ, even though the literature is still limited, and more clinical studies are needed. A systematic review by Ng et al., associates the effect of probiotic administration with the amelioration of side-effect symptoms of SCZ, mainly related to the antipsychotic therapy, such as perturbation of the microbiota composition that may lead to adverse metabolic effects, weight gain, constipation, and finally to systemic inflammation and neuroinflammation [220]. Lactobacillus rhamnosus GG and Bifidobacterium animalis subsp. lactis strain Bb12 have shown to have positive effects in improving intestinal barrier integrity and ameliorating bowel difficulties in patients with SCZ via modulation of inflammatory cytokines belonging to IL-17 family, but no significant impact on positive and negative syndrome scale (PANSS) psychiatric symptom scores [221]. Improvements in constipation and insulin resistance have been found by Nagamine et al. [222] after four weeks of treatment with BIO-THREE®, a mixture of Streptococcus faecalis, Bacillus mesentericus, and Clostridium butyricum, in SCZ patients, whereas similar results with no changes in PANSS were reported also by other clinical studies using the same mixture of probiotics (L. rhamnosus GG and B. animalis subsp. lactis strain Bb12) in patients with psychotic symptoms [213,223]. However, Okubo et al. (2019) reported that the probiotic strain Bifidobacterium breve A-1 was able to improve PANSS scores, depression, and anxiety in SCZ patients after four weeks of treatment. Moreover, the authors correlated the amelioration of symptoms with a modulation of IL-22 and TNF-related activation induced cytokines (TRANCE), involved in the intestinal barrier functions [224]. A combination of probiotic mixture (Bifidobacterium bifidum, Lactobacillus acidophilus, Lactobacillus fermentum, and Lactobacillus reuteri) and vitamin D have been administered in SCZ patients, showing amelioration of PANSS scores with reduced inflammation and enhanced plasma total antioxidant capacity [225]. Munawar et al. [226] extensively reviewed diverse nonpharmaceutical approaches in the treatment of SCZ, including probiotics and prebiotics, suggesting the use of a psychobiotic in SCZ as a promising challenge for clinical research.
Preclinical studies and human trials show that probiotics, mainly psychobiotics, can be beneficial for brain health and thereby they could be a promising alternative therapy for the treatment of NDDs. However, some aspects need to be considered and more deeply elucidated, such as the strain specificity, the probiotic dosage, time of treatment, and the precise mechanism of action at the molecular level. Moreover, there are some limitations, including individual differences (i.e., genetic background, environmental factors, diet, gender) and/or the low number of participants which remains a limit in producing high-quality clinical data [53].

7. Conclusions

Prenatal, natal, and postnatal adverse factors represent the underlying conditions for the onset of NDDs during brain development. NDDs are multifactorial disorders involving both a strong genetic component and environmental contributors. This wide and complex group of factors makes it difficult to find a trigger. The genetic and phenotypic complexity underlying NDDs are still the main obstacle to finding effective therapies. We have focused on the effects caused by an environmental factor, represented by antibiotic administration during different stages of CNS development. Antibiotic administration has been proposed as a possible therapy for ASD patients; however, this treatment can have side effects by affecting the gut microbiota homeostasis by targeting both pathogens and healthy commensal bacteria. It is now accepted that antibiotics alone or added to a genetic risk may perturb the gut–brain axis and have effects on the correct development of the brain. To date, mounting evidence from human and animal studies suggests that gut microbial targeting therapy may be beneficial as a new and safe method for treating individuals affected by NDDs. Microbiota is indeed influenced by different environmental factors before birth, during infancy, and during childhood, and can play a key role CNS development, influencing neurogenesis and microglial maturation. Alterations in the gut microbiome caused by antibiotics administration in children can lead to inappropriate neuronal maturation during critical phases of brain development. On the other hand, a particular class of probiotics, defined as “psychobiotics”, can specifically confer health benefits at the brain level. They can transiently colonize the gut, and in turn, restore the composition of a healthy gut microbiota by producing growth factors for beneficial microbes, for example during antibiotic therapy, by competing for nutrients and/or producing inhibiting molecules that protect from pathogens colonization, and by interacting with the intestinal mucosa and modulating the intestinal immune system as well as by producing and/or stimulating different types of neuroactive molecules directly involved in the two-way microbiota-brain interplay. In summary, evidence from preclinical and clinical studies provides support for the promising effect caused by probiotic administration. The future holds the exciting potential of probiotic-based therapies to prevent and cure the onset of NDDs.

Author Contributions

T.D., R.P., N.B., A.C., A.D.J. wrote the manuscript. T.D. prepared the artwork. All authors have read and agreed to the published version of the manuscript.

Funding

This paper was supported by the Autism Research Institute Grant 2021 to N.B.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The artwork was prepared using BioRender and Servier Medical Art.

Conflicts of Interest

The authors declare that there are no competing interest associated with this manuscript.

Abbreviations

5-hydroxytryptamine (5-HT); Attention Deficit Hyperactivity Disorder (ADHD); Autism Spectrum Disorder (ASD); Black Tan Brachyury (BTBR); Blood Brain Barrier (BBB); Brain-derived Neurotrophic Factor (BDNF); Central Nervous System (CNS); Chromodomain Helicase DNA Binding Protein 8 (CHD8); Enteric Nervous System (ENS); Gastrointestinal (GI); Hazard Risk Ratios (HR); Intellectual Disability (ID); Interleukin (IL-); Irritable Bowel Syndrome (IBS); Knock-out (KO); Maternal Immune Activation (MIA); Neurodevelopmental Disorders (NDDs); Neuroligin 3 (NLGN3); Positive And Negative Syndrome Scale (PANSS); Schizophrenia (SCZ); Short-Chain Fatty Acids (SCFAs).

References

  1. Boyce, W.T.; Sokolowski, M.B.; Robinson, G.E. Genes and environments, development and time. Proc. Natl. Acad. Sci. USA 2020, 117, 23235–23241. [Google Scholar] [CrossRef] [PubMed]
  2. Carlsson, T.; Molander, F.; Taylor, M.J.; Jonsson, U.; Bölte, S. Early environmental risk factors for neurodevelopmental disorders—A systematic review of twin and sibling studies. Dev. Psychopathol. 2021, 33, 1448–1495. [Google Scholar] [CrossRef] [PubMed]
  3. Niemi, M.E.K.; Martin, H.C.; Rice, D.L.; Gallone, G.; Gordon, S.; Kelemen, M.; McAloney, K.; McRae, J.; Radford, E.J.; Yu, S.; et al. Common genetic variants contribute to risk of rare severe neurodevelopmental disorders. Nature 2018, 562, 268–271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Parenti, I.; Rabaneda, L.G.; Schoen, H.; Novarino, G. Neurodevelopmental disorders: From genetics to functional pathways. Trends Neurosci. 2020, 43, 608–621. [Google Scholar] [CrossRef]
  5. Francés, L.; Quintero, J.; Fernández, A.; Ruiz, A.; Caules, J.; Fillon, G.; Hervás, A.; Soler, C.V. Current state of knowledge on the prevalence of neurodevelopmental disorders in childhood according to the DSM-5: A systematic review in accordance with the PRISMA criteria. Child Adolesc. Psychiatry Ment. Health 2022, 16, 27. [Google Scholar] [CrossRef]
  6. Antshel, K.M.; Barkley, R. Attention deficit hyperactivity disorder. Handb. Clin. Neurol. 2020, 174, 37–45. [Google Scholar] [CrossRef]
  7. Baio, J.; Wiggins, L.; Christensen, D.L.; Maenner, M.J.; Daniels, J.; Warren, Z.; Kurzius-Spencer, M.; Zahorodny, W.; Robinson Rosenberg, C.; White, T.; et al. Prevalence of Autism Spectrum Disorder Among Children Aged 8 Years—Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2014. MMWR Surveill. Summ. 2018, 67, 1–23. [Google Scholar] [CrossRef]
  8. Davidovitch, M.; Shmueli, D.; Rotem, R.S.; Bloch, A.M. Diagnosis despite clinical ambiguity: Physicians’ perspectives on the rise in Autism Spectrum disorder incidence. BMC Psychiatry 2021, 21, 150. [Google Scholar] [CrossRef]
  9. DuPaul, G.J.; Gormley, M.J.; Laracy, S.D. Comorbidity of LD and ADHD: Implications of DSM-5 for assessment and treatment. J. Learn. Disabil. 2013, 46, 43–51. [Google Scholar] [CrossRef]
  10. Hertz-Picciotto, I.; Schmidt, R.J.; Walker, C.K.; Bennett, D.H.; Oliver, M.; Shedd-Wise, K.M.; LaSalle, J.M.; Giulivi, C.; Puschner, B.; Thomas, J.; et al. A Prospective Study of Environmental Exposures and Early Biomarkers in Autism Spectrum Disorder: Design, Protocols, and Preliminary Data from the MARBLES Study. Environ. Health Perspect. 2018, 126, 117004. [Google Scholar] [CrossRef]
  11. Cardoso, A.R.; Lopes-Marques, M.; Silva, R.M.; Serrano, C.; Amorim, A.; Prata, M.J.; Azevedo, L. Essential genetic findings in neurodevelopmental disorders. Hum. Genom. 2019, 13, 31. [Google Scholar] [CrossRef]
  12. Satterstrom, F.K.; Kosmicki, J.A.; Wang, J.; Breen, M.S.; De Rubeis, S.; An, J.-Y.; Peng, M.; Collins, R.; Grove, J.; Klei, L.; et al. Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism. Cell 2020, 180, 568–584.e23. [Google Scholar] [CrossRef]
  13. Gandal, M.J.; Zhang, P.; Hadjimichael, E.; Walker, R.L.; Chen, C.; Liu, S.; Won, H.; van Bakel, H.; Varghese, M.; Wang, Y.; et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science 2018, 362. [Google Scholar] [CrossRef] [Green Version]
  14. An, J.-Y.; Lin, K.; Zhu, L.; Werling, D.M.; Dong, S.; Brand, H.; Wang, H.Z.; Zhao, X.; Schwartz, G.B.; Collins, R.L.; et al. Genome-wide de novo risk score implicates promoter variation in autism spectrum disorder. Science 2018, 362. [Google Scholar] [CrossRef] [Green Version]
  15. Sanders, S.J.; He, X.; Willsey, A.J.; Ercan-Sencicek, A.G.; Samocha, K.E.; Cicek, A.E.; Murtha, M.T.; Bal, V.H.; Bishop, S.L.; Dong, S.; et al. Insights into Autism Spectrum Disorder Genomic Architecture and Biology from 71 Risk Loci. Neuron 2015, 87, 1215–1233. [Google Scholar] [CrossRef] [Green Version]
  16. de la Torre-Ubieta, L.; Won, H.; Stein, J.L.; Geschwind, D.H. Advancing the understanding of autism disease mechanisms through genetics. Nat. Med. 2016, 22, 345–361. [Google Scholar] [CrossRef] [Green Version]
  17. Rasheed, M.; Khan, V.; Harripaul, R.; Siddiqui, M.; Malik, M.A.; Ullah, Z.; Zahid, M.; Vincent, J.B.; Ansar, M. Exome sequencing identifies novel and known mutations in families with intellectual disability. BMC Med. Genom. 2021, 14, 211. [Google Scholar] [CrossRef]
  18. Hamanaka, K.; Miyake, N.; Mizuguchi, T.; Miyatake, S.; Uchiyama, Y.; Tsuchida, N.; Sekiguchi, F.; Mitsuhashi, S.; Tsurusaki, Y.; Nakashima, M.; et al. Large-scale discovery of novel neurodevelopmental disorder-related genes through a unified analysis of single-nucleotide and copy number variants. Genome Med. 2022, 14, 40. [Google Scholar] [CrossRef]
  19. Leblond, C.S.; Cliquet, F.; Carton, C.; Huguet, G.; Mathieu, A.; Kergrohen, T.; Buratti, J.; Lemière, N.; Cuisset, L.; Bienvenu, T.; et al. Both rare and common genetic variants contribute to autism in the Faroe Islands. NPJ Genom. Med. 2019, 4, 1. [Google Scholar] [CrossRef] [Green Version]
  20. Chaste, P.; Leboyer, M. Autism risk factors: Genes, environment, and gene-environment interactions. Dialogues Clin. Neurosci. 2012, 14, 281–292. [Google Scholar] [CrossRef]
  21. Rahnama, M.; Tehrani, H.A.; Mirzaie, M.; Ziaee, V. Identification of key genes and convergent pathways disrupted in autism spectrum disorder via comprehensive bioinformatic analysis. Inform. Med. Unlocked 2021, 24, 100589. [Google Scholar] [CrossRef]
  22. Bonsi, P.; De Jaco, A.; Fasano, L.; Gubellini, P. Postsynaptic autism spectrum disorder genes and synaptic dysfunction. Neurobiol. Dis. 2022, 162, 105564. [Google Scholar] [CrossRef]
  23. Sacai, H.; Sakoori, K.; Konno, K.; Nagahama, K.; Suzuki, H.; Watanabe, T.; Watanabe, M.; Uesaka, N.; Kano, M. Autism spectrum disorder-like behavior caused by reduced excitatory synaptic transmission in pyramidal neurons of mouse prefrontal cortex. Nat. Commun. 2020, 11, 5140. [Google Scholar] [CrossRef] [PubMed]
  24. Trobiani, L.; Meringolo, M.; Diamanti, T.; Bourne, Y.; Marchot, P.; Martella, G.; Dini, L.; Pisani, A.; De Jaco, A.; Bonsi, P. The neuroligins and the synaptic pathway in Autism Spectrum Disorder. Neurosci. Biobehav. Rev. 2020, 119, 37–51. [Google Scholar] [CrossRef] [PubMed]
  25. Patel, S.; Masi, A.; Dale, R.C.; Whitehouse, A.J.O.; Pokorski, I.; Alvares, G.A.; Hickie, I.B.; Breen, E.; Guastella, A.J. Social impairments in autism spectrum disorder are related to maternal immune history profile. Mol. Psychiatry 2018, 23, 1794–1797. [Google Scholar] [CrossRef] [Green Version]
  26. Vyas, Y.; Cheyne, J.E.; Lee, K.; Jung, Y.; Cheung, P.Y.; Montgomery, J.M. Shankopathies in the developing brain in autism spectrum disorders. Front. Neurosci. 2021, 15, 775431. [Google Scholar] [CrossRef]
  27. Pohl, T.T.; Hörnberg, H. Neuroligins in neurodevelopmental conditions: How mouse models of de novo mutations can help us link synaptic function to social behavior. Neuronal Signal. 2022, 6, NS20210030. [Google Scholar] [CrossRef]
  28. Scattolin, M.A.d.A.; Resegue, R.M.; do Rosário, M.C. The impact of the environment on neurodevelopmental disorders in early childhood. J. Pediatr. 2022, 98 (Suppl. 1), S66–S72. [Google Scholar] [CrossRef]
  29. De Felice, A.; Ricceri, L.; Venerosi, A.; Chiarotti, F.; Calamandrei, G. Multifactorial origin of neurodevelopmental disorders: Approaches to understanding complex etiologies. Toxics 2015, 3, 89–129. [Google Scholar] [CrossRef]
  30. Mazina, V.; Gerdts, J.; Trinh, S.; Ankenman, K.; Ward, T.; Dennis, M.Y.; Girirajan, S.; Eichler, E.E.; Bernier, R. Epigenetics of autism-related impairment: Copy number variation and maternal infection. J. Dev. Behav. Pediatr. 2015, 36, 61–67. [Google Scholar] [CrossRef]
  31. Schaafsma, S.M.; Gagnidze, K.; Reyes, A.; Norstedt, N.; Månsson, K.; Francis, K.; Pfaff, D.W. Sex-specific gene-environment interactions underlying ASD-like behaviors. Proc. Natl. Acad. Sci. USA 2017, 114, 1383–1388. [Google Scholar] [CrossRef] [Green Version]
  32. Wang, C.; Geng, H.; Liu, W.; Zhang, G. Prenatal, perinatal, and postnatal factors associated with autism: A meta-analysis. Medicine 2017, 96, e6696. [Google Scholar] [CrossRef]
  33. Cheroni, C.; Caporale, N.; Testa, G. Autism spectrum disorder at the crossroad between genes and environment: Contributions, convergences, and interactions in ASD developmental pathophysiology. Mol. Autism 2020, 11, 69. [Google Scholar] [CrossRef]
  34. Pham, C.; Symeonides, C.; O’Hely, M.; Sly, P.D.; Knibbs, L.D.; Thomson, S.; Vuillermin, P.; Saffery, R.; Ponsonby, A.-L.; Barwon Infant Study Investigator Group. Early life environmental factors associated with autism spectrum disorder symptoms in children at age 2 years: A birth cohort study. Autism 2022, 26, 1864–1881. [Google Scholar] [CrossRef]
  35. Khogeer, A.A.; AboMansour, I.S.; Mohammed, D.A. The role of genetics, epigenetics, and the environment in ASD: A mini review. Epigenomes 2022, 6, 15. [Google Scholar] [CrossRef]
  36. Aversa, Z.; Atkinson, E.J.; Schafer, M.J.; Theiler, R.N.; Rocca, W.A.; Blaser, M.J.; LeBrasseur, N.K. Association of infant antibiotic exposure with childhood health outcomes. Mayo Clin. Proc. 2021, 96, 66–77. [Google Scholar] [CrossRef]
  37. Manco, M. Gut microbiota and developmental programming of the brain: From evidence in behavioral endophenotypes to novel perspective in obesity. Front. Cell. Infect. Microbiol. 2012, 2, 109. [Google Scholar] [CrossRef] [Green Version]
  38. Patrick, D.M.; Sbihi, H.; Dai, D.L.Y.; Al Mamun, A.; Rasali, D.; Rose, C.; Marra, F.; Boutin, R.C.T.; Petersen, C.; Stiemsma, L.T.; et al. Decreasing antibiotic use, the gut microbiota, and asthma incidence in children: Evidence from population-based and prospective cohort studies. Lancet Respir. Med. 2020, 8, 1094–1105. [Google Scholar] [CrossRef]
  39. Trasande, L.; Blustein, J.; Liu, M.; Corwin, E.; Cox, L.M.; Blaser, M.J. Infant antibiotic exposures and early-life body mass. Int. J. Obes. 2013, 37, 16–23. [Google Scholar] [CrossRef] [Green Version]
  40. Fan, H.; Gilbert, R.; O’Callaghan, F.; Li, L. Associations between macrolide antibiotics prescribing during pregnancy and adverse child outcomes in the UK: Population based cohort study. BMJ 2020, 368, m331. [Google Scholar] [CrossRef]
  41. Muanda, F.T.; Sheehy, O.; Bérard, A. Use of antibiotics during pregnancy and risk of spontaneous abortion. CMAJ 2017, 189, E625–E633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as major disruptors of gut microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef] [PubMed]
  43. Cox, L.M.; Weiner, H.L. Microbiota Signaling Pathways that Influence Neurologic Disease. Neurotherapeutics 2018, 15, 135–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cox, L.M.; Yamanishi, S.; Sohn, J.; Alekseyenko, A.V.; Leung, J.M.; Cho, I.; Kim, S.G.; Li, H.; Gao, Z.; Mahana, D.; et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014, 158, 705–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Margolis, K.G.; Cryan, J.F.; Mayer, E.A. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology 2021, 160, 1486–1501. [Google Scholar] [CrossRef]
  46. Cryan, J.F.; Dinan, T.G. Mind-altering microorganisms: The impact of the gut microbiota on brain and behaviour. Nat. Rev. Neurosci. 2012, 13, 701–712. [Google Scholar] [CrossRef]
  47. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.M.; Sandhu, K.V.; Bastiaanssen, T.F.S.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The Microbiota-Gut-Brain Axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef]
  48. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
  49. Ahmed, H.; Leyrolle, Q.; Koistinen, V.; Kärkkäinen, O.; Layé, S.; Delzenne, N.; Hanhineva, K. Microbiota-derived metabolites as drivers of gut-brain communication. Gut Microbes 2022, 14, 2102878. [Google Scholar] [CrossRef]
  50. Huang, F.; Wu, X. Brain neurotransmitter modulation by gut microbiota in anxiety and depression. Front. Cell Dev. Biol. 2021, 9, 649103. [Google Scholar] [CrossRef]
  51. Rastelli, M.; Cani, P.D.; Knauf, C. The gut microbiome influences host endocrine functions. Endocr. Rev. 2019, 40, 1271–1284. [Google Scholar] [CrossRef]
  52. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef] [Green Version]
  53. Lee, J.E.; Walton, D.; O’Connor, C.P.; Wammes, M.; Burton, J.P.; Osuch, E.A. Drugs, guts, brains, but not rock and roll: The need to consider the role of gut microbiota in contemporary mental health and wellness of emerging adults. Int. J. Mol. Sci. 2022, 23, 6643. [Google Scholar] [CrossRef]
  54. 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. [Google Scholar] [CrossRef]
  55. Larraufie, P.; Martin-Gallausiaux, C.; Lapaque, N.; Dore, J.; Gribble, F.M.; Reimann, F.; Blottiere, H.M. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep. 2018, 8, 74. [Google Scholar] [CrossRef] [Green Version]
  56. Abdalkareem Jasim, S.; Jade Catalan Opulencia, M.; Alexis Ramírez-Coronel, A.; Kamal Abdelbasset, W.; Hasan Abed, M.; Markov, A.; Raheem Lateef Al-Awsi, G.; Azamatovich Shamsiev, J.; Thaeer Hammid, A.; Nader Shalaby, M.; et al. The emerging role of microbiota-derived short-chain fatty acids in immunometabolism. Int. Immunopharmacol. 2022, 110, 108983. [Google Scholar] [CrossRef]
  57. Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [Green Version]
  58. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  59. Fung, T.C. The microbiota-immune axis as a central mediator of gut-brain communication. Neurobiol. Dis. 2020, 136, 104714. [Google Scholar] [CrossRef]
  60. Cenit, M.C.; Sanz, Y.; Codoñer-Franch, P. Influence of gut microbiota on neuropsychiatric disorders. World J. Gastroenterol. 2017, 23, 5486–5498. [Google Scholar] [CrossRef] [Green Version]
  61. Welcome, M.O. Gut Microbiota Disorder, Gut Epithelial and Blood-Brain Barrier Dysfunctions in Etiopathogenesis of Dementia: Molecular Mechanisms and Signaling Pathways. Neuromolecular Med. 2019, 21, 205–226. [Google Scholar] [CrossRef] [PubMed]
  62. Omenetti, S.; Bussi, C.; Metidji, A.; Iseppon, A.; Lee, S.; Tolaini, M.; Li, Y.; Kelly, G.; Chakravarty, P.; Shoaie, S.; et al. The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells. Immunity 2019, 51, 77–89.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Gao, K.; Mu, C.-L.; Farzi, A.; Zhu, W.-Y. Tryptophan metabolism: A link between the gut microbiota and brain. Adv. Nutr. 2020, 11, 709–723. [Google Scholar] [CrossRef] [PubMed]
  64. Gheorghe, C.E.; Martin, J.A.; Manriquez, F.V.; Dinan, T.G.; Cryan, J.F.; Clarke, G. Focus on the essentials: Tryptophan metabolism and the microbiome-gut-brain axis. Curr. Opin. Pharmacol. 2019, 48, 137–145. [Google Scholar] [CrossRef] [PubMed]
  65. Sharma, R.; Gupta, D.; Mehrotra, R.; Mago, P. Psychobiotics: The Next-Generation Probiotics for the Brain. Curr. Microbiol. 2021, 78, 449–463. [Google Scholar] [CrossRef]
  66. Foster, J.A.; McVey Neufeld, K.-A. Gut-brain axis: How the microbiome influences anxiety and depression. Trends Neurosci. 2013, 36, 305–312. [Google Scholar] [CrossRef]
  67. Israelyan, N.; Margolis, K.G. Serotonin as a link between the gut-brain-microbiome axis in autism spectrum disorders. Pharmacol. Res. 2018, 132, 1–6. [Google Scholar] [CrossRef]
  68. Auteri, M.; Zizzo, M.G.; Serio, R. GABA and GABA receptors in the gastrointestinal tract: From motility to inflammation. Pharmacol. Res. 2015, 93, 11–21. [Google Scholar] [CrossRef]
  69. Kim, J.A.; Park, M.S.; Kang, S.A.; Ji, G.E. Production of γ-aminobutyric acid during fermentation of Gastrodia elata Bl. by co-culture of Lactobacillus brevis GABA 100 with Bifidobacterium bifidum BGN4. Food Sci. Biotechnol. 2014, 23, 459–466. [Google Scholar] [CrossRef]
  70. Diez-Gutiérrez, L.; San Vicente, L.; Barrón, L.J.R.; Villarán, M.d.C.; Chávarri, M. Gamma-aminobutyric acid and probiotics: Multiple health benefits and their future in the global functional food and nutraceuticals market. J. Funct. Foods 2020, 64, 103669. [Google Scholar] [CrossRef]
  71. Mele, M.; Costa, R.O.; Duarte, C.B. Alterations in GABAA-Receptor Trafficking and Synaptic Dysfunction in Brain Disorders. Front. Cell. Neurosci. 2019, 13, 77. [Google Scholar] [CrossRef]
  72. Golofast, B.; Vales, K. The connection between microbiome and schizophrenia. Neurosci. Biobehav. Rev. 2020, 108, 712–731. [Google Scholar] [CrossRef]
  73. Dash, S.; Syed, Y.A.; Khan, M.R. Understanding the role of the gut microbiome in brain development and its association with neurodevelopmental psychiatric disorders. Front. Cell Dev. Biol. 2022, 10, 880544. [Google Scholar] [CrossRef]
  74. Collins, S.M.; Surette, M.; Bercik, P. The interplay between the intestinal microbiota and the brain. Nat. Rev. Microbiol. 2012, 10, 735–742. [Google Scholar] [CrossRef]
  75. Góralczyk-Bińkowska, A.; Szmajda-Krygier, D.; Kozłowska, E. The Microbiota-Gut-Brain Axis in Psychiatric Disorders. Int. J. Mol. Sci. 2022, 23, 11245. [Google Scholar] [CrossRef]
  76. Ferguson, B.J.; Dovgan, K.; Takahashi, N.; Beversdorf, D.Q. The relationship among gastrointestinal symptoms, problem behaviors, and internalizing symptoms in children and adolescents with autism spectrum disorder. Front. Psychiatry 2019, 10, 194. [Google Scholar] [CrossRef] [Green Version]
  77. 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 2019, 56, 11. [Google Scholar] [CrossRef] [Green Version]
  78. Kim, A.; Zisman, C.R.; Holingue, C. Influences of the immune system and microbiome on the etiology of ASD and GI symptomology of autistic individuals. In Current Topics in Behavioral Neurosciences; Springer: Berlin, Heidelberg, 2022. [Google Scholar] [CrossRef]
  79. Kelly, J.R.; Kennedy, P.J.; Cryan, J.F.; Dinan, T.G.; Clarke, G.; Hyland, N.P. Breaking down the barriers: The gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front. Cell. Neurosci. 2015, 9, 392. [Google Scholar] [CrossRef] [Green Version]
  80. 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, 40–45. [Google Scholar] [CrossRef]
  81. Vuong, H.E.; Hsiao, E.Y. Emerging roles for the gut microbiome in autism spectrum disorder. Biol. Psychiatry 2017, 81, 411–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Aarts, E.; Ederveen, T.H.A.; Naaijen, J.; Zwiers, M.P.; Boekhorst, J.; Timmerman, H.M.; Smeekens, S.P.; Netea, M.G.; Buitelaar, J.K.; Franke, B.; et al. Gut microbiome in ADHD and its relation to neural reward anticipation. PLoS ONE 2017, 12, e0183509. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Checa-Ros, A.; Jeréz-Calero, A.; Molina-Carballo, A.; Campoy, C.; Muñoz-Hoyos, A. Current evidence on the role of the gut microbiome in ADHD pathophysiology and therapeutic implications. Nutrients 2021, 13, 249. [Google Scholar] [CrossRef] [PubMed]
  85. Richarte, V.; Sánchez-Mora, C.; Corrales, M.; Fadeuilhe, C.; Vilar-Ribó, L.; Arribas, L.; Garcia, E.; Rosales-Ortiz, S.K.; Arias-Vasquez, A.; Soler-Artigas, M.; et al. Gut microbiota signature in treatment-naïve attention-deficit/hyperactivity disorder. Transl. Psychiatry 2021, 11, 382. [Google Scholar] [CrossRef] [PubMed]
  86. Lacorte, E.; Gervasi, G.; Bacigalupo, I.; Vanacore, N.; Raucci, U.; Parisi, P. A systematic review of the microbiome in children with neurodevelopmental disorders. Front. Neurol. 2019, 10, 727. [Google Scholar] [CrossRef]
  87. Pärtty, A.; Kalliomäki, M.; Wacklin, P.; Salminen, S.; Isolauri, E. A possible link between early probiotic intervention and the risk of neuropsychiatric disorders later in childhood: A randomized trial. Pediatr. Res. 2015, 77, 823–828. [Google Scholar] [CrossRef] [Green Version]
  88. Clapp, M.; Aurora, N.; Herrera, L.; Bhatia, M.; Wilen, E.; Wakefield, S. Gut microbiota’s effect on mental health: The gut-brain axis. Clin. Pract. 2017, 7, 987. [Google Scholar] [CrossRef]
  89. Yeh, T.-C.; Bai, Y.-M.; Tsai, S.-J.; Chen, T.-J.; Liang, C.-S.; Chen, M.-H. Risks of Major Mental Disorders and Irritable Bowel Syndrome among the Offspring of Parents with Irritable Bowel Syndrome: A Nationwide Study. Int. J. Environ. Res. Public Health 2021, 18, 4679. [Google Scholar] [CrossRef]
  90. Shah, E.; Rezaie, A.; Riddle, M.; Pimentel, M. Psychological disorders in gastrointestinal disease: Epiphenomenon, cause or consequence? Ann. Gastroenterol. 2014, 27, 224–230. [Google Scholar]
  91. Klein-Petersen, A.W.; Köhler-Forsberg, O.; Benros, M.E. Infections, antibiotic treatment and the Microbiome in relation to schizophrenia. Schizophr. Res. 2021, 234, 71–77. [Google Scholar] [CrossRef]
  92. Severance, E.G.; Yolken, R.H.; Eaton, W.W. Autoimmune diseases, gastrointestinal disorders and the microbiome in schizophrenia: More than a gut feeling. Schizophr. Res. 2016, 176, 23–35. [Google Scholar] [CrossRef] [Green Version]
  93. Patrono, E.; Svoboda, J.; Stuchlík, A. Schizophrenia, the gut microbiota, and new opportunities from optogenetic manipulations of the gut-brain axis. Behav. Brain Funct. 2021, 17, 7. [Google Scholar] [CrossRef]
  94. Socała, K.; Doboszewska, U.; Szopa, A.; Serefko, A.; Włodarczyk, M.; Zielińska, A.; Poleszak, E.; Fichna, J.; Wlaź, P. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol. Res. 2021, 172, 105840. [Google Scholar] [CrossRef]
  95. Zhu, F.; Ju, Y.; Wang, W.; Wang, Q.; Guo, R.; Ma, Q.; Sun, Q.; Fan, Y.; Xie, Y.; Yang, Z.; et al. Metagenome-wide association of gut microbiome features for schizophrenia. Nat. Commun. 2020, 11, 1612. [Google Scholar] [CrossRef] [Green Version]
  96. Castro-Nallar, E.; Bendall, M.L.; Pérez-Losada, M.; Sabuncyan, S.; Severance, E.G.; Dickerson, F.B.; Schroeder, J.R.; Yolken, R.H.; Crandall, K.A. Composition, taxonomy and functional diversity of the oropharynx microbiome in individuals with schizophrenia and controls. PeerJ 2015, 3, e1140. [Google Scholar] [CrossRef]
  97. Bojović, K.; Ignjatović, Ð.-D.I.; Soković Bajić, S.; Vojnović Milutinović, D.; Tomić, M.; Golić, N.; Tolinački, M. Gut microbiota dysbiosis associated with altered production of short chain fatty acids in children with neurodevelopmental disorders. Front. Cell. Infect. Microbiol. 2020, 10, 223. [Google Scholar] [CrossRef]
  98. Jolanta Wasilewska, J.; Klukowski, M. Gastrointestinal symptoms and autism spectrum disorder: Links and risks—A possible new overlap syndrome. PHMT 2015, 2015, 153–166. [Google Scholar] [CrossRef] [Green Version]
  99. Zhang, D.-M.; Ye, J.-X.; Mu, J.-S.; Cui, X.-P. Efficacy of Vitamin B Supplementation on Cognition in Elderly Patients With Cognitive-Related Diseases. J. Geriatr. Psychiatry Neurol. 2017, 30, 50–59. [Google Scholar] [CrossRef]
  100. Sarubbo, F.; Cavallucci, V.; Pani, G. The influence of gut microbiota on neurogenesis: Evidence and hopes. Cells 2022, 11, 382. [Google Scholar] [CrossRef]
  101. Jiang, H.; Ling, Z.; Zhang, Y.; Mao, H.; Ma, Z.; Yin, Y.; Wang, W.; Tang, W.; Tan, Z.; Shi, J.; et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 2015, 48, 186–194. [Google Scholar] [CrossRef] [PubMed]
  102. Sgritta, M.; Dooling, S.W.; Buffington, S.A.; Momin, E.N.; Francis, M.B.; Britton, R.A.; Costa-Mattioli, M. Mechanisms Underlying Microbial-Mediated Changes in Social Behavior in Mouse Models of Autism Spectrum Disorder. Neuron 2019, 101, 246–259.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Grenham, S.; Clarke, G.; Cryan, J.F.; Dinan, T.G. Brain-gut-microbe communication in health and disease. Front. Physiol. 2011, 2, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Sharon, G.; Cruz, N.J.; Kang, D.-W.; Gandal, M.J.; Wang, B.; Kim, Y.-M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; Lane, C.J.; et al. Human Gut Microbiota from Autism Spectrum Disorder Promote Behavioral Symptoms in Mice. Cell 2019, 177, 1600–1618.e17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Alharthi, A.; Alhazmi, S.; Alburae, N.; Bahieldin, A. The human gut microbiome as a potential factor in autism spectrum disorder. Int. J. Mol. Sci. 2022, 23, 1363. [Google Scholar] [CrossRef]
  106. Bölte, S.; Girdler, S.; Marschik, P.B. The contribution of environmental exposure to the etiology of autism spectrum disorder. Cell. Mol. Life Sci. 2019, 76, 1275–1297. [Google Scholar] [CrossRef] [Green Version]
  107. Nithianantharajah, J.; Balasuriya, G.K.; Franks, A.E.; Hill-Yardin, E.L. Using Animal Models to Study the Role of the Gut-Brain Axis in Autism. Curr. Dev. Disord. Rep. 2017, 4, 28–36. [Google Scholar] [CrossRef] [Green Version]
  108. Jamain, S.; Quach, H.; Betancur, C.; Råstam, M.; Colineaux, C.; Gillberg, I.C.; Soderstrom, H.; Giros, B.; Leboyer, M.; Gillberg, C.; et al. Mutations of the X-linked genes encoding neuroligins NLGN3 and NLGN4 are associated with autism. Nat. Genet. 2003, 34, 27–29. [Google Scholar] [CrossRef] [Green Version]
  109. Hosie, S.; Ellis, M.; Swaminathan, M.; Ramalhosa, F.; Seger, G.O.; Balasuriya, G.K.; Gillberg, C.; Råstam, M.; Churilov, L.; McKeown, S.J.; et al. Gastrointestinal dysfunction in patients and mice expressing the autism-associated R451C mutation in neuroligin-3. Autism Res. 2019, 12, 1043–1056. [Google Scholar] [CrossRef] [Green Version]
  110. Herath, M.; Bornstein, J.C.; Hill-Yardin, E.L.; Franks, A.E. The autism-associated Neuroligin-3 R451C mutation alters mucus density and the spatial distribution of bacteria in the mouse gastrointestinal tract. BioRxiv 2022. [Google Scholar] [CrossRef]
  111. Leembruggen, A.J.L.; Balasuriya, G.K.; Zhang, J.; Schokman, S.; Swiderski, K.; Bornstein, J.C.; Nithianantharajah, J.; Hill-Yardin, E.L. Colonic dilation and altered ex vivo gastrointestinal motility in the neuroligin-3 knockout mouse. Autism Res. 2020, 13, 691–701. [Google Scholar] [CrossRef]
  112. Sauer, A.K.; Bockmann, J.; Steinestel, K.; Boeckers, T.M.; Grabrucker, A.M. Altered intestinal morphology and microbiota composition in the autism spectrum disorders associated SHANK3 mouse model. Int. J. Mol. Sci. 2019, 20, 2134. [Google Scholar] [CrossRef] [Green Version]
  113. Tabouy, L.; Getselter, D.; Ziv, O.; Karpuj, M.; Tabouy, T.; Lukic, I.; Maayouf, R.; Werbner, N.; Ben-Amram, H.; Nuriel-Ohayon, M.; et al. Dysbiosis of microbiome and probiotic treatment in a genetic model of autism spectrum disorders. Brain Behav. Immun. 2018, 73, 310–319. [Google Scholar] [CrossRef]
  114. Alotaibi, M.; Ramzan, K. A de novo variant of CHD8 in a patient with autism spectrum disorder. Discoveries 2020, 8, e107. [Google Scholar] [CrossRef]
  115. Bernier, R.; Golzio, C.; Xiong, B.; Stessman, H.A.; Coe, B.P.; Penn, O.; Witherspoon, K.; Gerdts, J.; Baker, C.; Vulto-van Silfhout, A.T.; et al. Disruptive CHD8 mutations define a subtype of autism early in development. Cell 2014, 158, 263–276. [Google Scholar] [CrossRef] [Green Version]
  116. Yu, Y.; Zhang, B.; Ji, P.; Zuo, Z.; Huang, Y.; Wang, N.; Liu, C.; Liu, S.-J.; Zhao, F. Changes to gut amino acid transporters and microbiome associated with increased E/I ratio in Chd8+/- mouse model of ASD-like behavior. Nat. Commun. 2022, 13, 1151. [Google Scholar] [CrossRef]
  117. Katayama, Y.; Nishiyama, M.; Shoji, H.; Ohkawa, Y.; Kawamura, A.; Sato, T.; Suyama, M.; Takumi, T.; Miyakawa, T.; Nakayama, K.I. CHD8 haploinsufficiency results in autistic-like phenotypes in mice. Nature 2016, 537, 675–679. [Google Scholar] [CrossRef]
  118. Chatterjee, I.; Getselter, D.; Ghaneem, N.; Bel, S.; Elliott, E. CHD8 regulates gut epithelial cell function and affects autism-related behaviours through the gut-brain axis. BioRxiv 2021. [Google Scholar] [CrossRef]
  119. Gaudissard, J.; Ginger, M.; Premoli, M.; Memo, M.; Frick, A.; Pietropaolo, S. Behavioral abnormalities in the Fmr1-KO2 mouse model of fragile X syndrome: The relevance of early life phases. Autism Res. 2017, 10, 1584–1596. [Google Scholar] [CrossRef]
  120. Altimiras, F.; Garcia, J.A.; Palacios-García, I.; Hurley, M.J.; Deacon, R.; González, B.; Cogram, P. Altered gut microbiota in a fragile X syndrome mouse model. Front. Neurosci. 2021, 15, 653120. [Google Scholar] [CrossRef]
  121. Meyza, K.Z.; Blanchard, D.C. The BTBR mouse model of idiopathic autism—Current view on mechanisms. Neurosci. Biobehav. Rev. 2017, 76, 99–110. [Google Scholar] [CrossRef] [PubMed]
  122. Coretti, L.; Cristiano, C.; Florio, E.; Scala, G.; Lama, A.; Keller, S.; Cuomo, M.; Russo, R.; Pero, R.; Paciello, O.; et al. Sex-related alterations of gut microbiota composition in the BTBR mouse model of autism spectrum disorder. Sci. Rep. 2017, 7, 45356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Nyangahu, D.D.; Lennard, K.S.; Brown, B.P.; Darby, M.G.; Wendoh, J.M.; Havyarimana, E.; Smith, P.; Butcher, J.; Stintzi, A.; Mulder, N.; et al. Disruption of maternal gut microbiota during gestation alters offspring microbiota and immunity. Microbiome 2018, 6, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Di Gesù, C.M.; Matz, L.M.; Buffington, S.A. Diet-induced dysbiosis of the maternal gut microbiome in early life programming of neurodevelopmental disorders. Neurosci. Res. 2021, 168, 3–19. [Google Scholar] [CrossRef] [PubMed]
  125. Wong, G.C.; Montgomery, J.M.; Taylor, M.W. The Gut-Microbiota-Brain Axis in Autism Spectrum Disorder. In Autism Spectrum Disorders; Grabrucker, A.M., Ed.; Exon Publications: Brisbane, Australia, 2021; ISBN 9780645001785. [Google Scholar]
  126. Lammert, C.R.; Lukens, J.R. Modeling Autism-Related Disorders in Mice with Maternal Immune Activation (MIA). Methods Mol. Biol. 2019, 1960, 227–236. [Google Scholar] [CrossRef] [PubMed]
  127. Kim, S.; Kim, H.; Yim, Y.S.; Ha, S.; Atarashi, K.; Tan, T.G.; Longman, R.S.; Honda, K.; Littman, D.R.; Choi, G.B.; et al. Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549, 528–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Hsiao, E.Y.; McBride, S.W.; Hsien, S.; Sharon, G.; Hyde, E.R.; McCue, T.; Codelli, J.A.; Chow, J.; Reisman, S.E.; Petrosino, J.F.; et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013, 155, 1451–1463. [Google Scholar] [CrossRef] [Green Version]
  129. Li, W.; Chen, M.; Feng, X.; Song, M.; Shao, M.; Yang, Y.; Zhang, L.; Liu, Q.; Lv, L.; Su, X. Maternal immune activation alters adult behavior, intestinal integrity, gut microbiota and the gut inflammation. Brain Behav. 2021, 11, e02133. [Google Scholar] [CrossRef]
  130. Hermansson, H.; Kumar, H.; Collado, M.C.; Salminen, S.; Isolauri, E.; Rautava, S. Breast milk microbiota is shaped by mode of delivery and intrapartum antibiotic exposure. Front. Nutr. 2019, 6, 4. [Google Scholar] [CrossRef] [Green Version]
  131. Daliry, A.; Pereira, E.N.G.d.S. Role of Maternal Microbiota and Nutrition in Early-Life Neurodevelopmental Disorders. Nutrients 2021, 13, 3533. [Google Scholar] [CrossRef]
  132. Tau, G.Z.; Peterson, B.S. Normal development of brain circuits. Neuropsychopharmacology 2010, 35, 147–168. [Google Scholar] [CrossRef]
  133. Doi, M.; Usui, N.; Shimada, S. Prenatal environment and neurodevelopmental disorders. Front. Endocrinol. 2022, 13, 860110. [Google Scholar] [CrossRef]
  134. Stokholm, J.; Schjørring, S.; Eskildsen, C.E.; Pedersen, L.; Bischoff, A.L.; Følsgaard, N.; Carson, C.G.; Chawes, B.L.K.; Bønnelykke, K.; Mølgaard, A.; et al. Antibiotic use during pregnancy alters the commensal vaginal microbiota. Clin. Microbiol. Infect. 2014, 20, 629–635. [Google Scholar] [CrossRef] [Green Version]
  135. Persaud, R.R.; Azad, M.B.; Chari, R.S.; Sears, M.R.; Becker, A.B.; Kozyrskyj, A.L.; CHILD Study Investigators. Perinatal antibiotic exposure of neonates in Canada and associated risk factors: A population-based study. J. Matern. Fetal Neonatal Med. 2015, 28, 1190–1195. [Google Scholar] [CrossRef]
  136. Koebnick, C.; Tartof, S.Y.; Sidell, M.A.; Rozema, E.; Chung, J.; Chiu, V.Y.; Taylor, Z.W.; Xiang, A.H.; Getahun, D. Effect of In-Utero Antibiotic Exposure on Childhood Outcomes: Methods and Baseline Data of the Fetal Antibiotic EXposure (FAX) Cohort Study. JMIR Res. Protoc. 2019, 8, e12065. [Google Scholar] [CrossRef] [Green Version]
  137. Petersen, I.; Gilbert, R.; Evans, S.; Ridolfi, A.; Nazareth, I. Oral antibiotic prescribing during pregnancy in primary care: UK population-based study. J. Antimicrob. Chemother. 2010, 65, 2238–2246. [Google Scholar] [CrossRef] [Green Version]
  138. Kenyon, S.; Pike, K.; Jones, D.R.; Brocklehurst, P.; Marlow, N.; Salt, A.; Taylor, D.J. Childhood outcomes after prescription of antibiotics to pregnant women with spontaneous preterm labour: 7-year follow-up of the ORACLE II trial. Lancet 2008, 372, 1319–1327. [Google Scholar] [CrossRef] [Green Version]
  139. Andersen, J.T.; Petersen, M.; Jimenez-Solem, E.; Rasmussen, J.N.; Andersen, N.L.; Afzal, S.; Broedbaek, K.; Hjelvang, B.R.; Køber, L.; Torp-Pedersen, C.; et al. Trimethoprim Use prior to Pregnancy and the Risk of Congenital Malformation: A Register-Based Nationwide Cohort Study. Obstet. Gynecol. Int. 2013, 2013, 364526. [Google Scholar] [CrossRef] [Green Version]
  140. Hamad, A.F.; Alessi-Severini, S.; Mahmud, S.; Brownell, M.; Kuo, I.F. Prenatal antibiotic exposure and risk of attention-deficit/hyperactivity disorder: A population-based cohort study. CMAJ 2020, 192, E527–E535. [Google Scholar] [CrossRef]
  141. Wimberley, T.; Agerbo, E.; Pedersen, C.B.; Dalsgaard, S.; Horsdal, H.T.; Mortensen, P.B.; Thompson, W.K.; Köhler-Forsberg, O.; Yolken, R.H. Otitis media, antibiotics, and risk of autism spectrum disorder. Autism Res. 2018, 11, 1432–1440. [Google Scholar] [CrossRef]
  142. Atladóttir, H.Ó.; Henriksen, T.B.; Schendel, D.E.; Parner, E.T. Autism after infection, febrile episodes, and antibiotic use during pregnancy: An exploratory study. Pediatrics 2012, 130, e1447–e1454. [Google Scholar] [CrossRef] [PubMed]
  143. Lavebratt, C.; Yang, L.L.; Giacobini, M.; Forsell, Y.; Schalling, M.; Partonen, T.; Gissler, M. Early exposure to antibiotic drugs and risk for psychiatric disorders: A population-based study. Transl. Psychiatry 2019, 9, 317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tochitani, S.; Ikeno, T.; Ito, T.; Sakurai, A.; Yamauchi, T.; Matsuzaki, H. Administration of Non-Absorbable Antibiotics to Pregnant Mice to Perturb the Maternal Gut Microbiota Is Associated with Alterations in Offspring Behavior. PLoS ONE 2016, 11, e0138293. [Google Scholar] [CrossRef] [PubMed]
  145. Harshaw, C.; Kojima, S.; Wellman, C.L.; Demas, G.E.; Morrow, A.L.; Taft, D.H.; Kenkel, W.M.; Leffel, J.K.; Alberts, J.R. Maternal antibiotics disrupt microbiome, behavior, and temperature regulation in unexposed infant mice. Dev. Psychobiol. 2022, 64, e22289. [Google Scholar] [CrossRef]
  146. Degroote, S.; Hunting, D.J.; Baccarelli, A.A.; Takser, L. Maternal gut and fetal brain connection: Increased anxiety and reduced social interactions in Wistar rat offspring following peri-conceptional antibiotic exposure. Prog. Neuropsychopharmacol. Biol. Psychiatry 2016, 71, 76–82. [Google Scholar] [CrossRef]
  147. Vuong, H.E.; Pronovost, G.N.; Williams, D.W.; Coley, E.J.L.; Siegler, E.L.; Qiu, A.; Kazantsev, M.; Wilson, C.J.; Rendon, T.; Hsiao, E.Y. The maternal microbiome modulates fetal neurodevelopment in mice. Nature 2020, 586, 281–286. [Google Scholar] [CrossRef]
  148. Mateos-Aparicio, P.; Rodríguez-Moreno, A. The impact of studying brain plasticity. Front. Cell. Neurosci. 2019, 13, 66. [Google Scholar] [CrossRef] [Green Version]
  149. Smith, K.E.; Pollak, S.D. Early life stress and development: Potential mechanisms for adverse outcomes. J. Neurodev. Disord. 2020, 12, 34. [Google Scholar] [CrossRef]
  150. Sturman, D.A.; Moghaddam, B. The neurobiology of adolescence: Changes in brain architecture, functional dynamics, and behavioral tendencies. Neurosci. Biobehav. Rev. 2011, 35, 1704–1712. [Google Scholar] [CrossRef] [Green Version]
  151. Duong, Q.A.; Pittet, L.F.; Curtis, N.; Zimmermann, P. Antibiotic exposure and adverse long-term health outcomes in children: A systematic review and meta-analysis. J. Infect. 2022, 85, 213–300. [Google Scholar] [CrossRef]
  152. Rogers, G.B.; Keating, D.J.; Young, R.L.; Wong, M.L.; Licinio, J.; Wesselingh, S. From gut dysbiosis to altered brain function and mental illness: Mechanisms and pathways. Mol. Psychiatry 2016, 21, 738–748. [Google Scholar] [CrossRef]
  153. Łukasik, J.; Patro-Gołąb, B.; Horvath, A.; Baron, R.; Szajewska, H.; SAWANTI Working Group. Early life exposure to antibiotics and autism spectrum disorders: A systematic review. J. Autism Dev. Disord. 2019, 49, 3866–3876. [Google Scholar] [CrossRef] [Green Version]
  154. Neuman, H.; Forsythe, P.; Uzan, A.; Avni, O.; Koren, O. Antibiotics in early life: Dysbiosis and the damage done. FEMS Microbiol. Rev. 2018, 42, 489–499. [Google Scholar] [CrossRef] [Green Version]
  155. Volker, E.; Tessier, C.; Rodriguez, N.; Yager, J.; Kozyrskyj, A. Pathways of atopic disease and neurodevelopmental impairment: Assessing the evidence for infant antibiotics. Expert Rev. Clin. Immunol. 2022, 18, 901–922. [Google Scholar] [CrossRef]
  156. Bittker, S.S.; Bell, K.R. Acetaminophen, antibiotics, ear infection, breastfeeding, vitamin D drops, and autism: An epidemiological study. Neuropsychiatr. Dis. Treat. 2018, 14, 1399–1414. [Google Scholar] [CrossRef] [Green Version]
  157. Fallon, J. Could one of the most widely prescribed antibiotics amoxicillin/clavulanate “augmentin” be a risk factor for autism? Med. Hypotheses 2005, 64, 312–315. [Google Scholar] [CrossRef]
  158. Niehus, R.; Lord, C. Early medical history of children with autism spectrum disorders. J. Dev. Behav. Pediatr. 2006, 27, S120–S127. [Google Scholar] [CrossRef]
  159. Köhler, O.; Petersen, L.; Mors, O.; Mortensen, P.B.; Yolken, R.H.; Gasse, C.; Benros, M.E. Infections and exposure to anti-infective agents and the risk of severe mental disorders: A nationwide study. Acta Psychiatr. Scand. 2017, 135, 97–105. [Google Scholar] [CrossRef]
  160. Köhler-Forsberg, O.; Petersen, L.; Gasse, C.; Mortensen, P.B.; Dalsgaard, S.; Yolken, R.H.; Mors, O.; Benros, M.E. A nationwide study in denmark of the association between treated infections and the subsequent risk of treated mental disorders in children and adolescents. JAMA Psychiatry 2019, 76, 271–279. [Google Scholar] [CrossRef] [Green Version]
  161. Slykerman, R.F.; Thompson, J.; Waldie, K.E.; Murphy, R.; Wall, C.; Mitchell, E.A. Antibiotics in the first year of life and subsequent neurocognitive outcomes. Acta Paediatr. 2017, 106, 87–94. [Google Scholar] [CrossRef]
  162. Hamad, A.F.; Alessi-Severini, S.; Mahmud, S.M.; Brownell, M.; Kuo, I.F. Antibiotic Exposure in the First Year of Life and the Risk of Attention-Deficit/Hyperactivity Disorder: A Population-Based Cohort Study. Am. J. Epidemiol. 2019, 188, 1923–1931. [Google Scholar] [CrossRef] [PubMed]
  163. Tierney, A.L.; Nelson, C.A. Brain development and the role of experience in the early years. Zero Three 2009, 30, 9–13. [Google Scholar] [PubMed]
  164. Slykerman, R.F.; Coomarasamy, C.; Wickens, K.; Thompson, J.M.D.; Stanley, T.V.; Barthow, C.; Kang, J.; Crane, J.; Mitchell, E.A. Exposure to antibiotics in the first 24 months of life and neurocognitive outcomes at 11 years of age. Psychopharmacology 2019, 236, 1573–1582. [Google Scholar] [CrossRef] [PubMed]
  165. Slob, E.M.A.; Brew, B.K.; Vijverberg, S.J.H.; Dijs, T.; van Beijsterveldt, C.E.M.; Koppelman, G.H.; Bartels, M.; Dolan, C.V.; Larsson, H.; Lundström, S.; et al. Early-life antibiotic use and risk of attention-deficit hyperactivity disorder and autism spectrum disorder: Results of a discordant twin study. Int. J. Epidemiol. 2021, 50, 475–484. [Google Scholar] [CrossRef] [PubMed]
  166. Axelsson, P.B.; Clausen, T.D.; Petersen, A.H.; Hageman, I.; Pinborg, A.; Kessing, L.V.; Bergholt, T.; Rasmussen, S.C.; Keiding, N.; Løkkegaard, E.C.L. Investigating the effects of cesarean delivery and antibiotic use in early childhood on risk of later attention deficit hyperactivity disorder. J. Child Psychol. Psychiatry 2019, 60, 151–159. [Google Scholar] [CrossRef]
  167. Karlsson, H.; Sjöqvist, H.; Brynge, M.; Gardner, R.; Dalman, C. Childhood infections and autism spectrum disorders and/or intellectual disability: A register-based cohort study. J. Neurodev. Disord. 2022, 14, 12. [Google Scholar] [CrossRef]
  168. Nudel, R.; Wang, Y.; Appadurai, V.; Schork, A.J.; Buil, A.; Agerbo, E.; Bybjerg-Grauholm, J.; Børglum, A.D.; Daly, M.J.; Mors, O.; et al. A large-scale genomic investigation of susceptibility to infection and its association with mental disorders in the Danish population. Transl. Psychiatry 2019, 9, 283. [Google Scholar] [CrossRef] [Green Version]
  169. Gau, S.S.-F.; Chang, L.-Y.; Huang, L.-M.; Fan, T.-Y.; Wu, Y.-Y.; Lin, T.-Y. Attention-deficit/hyperactivity-related symptoms among children with enterovirus 71 infection of the central nervous system. Pediatrics 2008, 122, e452–e458. [Google Scholar] [CrossRef]
  170. Singh, G.; Prabhakar, S. The association between central nervous system (CNS) infections and epilepsy: Epidemiological approaches and microbiological and epileptological perspectives. Epilepsia 2008, 49, 2–7. [Google Scholar] [CrossRef]
  171. Koponen, H.; Rantakallio, P.; Veijola, J.; Jones, P.; Jokelainen, J.; Isohanni, M. Childhood central nervous system infections and risk for schizophrenia. Eur. Arch. Psychiatry Clin. Neurosci. 2004, 254, 9–13. [Google Scholar] [CrossRef]
  172. Sabourin, K.R.; Reynolds, A.; Schendel, D.; Rosenberg, S.; Croen, L.A.; Pinto-Martin, J.A.; Schieve, L.A.; Newschaffer, C.; Lee, L.-C.; DiGuiseppi, C. Infections in children with autism spectrum disorder: Study to Explore Early Development (SEED). Autism Res. 2019, 12, 136–146. [Google Scholar] [CrossRef]
  173. Pedersen, E.M.J.; Köhler-Forsberg, O.; Nordentoft, M.; Christensen, R.H.B.; Mortensen, P.B.; Petersen, L.; Benros, M.E. Infections of the central nervous system as a risk factor for mental disorders and cognitive impairment: A nationwide register-based study. Brain Behav. Immun. 2020, 88, 668–674. [Google Scholar] [CrossRef]
  174. Adams, D.J.; Susi, A.; Erdie-Lalena, C.R.; Gorman, G.; Hisle-Gorman, E.; Rajnik, M.; Elrod, M.; Nylund, C.M. Otitis Media and Related Complications Among Children with Autism Spectrum Disorders. J. Autism Dev. Disord. 2016, 46, 1636–1642. [Google Scholar] [CrossRef]
  175. Sadaka, Y.; Freedman, J.; Ashkenazi, S.; Vinker, S.; Golan-Cohen, A.; Green, I.; Israel, A.; Eran, A.; Merzon, E. The Effect of Antibiotic Treatment of Early Childhood Shigellosis on Long-Term Prevalence of Attention Deficit/Hyperactivity Disorder. Children 2021, 8, 880. [Google Scholar] [CrossRef]
  176. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The central nervous system and the gut microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef] [Green Version]
  177. Diaz Heijtz, R.; Wang, S.; Anuar, F.; Qian, Y.; Björkholm, B.; Samuelsson, A.; Hibberd, M.L.; Forssberg, H.; Pettersson, S. Normal gut microbiota modulates brain development and behavior. Proc. Natl. Acad. Sci. USA 2011, 108, 3047–3052. [Google Scholar] [CrossRef] [Green Version]
  178. Guida, F.; Turco, F.; Iannotta, M.; De Gregorio, D.; Palumbo, I.; Sarnelli, G.; Furiano, A.; Napolitano, F.; Boccella, S.; Luongo, L.; et al. Antibiotic-induced microbiota perturbation causes gut endocannabinoidome changes, hippocampal neuroglial reorganization and depression in mice. Brain Behav. Immun. 2018, 67, 230–245. [Google Scholar] [CrossRef]
  179. Ogawa, Y.; Miyoshi, C.; Obana, N.; Yajima, K.; Hotta-Hirashima, N.; Ikkyu, A.; Kanno, S.; Soga, T.; Fukuda, S.; Yanagisawa, M. Gut microbiota depletion by chronic antibiotic treatment alters the sleep/wake architecture and sleep EEG power spectra in mice. Sci. Rep. 2020, 10, 19554. [Google Scholar] [CrossRef]
  180. Schmidtner, A.K.; Slattery, D.A.; Gläsner, J.; Hiergeist, A.; Gryksa, K.; Malik, V.A.; Hellmann-Regen, J.; Heuser, I.; Baghai, T.C.; Gessner, A.; et al. Minocycline alters behavior, microglia and the gut microbiome in a trait-anxiety-dependent manner. Transl. Psychiatry 2019, 9, 223. [Google Scholar] [CrossRef] [Green Version]
  181. Leclercq, S.; Mian, F.M.; Stanisz, A.M.; Bindels, L.B.; Cambier, E.; Ben-Amram, H.; Koren, O.; Forsythe, P.; Bienenstock, J. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 2017, 8, 15062. [Google Scholar] [CrossRef] [Green Version]
  182. Bistoletti, M.; Caputi, V.; Baranzini, N.; Marchesi, N.; Filpa, V.; Marsilio, I.; Cerantola, S.; Terova, G.; Baj, A.; Grimaldi, A.; et al. Antibiotic treatment-induced dysbiosis differently affects BDNF and TrkB expression in the brain and in the gut of juvenile mice. PLoS ONE 2019, 14, e0212856. [Google Scholar] [CrossRef] [PubMed]
  183. Desbonnet, L.; Clarke, G.; Traplin, A.; O’Sullivan, O.; Crispie, F.; Moloney, R.D.; Cotter, P.D.; Dinan, T.G.; Cryan, J.F. Gut microbiota depletion from early adolescence in mice: Implications for brain and behaviour. Brain Behav. Immun. 2015, 48, 165–173. [Google Scholar] [CrossRef] [PubMed]
  184. Perna, J.; Lu, J.; Mullen, B.; Liu, T.; Tjia, M.; Weiser, S.; Ackman, J.; Zuo, Y. Perinatal penicillin exposure affects cortical development and sensory processing. Front. Mol. Neurosci. 2021, 14, 704219. [Google Scholar] [CrossRef] [PubMed]
  185. Helaly, A.M.N.; El-Attar, Y.A.; Khalil, M.; Ahmed Ghorab, D.S.E.-D.; El-Mansoury, A.M. Antibiotic abuse induced histopathological and neurobehavioral disorders in mice. Curr. Drug Saf. 2019, 14, 199–208. [Google Scholar] [CrossRef] [PubMed]
  186. Atli, O.; Demir-Ozkay, U.; Ilgin, S.; Aydin, T.H.; Akbulut, E.N.; Sener, E. Evidence for neurotoxicity associated with amoxicillin in juvenile rats. Hum. Exp. Toxicol. 2016, 35, 866–876. [Google Scholar] [CrossRef]
  187. Volkova, A.; Ruggles, K.; Schulfer, A.; Gao, Z.; Ginsberg, S.D.; Blaser, M.J. Effects of early-life penicillin exposure on the gut microbiome and frontal cortex and amygdala gene expression. iScience 2021, 24, 102797. [Google Scholar] [CrossRef]
  188. Liu, G.; Yu, Q.; Tan, B.; Ke, X.; Zhang, C.; Li, H.; Zhang, T.; Lu, Y. Gut dysbiosis impairs hippocampal plasticity and behaviors by remodeling serum metabolome. Gut Microbes 2022, 14, 2104089. [Google Scholar] [CrossRef]
  189. Zimmermann, P.; Curtis, N. The effect of antibiotics on the composition of the intestinal microbiota—A systematic review. J. Infect. 2019, 79, 471–489. [Google Scholar] [CrossRef]
  190. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022, 11, e1260. [Google Scholar] [CrossRef]
  191. Zheng, J.; Wittouck, S.; Salvetti, E.; Franz, C.M.A.P.; Harris, H.M.B.; Mattarelli, P.; O’Toole, P.W.; Pot, B.; Vandamme, P.; Walter, J.; et al. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int. J. Syst. Evol. Microbiol. 2020, 70, 2782–2858. [Google Scholar] [CrossRef]
  192. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
  193. Wieërs, G.; Belkhir, L.; Enaud, R.; Leclercq, S.; Philippart de Foy, J.-M.; Dequenne, I.; de Timary, P.; Cani, P.D. How probiotics affect the microbiota. Front. Cell. Infect. Microbiol. 2019, 9, 454. [Google Scholar] [CrossRef] [Green Version]
  194. Hao, W.-Z.; Li, X.-J.; Zhang, P.-W.; Chen, J.-X. A review of antibiotics, depression, and the gut microbiome. Psychiatry Res. 2020, 284, 112691. [Google Scholar] [CrossRef]
  195. Wall, R.; Cryan, J.F.; Ross, R.P.; Fitzgerald, G.F.; Dinan, T.G.; Stanton, C. Bacterial neuroactive compounds produced by psychobiotics. Adv. Exp. Med. Biol. 2014, 817, 221–239. [Google Scholar] [CrossRef]
  196. Lyte, M. Probiotics function mechanistically as delivery vehicles for neuroactive compounds: Microbial endocrinology in the design and use of probiotics. Bioessays 2011, 33, 574–581. [Google Scholar] [CrossRef]
  197. Yong, S.J.; Tong, T.; Chew, J.; Lim, W.L. Antidepressive mechanisms of probiotics and their therapeutic potential. Front. Neurosci. 2019, 13, 1361. [Google Scholar] [CrossRef] [Green Version]
  198. O’Mahony, S.M.; Clarke, G.; Borre, Y.E.; Dinan, T.G.; Cryan, J.F. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav. Brain Res. 2015, 277, 32–48. [Google Scholar] [CrossRef]
  199. Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F.; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
  200. Garcia-Gonzalez, N.; Battista, N.; Prete, R.; Corsetti, A. Health-Promoting Role of Lactiplantibacillus plantarum Isolated from Fermented Foods. Microorganisms 2021, 9, 349. [Google Scholar] [CrossRef]
  201. Chong, H.X.; Yusoff, N.A.A.; Hor, Y.Y.; Lew, L.C.; Jaafar, M.H.; Choi, S.B.; Yusoff, M.S.B.; Wahid, N.; Abdullah, M.F.I.L.; Zakaria, N.; et al. Lactobacillus plantarum DR7 alleviates stress and anxiety in adults: A randomised, double-blind, placebo-controlled study. Benef. Microbes 2019, 10, 355–373. [Google Scholar] [CrossRef]
  202. Liu, G.; Chong, H.-X.; Chung, F.Y.-L.; Li, Y.; Liong, M.-T. Lactobacillus plantarum DR7 Modulated Bowel Movement and Gut Microbiota Associated with Dopamine and Serotonin Pathways in Stressed Adults. Int. J. Mol. Sci. 2020, 21, 4608. [Google Scholar] [CrossRef] [PubMed]
  203. Hwang, Y.-H.; Park, S.; Paik, J.-W.; Chae, S.-W.; Kim, D.-H.; Jeong, D.-G.; Ha, E.; Kim, M.; Hong, G.; Park, S.-H.; et al. Efficacy and Safety of Lactobacillus Plantarum C29-Fermented Soybean (DW2009) in Individuals with Mild Cognitive Impairment: A 12-Week, Multi-Center, Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2019, 11, 305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Bravo, J.A.; Forsythe, P.; Chew, M.V.; Escaravage, E.; Savignac, H.M.; Dinan, T.G.; Bienenstock, J.; Cryan, J.F. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc. Natl. Acad. Sci. USA 2011, 108, 16050–16055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Messaoudi, M.; Lalonde, R.; Violle, N.; Javelot, H.; Desor, D.; Nejdi, A.; Bisson, J.-F.; Rougeot, C.; Pichelin, M.; Cazaubiel, M.; et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human subjects. Br. J. Nutr. 2011, 105, 755–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Desbonnet, L.; Garrett, L.; Clarke, G.; Kiely, B.; Cryan, J.F.; Dinan, T.G. Effects of the probiotic Bifidobacterium infantis in the maternal separation model of depression. Neuroscience 2010, 170, 1179–1188. [Google Scholar] [CrossRef]
  207. Marler, S.; Ferguson, B.J.; Lee, E.B.; Peters, B.; Williams, K.C.; McDonnell, E.; Macklin, E.A.; Levitt, P.; Gillespie, C.H.; Anderson, G.M.; et al. Brief report: Whole blood serotonin levels and gastrointestinal symptoms in autism spectrum disorder. J. Autism Dev. Disord. 2016, 46, 1124–1130. [Google Scholar] [CrossRef] [Green Version]
  208. Liu, Y.-W.; Liu, W.-H.; Wu, C.-C.; Juan, Y.-C.; Wu, Y.-C.; Tsai, H.-P.; Wang, S.; Tsai, Y.-C. Psychotropic effects of Lactobacillus plantarum PS128 in early life-stressed and naïve adult mice. Brain Res. 2016, 1631, 1–12. [Google Scholar] [CrossRef] [Green Version]
  209. Nimgampalle, M.; Kuna, Y. Anti-Alzheimer Properties of Probiotic, Lactobacillus plantarum MTCC 1325 in Alzheimer’s Disease induced Albino Rats. J. Clin. Diagn. Res. 2017, 11, KC01–KC05. [Google Scholar] [CrossRef]
  210. Liang, S.; Wang, T.; Hu, X.; Luo, J.; Li, W.; Wu, X.; Duan, Y.; Jin, F. Administration of Lactobacillus helveticus NS8 improves behavioral, cognitive, and biochemical aberrations caused by chronic restraint stress. Neuroscience 2015, 310, 561–577. [Google Scholar] [CrossRef]
  211. Desbonnet, L.; Clarke, G.; Shanahan, F.; Dinan, T.G.; Cryan, J.F. Microbiota is essential for social development in the mouse. Mol. Psychiatry 2014, 19, 146–148. [Google Scholar] [CrossRef] [Green Version]
  212. 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. [Google Scholar] [CrossRef]
  213. Dickerson, F.B.; Stallings, C.; Origoni, A.; Katsafanas, E.; Savage, C.L.G.; Schweinfurth, L.A.B.; Goga, J.; Khushalani, S.; Yolken, R.H. Effect of probiotic supplementation on schizophrenia symptoms and association with gastrointestinal functioning: A randomized, placebo-controlled trial. Prim. Care Companion CNS Disord. 2014, 16. [Google Scholar] [CrossRef] [Green Version]
  214. Gómez-Eguílaz, M.; Ramón-Trapero, J.L.; Pérez-Martínez, L.; Blanco, J.R. The beneficial effect of probiotics as a supplementary treatment in drug-resistant epilepsy: A pilot study. Benef. Microbes 2018, 9, 875–881. [Google Scholar] [CrossRef]
  215. Kim, B.; Hong, V.M.; Yang, J.; Hyun, H.; Im, J.J.; Hwang, J.; Yoon, S.; Kim, J.E. A Review of Fermented Foods with Beneficial Effects on Brain and Cognitive Function. Prev. Nutr. Food Sci. 2016, 21, 297–309. [Google Scholar] [CrossRef] [Green Version]
  216. 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, 183. [Google Scholar] [CrossRef] [Green Version]
  217. Santocchi, E.; Guiducci, L.; Prosperi, M.; Calderoni, S.; Gaggini, M.; Apicella, F.; Tancredi, R.; Billeci, L.; Mastromarino, P.; Grossi, E.; et al. Effects of probiotic supplementation on gastrointestinal, sensory and core symptoms in autism spectrum disorders: A randomized controlled trial. Front. Psychiatry 2020, 11, 550593. [Google Scholar] [CrossRef]
  218. Kalenik, A.; Kardaś, K.; Rahnama, A.; Sirojć, K.; Wolańczyk, T. Gut microbiota and probiotic therapy in ADHD: A review of current knowledge. Prog. Neuropsychopharmacol. Biol. Psychiatry 2021, 110, 110277. [Google Scholar] [CrossRef]
  219. Arteaga-Henríquez, G.; Rosales-Ortiz, S.K.; Arias-Vásquez, A.; Bitter, I.; Ginsberg, Y.; Ibañez-Jimenez, P.; Kilencz, T.; Lavebratt, C.; Matura, S.; Reif, A.; et al. Treating impulsivity with probiotics in adults (PROBIA): Study protocol of a multicenter, double-blind, randomized, placebo-controlled trial. Trials 2020, 21, 161. [Google Scholar] [CrossRef] [Green Version]
  220. Ng, Q.X.; Soh, A.Y.S.; Venkatanarayanan, N.; Ho, C.Y.X.; Lim, D.Y.; Yeo, W.-S. A systematic review of the effect of probiotic supplementation on schizophrenia symptoms. Neuropsychobiology 2019, 78, 1–6. [Google Scholar] [CrossRef]
  221. Tomasik, J.; Yolken, R.H.; Bahn, S.; Dickerson, F.B. Immunomodulatory Effects of Probiotic Supplementation in Schizophrenia Patients: A Randomized, Placebo-Controlled Trial. Biomark Insights 2015, 10, 47–54. [Google Scholar] [CrossRef]
  222. Nagamine, T. The potential of probiotics in the treatment of schizophrenia. CNPT 2021, 12, 18–22. [Google Scholar] [CrossRef]
  223. Severance, E.G.; Gressitt, K.L.; Stallings, C.R.; Katsafanas, E.; Schweinfurth, L.A.; Savage, C.L.G.; Adamos, M.B.; Sweeney, K.M.; Origoni, A.E.; Khushalani, S.; et al. Probiotic normalization of Candida albicans in schizophrenia: A randomized, placebo-controlled, longitudinal pilot study. Brain Behav. Immun. 2017, 62, 41–45. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Okubo, R.; Koga, M.; Katsumata, N.; Odamaki, T.; Matsuyama, S.; Oka, M.; Narita, H.; Hashimoto, N.; Kusumi, I.; Xiao, J.; et al. Effect of bifidobacterium breve A-1 on anxiety and depressive symptoms in schizophrenia: A proof-of-concept study. J. Affect. Disord. 2019, 245, 377–385. [Google Scholar] [CrossRef] [PubMed]
  225. Ghaderi, A.; Banafshe, H.R.; Mirhosseini, N.; Moradi, M.; Karimi, M.-A.; Mehrzad, F.; Bahmani, F.; Asemi, Z. Clinical and metabolic response to vitamin D plus probiotic in schizophrenia patients. BMC Psychiatry 2019, 19, 77. [Google Scholar] [CrossRef] [Green Version]
  226. Munawar, N.; Ahmad, A.; Anwar, M.A.; Muhammad, K. Modulation of Gut Microbial Diversity through Non-Pharmaceutical Approaches to Treat Schizophrenia. Int. J. Mol. Sci. 2022, 23, 2625. [Google Scholar] [CrossRef]
  227. Bravo, R.; Vicencio, J.M.; Parra, V.; Troncoso, R.; Munoz, J.P.; Bui, M.; Quiroga, C.; Rodriguez, A.E.; Verdejo, H.E.; Ferreira, J.; et al. Increased ER-mitochondrial coupling promotes mitochondrial respiration and bioenergetics during early phases of ER stress. J. Cell Sci. 2011, 124, 2511. [Google Scholar] [CrossRef] [Green Version]
  228. Lew, L.-C.; Hor, Y.-Y.; Yusoff, N.A.A.; Choi, S.-B.; Yusoff, M.S.B.; Roslan, N.S.; Ahmad, A.; Mohammad, J.A.M.; Abdullah, M.F.I.L.; Zakaria, N.; et al. Probiotic Lactobacillus plantarum P8 alleviated stress and anxiety while enhancing memory and cognition in stressed adults: A randomised, double-blind, placebo-controlled study. Clin. Nutr. 2019, 38, 2053–2064. [Google Scholar] [CrossRef]
  229. Rao, A.V.; Bested, A.C.; Beaulne, T.M.; Katzman, M.A.; Iorio, C.; Berardi, J.M.; Logan, A.C. A randomized, double-blind, placebo-controlled pilot study of a probiotic in emotional symptoms of chronic fatigue syndrome. Gut Pathog. 2009, 1, 6. [Google Scholar] [CrossRef] [Green Version]
  230. Liu, Y.-W.; Liong, M.T.; Chung, Y.-C.E.; Huang, H.-Y.; Peng, W.-S.; Cheng, Y.-F.; Lin, Y.-S.; Wu, Y.-Y.; Tsai, Y.-C. Effects of Lactobacillus plantarum PS128 on Children with Autism Spectrum Disorder in Taiwan: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2019, 11, 820. [Google Scholar] [CrossRef]
Figure 1. The gut–brain axis and the main effects during healthy and pathological conditions.
Figure 1. The gut–brain axis and the main effects during healthy and pathological conditions.
Antibiotics 11 01767 g001
Table 1. Effect of 3 antibiotic classes on human gut bacteria levels [189,190].
Table 1. Effect of 3 antibiotic classes on human gut bacteria levels [189,190].
AntibioticsHuman Gut Bacteria
Penicillin
(Amoxicillin, ampicillin, Oxacillin, PenV)
↑Enterobacteria
Bacteroidaceae
↓Bifidobacteria
↓Lactobacilli
↓Eubacteria
Cephalosporins
(Cefalor, Cafotaxime, Cefuroxime, Cefepime)
↑Clostridia
Bacteroides spp.
E. coli
↓Bifidobacteria
Enterobacteriaceae
Macrolides
(Azithromycin, Clarithromycin, Erythromycin, Spiramycin)
↑Enterococci
↑Streptococci
Bacteroidetes
↑Enterobacteria
↓Actinobacteria
Clostriales spp.
Veillonella spp.
↓decrease; ↑increase
Table 2. Preclinical and clinical evidence of the use of probiotics in NDDs.
Table 2. Preclinical and clinical evidence of the use of probiotics in NDDs.
BacteriaHealth ConditionExperimental ModelMain OutcomesReference
L. rhamnosus JB-1Healthy conditionAdult male BALB/c mice↓stress-induced corticosterone and anxiety- and depression-related behavior
Modulation of GABA expression at the brain level via the vagus nerve
[227]
L. plantarum DR7Mental stress conditionAdult patients↓stress; ↓anxiety; ↑cognitive functions
↑dopamine and norepinephrine
↓plasma cortisol and pro- inflammatory cytokines
[201]
L. plantarum DR7Mental stress conditionAdult patients↓stress; ↓anxiety; ↑cognitive functions
Modulation of stress-induced bowel movement and gut microbiota in association with dopamine and serotonin
[202]
L. plantarum P8Mental stress conditionAdult patients↓stress; ↓anxiety; ↑memory and cognitive traits
↓pro-inflammatory markers
[228]
L. plantarum MTCC1325Neurodegenerative disordersAlbino ratsBehavioral changes; ↓cognitive deficits
↓acetylcholine levels
[209]
L. plantarum C29Adult with mild cognitive impairmentsAdult patients↑cognitive functions especially in the attention domain
↑serum BDNF levels
[203]
L. casei ShirotaChronic fatigue syndromeAdult patients↓anxiety symptoms with modulation of gut microbiota[229]
B. infantis 35624Maternal separation (MS) modelMS adult rat offspringsNormalization of the immune response; ↓behavioral deficits
↓noradrenaline in the brain
↓depression-like behavior;
↓5-HT, noradrenaline and dopamine levels; ↑peripheral IL6
[206]
L. helveticus R0052 combined with B. longum R0175Induced stressAdult Wistar rats↓anxiety-like behavior; ↓stress-induced gastrointestinal discomfort[205]
L. helveticus R0052 combined with B. longum R0175Induced stressAdult patients↓psychological distress; ↓stress-induced gastrointestinal discomfort[205]
L. helveticus NS8Chronic restraint induced stressSprague-Dawley rats↑intestinal barrier and BBB; ↓global inflammation status
↑BDNF; ↓serotonin and noradrenaline in the hippocampus
↑circulating anti-inflammatory cytokines
[210]
L. plantarum PS128Early life stress conditionAdult
C57BL/6J mice
↓locomotor activities anxiety-like and depression-like behaviors
↓serum corticosterone and inflammatory cytokine levels
↑anti-inflammatory cytokine levels
↑dopamine and serotonin in the prefrontal cortex
[208]
L. plantarum PS128ASDChildren↓Age-dependent autism symptoms; ↓behavioral aspects, such as disruptive and rule-breaking attitudes and hyperactivity/impulsivity[230]
Bacteroides fragilisASDMaternal immune activation murine model↓ASD-related defects in communicative, stereotypic, anxiety-like and sensorimotor behaviors
Restoration of gut permeability and modulation of gut microbial composition
[128]
Vivomixx VSL#3ASD12-year-old boy↓severity of abdominal symptoms; ↓Autistic core symptoms
Modulation of gut microbiota and positive regulation of intestinal barrier
[212]
Vivomixx VSL#3ASDPreschool children↓GI symptoms; ↑multisensory processing, adaptive functions, and developmental pathways[216,217]
L. acidophilus DSM32241, L. plantarum DSM32244, L. casei DSM32243, L. helveticus DSM32242, L. brevis DSM11988, B. lactis DSM32246, B. lactis DSM32247, and S. salivarius subsp. thermophilus DSM32245.Drug-resistant EpilepsyAdult patients↓epileptic seizures; ↑quality of life
↓serum IL-6 and sCD14; ↑serum GABA
[214]
L. rhamnosus GGADHDInfantsPreventive effect in reducing the risk of developing ADHD[87]
L. rhamnosus GG combined with B. animalis subsp. lactis Bb12SCZAdult patients↓severe bowel difficulty and prevention of common somatic symptoms associated with SCZ
Positive effects on digestion and on GI disorders such as chronic constipation
[213]
L. rhamnosus GG combined with B. animalis subsp. lactis Bb12SCZAdult patients↑intestinal barrier integrity; ↓bowel difficulties via modulation of inflammatory cytokines belonging to IL17 family
no significant impact on positive and negative syndrome scale (PANSS) psychiatric symptom scores
[221]
BIO-THREE®SCZAdult patients↓constipation; ↓insulin resistance; ↓intestinal inflammation[222]
B. breve A-1SCZAdult patientsImproved PANSS scores; ↓depression; ↓anxiety
↓symptoms with a modulation of IL22 and tumor necrosis factor-related activation induced cytokines (TRANCE)
[229]
B. bifidum, L. acidophilus, L. fermentum and L. reuteri combined with vitamin DSCZAdult patientsAmelioration of PANSS scores
↓inflammation; ↑plasma total antioxidant capacity
[225]
↓decrease; ↑increase
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Diamanti, T.; Prete, R.; Battista, N.; Corsetti, A.; De Jaco, A. Exposure to Antibiotics and Neurodevelopmental Disorders: Could Probiotics Modulate the Gut–Brain Axis? Antibiotics 2022, 11, 1767. https://doi.org/10.3390/antibiotics11121767

AMA Style

Diamanti T, Prete R, Battista N, Corsetti A, De Jaco A. Exposure to Antibiotics and Neurodevelopmental Disorders: Could Probiotics Modulate the Gut–Brain Axis? Antibiotics. 2022; 11(12):1767. https://doi.org/10.3390/antibiotics11121767

Chicago/Turabian Style

Diamanti, Tamara, Roberta Prete, Natalia Battista, Aldo Corsetti, and Antonella De Jaco. 2022. "Exposure to Antibiotics and Neurodevelopmental Disorders: Could Probiotics Modulate the Gut–Brain Axis?" Antibiotics 11, no. 12: 1767. https://doi.org/10.3390/antibiotics11121767

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop