Abstract
The intestinal microbiota are important in proper human growth and development before and after birth, during infancy and childhood. Microbial composition may yield insights into the temporal development of microbial communities and vulnerabilities to disorders of microbial ecology such as recurrent Clostridium difficile infection. Discoveries of key microbiome features of carbohydrate and amino acid metabolism are lending new insights into possible new therapies or preventative strategies for inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS). In this review, we summarize the current understanding of the development of the pediatric gastrointestinal microbiome, the influence of the microbiome on the developing brain through the gut-brain axis, and the impact of dysbiosis on the development of disease. Microbial dysbiosis will be explored in the context of pediatric allergy and asthma, recurrent C. difficile infection, IBD, IBS, and metabolic disorders. The central premise is that the human intestinal microbiome plays a vital role throughout human life beginning in the prenatal period and extending throughout childhood in health and disease.
Keywords: children, early life, gut-brain axis, gut microbes, microbiota, neonatal
INTRODUCTION
The human intestine harbors trillions of microbial cells which form a symbiotic relationship with the host and play a vital role in both health and disease. While the specific microbial composition varies among healthy individuals, the functional repertoire of the microbiome is conserved1. These microbes play important roles in mammalian homeostasis, including providing essential nutrients2, 3, metabolizing dietary fiber into short chain fatty acids4, and ensuring proper development of the immune system5. Therefore, the gut microbiota is considered a crucial factor for proper early life development and lifelong health. However, when the balance of the intestinal microbiota becomes disrupted, alterations can lead to immunologic dysregulation and the development of diseases including Clostridium difficile infection6, inflammatory bowel disease7, 8, irritable bowel syndrome9, 10, asthma11, obesity12, and neurodevelopmental disorders such as autism13. In this review, we describe the development of the pediatric microbiome, starting in utero and progressing through infancy, childhood and adolescence. We then discuss the impact of the microbiome on the developing brain and neural function through the gut-brain axis. We conclude with a discussion on the impact of dysbiosis on disease development.
ESTABLISHMENT OF EARLY LIFE INTESTINAL MICROBIOME
Microbial Colonization of the Neonatal Gut
For many years it was believed that the fetus’ in utero environment was sterile, with infant gut colonization beginning at the time of delivery. However, recent work demonstrating the presence of a microbial community in the meconium14, 15 has challenged this notion. While still controversial16, it is now clear that microbial colonization of the infant gut may begin prior to birth as additional evidence suggests microbial colonization of the placenta17, 18, amniotic fluid19, 20, and the umbilical cord21. Aagaard et al17 collected 320 placental specimens under sterile conditions and found a unique placental microbiome niche, which most closely resembled the human oral microbiome17. Furthermore, a randomized, double blind, placebo-controlled trial demonstrated that maternal probiotic supplementation could affect the expression of Toll-like receptor (TLR)-related genes in both the placenta and the fetal intestine.19 This finding suggested that the fetal intestinal immune gene expression profile could be affected by microbial contact in utero. Similarities between the unique microbiota composition of the placenta and amniotic fluid to that of the infant meconium further suggests a prenatal microbial transfer from mother to fetus.22 Importantly, variations in the placental microbiome have been shown to associate with preterm birth17 as well as low birth weight in full term infants.18
In addition to potential in utero environmental influences, many factors have been found to contribute to early intestinal colonization, such as gestational age at birth. Studies have shown that the intestinal microbiota of preterm infants differs from that of healthy term infants23, with preterm infant microbiomes being dominated by Enterobacter, Staphylococcus, and Enterococcus24, 25. Prematurity is associated with a high risk for neonatal complications and can lead to significant morbidity and mortality26. These premature neonates are often exposed to prolonged hospitalizations, antibiotics, and formula feeding which may all disrupt the maturation of health-associated microbial communities27. Importantly, alterations in the microbiome of preterm infants have been correlated with increased risk for complications such a necrotizing enterocolitis28, 29 and late-onset sepsis30, 31.
Another major influence on the infant gut microbiome is infant diet. Breast-fed infants have microbiota enriched in Lactobacillus, Staphylococcus, and Bifidobacterium, as compared to formula-fed infants with microbiomes dominated by Roseburia, Clostridium, and Anaerostipes32. Formula-fed infants have greater quantities of microbes associated with inflammation, with a more rapid maturation of their microbiome toward that of an adult-type composition27, 32–34. Conversely, studies have shown that human milk isolates contain symbiotic and potentially probiotic microbes32, 33. Human milk oligosaccharides, the 3rd largest component in human milk, are considered prebiotic, with antimicrobial and antiadhesive properties thought to be protective to the infant35–38. Interestingly, breast-fed infants have reduced microbial diversity than their formula-fed counterparts39. However, this reduced diversity is associated with an increase in genes relevant for degradation of human milk oligosaccharides (HMOs)39. HMOs, in turn, are able to amplify the presence of specific bacterial populations in the infant gut35.
Development of the infant intestinal microbiota
During the first year of an infant’s life, the relatively simple neonatal microbiome matures and develops into a more complex microbiome, with a composition more representative of an adult gastrointestinal tract enriched in Bacteroides and Firmicutes32, 40. During the first year of life, the infant’s microbiome also gains functionality similar to their mother’s gut metagenome, with decreasing inter-individual variation over time32, 40. An increased number of bacterial genes relevant for plant polysaccharide metabolism primes the infant microbiome for the adult diet even before the introduction of solid foods41. Once solid foods are introduced, there is a sustained shift in the microbial composition with an increase in Bacteroidetes. Additional modifications include increased short chain fatty acids in the stool, and expression of genes relevant for carbohydrate metabolism, vitamin biosynthesis, and xenobiotic degredation41.
During the early infant development period many exposures can influence the progression of the intestinal microbiota. For example, antibiotic treatment during this period of early life development can dramatically alter the intestinal microbiota structure40–42. Similarly, exposure to less sanitary environments, including contact with household pets and siblings, have significant effects on the developing microbiome42, 43. In fact, the number of older siblings positively correlates with bacterial diversity and richness at 18 months of age, with increasing relative abundances of Firmicutes and Bacteroidetes in infants with more siblings44.
Conversely, the microbiome also affects the general health status of the infant or child. A longitudinal comparative study of Malawian twins discordant for kwashiorkor found that the malnourished twin displayed abnormal microbiome signatures compared to the healthy twin45. As proof of concept that the microbiome was a causal factor in the development of kwashiorkor phenotype, frozen fecal communities from the discordant twin pairs were transplanted into gnotobiotic mice. The mice receiving kwashiorkor microbiome exhibited marked weight loss with accompanied perturbations in amino acid, carbohydrate and intermediary metabolism45.
Development of pediatric and adolescent intestinal microbiota
While some investigators have suggested that the pediatric microbiome reaches a relatively stable, adult-like configuration within the first 3 years of life32, 46, other studies have demonstrated continued development through childhood into the teenage years47–49. In a study comparing the intestinal microbiota of 1–4 year old children to heathy adults, the adult microbiome had significantly greater diversity (abundance and richness) than young children48. At the phylum-like level, the predominant bacterial groups were similar, including Firmicutes, Bacteroidetes and Actinobacteria48. However, at the genus level, multiple phylogenetic groups were significantly different between the children and adult populations48.
A study comparing the fecal microbiota of adolescents (11–18 years of age) to healthy adults found that the number of detected species were similar between groups, but the relative abundances of genera differentiated adolescents from adults, suggesting that the microbiomes differed, even into adolescence49. Hollister et al47 compared 7–12 year old children to adults, and found that similar to adults, the pediatric gut microbiome was largely composed of Bacteroidetes and Firmicutes (Figure 1). However, the relative abundances of these bacteria differed from adults, with relatively lesser abundances of Bacteroidetes and greater abundances of Firmicutes and Actinobacteria47. They also found that while many taxa were shared between pediatric and adult samples, the distribution was significantly different, with children having greater abundances of bacteria belonging to the genera Faecalibacterium, Dialister, Roseburia, Ruminococcus, and Bifidobacterium47.
Figure 1.
Healthy pediatric and adult gastrointestinal tracts differ in relative abundances of gut bacterial taxa. (a) Phylum level relative abundances via 16S rRNA gene sequencing. Genus level relative abundances by (b) 16S sequencing and (c) shotgun metagenomic sequencing. (Adapted from Hollister et al.47)
Importantly, Hollister et al47 also characterized the metagenomic profiles of pediatric and adult microbiomes. Children demonstrated an enrichment of genes which may support ongoing development, including genes involved in vitamin synthesis, de novo folate synthesis, and amino acid metabolism47. Meanwhile, adults were enriched in pathways previously linked to inflammation, including genes involved in oxidative phosphorylation, lipopolysaccharide biosynthesis, flagellar assembly, and steroid hormone biosynthesis47. While the intestinal communities of children shared 35–46% similarity to each other taxonomically, they had substantially greater overlap at the functional level, with >90% similarity of the ortholog group and pathway levels47. This difference implies that the functional capacity of microbes present in the pediatric gastrointestinal tract is more highly conserved than microbial composition.
THE GUT-BRAIN AXIS
Impact on brain development
The human brain undergoes rapid growth during the perinatal period, corresponding to dramatic changes in the maternal microbiota50. Mothers demonstrate an increase in Proteobacteria and Actinobacteria, and a decreased richness as they progress from the first to the third trimester of pregnancy50. While these changes are often correlated with metabolic syndrome in nonpregnant females, in the setting of pregnancy these changes are beneficial in promoting energy storage and allowing for adequate growth of the fetus50.
Many studies have demonstrated the importance of the microbiota during brain development, including the microbiome’s indirect effect on tryptophan metabolism and serotonin (5-HT) synthesis (Figure 2). 5-HT is known to be crucial to CNS development51. Knock-in mice lacking the tryptophan hydroxylase 2 gene, demonstrated that a lack of brain 5-HT caused improper wiring of the brain that may lead to long-term changes and neurodevelopmental disorders51. When compared to specific pathogen free (SPF) mice, germ-free mice have increased motor activity and decreased anxiety, as well as altered levels of neurotransmitters such as noradrenaline, dopamine and 5-HT52. Importantly, these abnormalities can be prevented by exposing the mice to gut microbiota early in life52. Germ-free mice also have a decreased kynurenine:tryptophan ratio compared to conventionally raised mice, which normalizes upon exposure to gut microbiota immediately after weaning53. Similarly, aberrant anxiety responses were corrected with bacterial colonization of the gut in these animals53. Furthermore, rats treated with Bifidobacterium infantis showed reduced 5-HIAA concentrations in the frontal cortex, and increased plasma concentrations of tryptophan and kynurenic acid compared to controls54. Gut bacteria can also have direct effects on the metabolism and synthesis of tryptophan and 5-HT55. For example, there is in vitro evidence that some bacterial strains can produce 5-HT from tryptophan55. Meanwhile, some bacteria are able to synthesize tryptophan using enzymes such as tryptophan synthase56, 57, while others degrade tryptophan with tryptophanase enzyme57, 58.
Figure 2.
The bidirectional gut-brain axis. The gut-brain axis is a complex interplay between the central nervous system, the neuroendocrine and neuroimmune systems, the autonomic nervous system, the enteric nervous system, and the microbiota. 5-HT, 5-hydroxytryptamine. DC, dendritic cell. GABA, γ-aminobutyric acid. (Adapted from Collins et al.136)
Colonic bacteria have an important role of fermenting carbohydrates and proteins to produce metabolites, including short chain fatty acids (SCFA), which are essential for human health4. Erny et al determined that SCFA regulate microglial homeostasis59. Therefore, defective microglia were found in mice with altered microbiota, including germ-free mice, mice with temporal eradication of microbiota, and mice with limited microbial complexity59. Exposure to an indigenous microbiota during early development is also crucial for the development of a normal hypothalamic-pituitary-adrenal (HPA) system60. Germ-free mice display an exaggerated HPA stress response, which was reversible with exposure to specific pathogen-free (SPF) feces during early development60. However, reconstitution with SPF feces at a later developmental stage was ineffective at reducing the aberrant stress response60. This study demonstrates the important role that the microbiota play during early postnatal brain development, while brain plasticity may still be preserved.
Bidirectional gut-brain axis and its impact on brain function
The brain-gut-microbiota axis is a complex interplay between the CNS, the neuroendocrine and neuroimmune systems, the sympathetic and parasympathetic arms of the autonomic nervous system, the enteric nervous system, and the microbiota10. The communication throughout this axis is bidirectional, with brain signals affecting gastrointestinal tract motor, sensory and secretory functions, and simultaneous visceral signaling from the GI tract affecting brain function61 (Figure 2).
Long-term treatment with the probiotic L. rhamnosus (JB-1) led to decreased levels of stressinduced corticosteroids, depressive symptoms, and anxiety62. Treatment with L. rhamnosus also induced region-specific alterations in expression of GABAB1b and GABAAα262. Alterations in GABA expression are implicated in depression and anxiety disorders. Importantly these changes were not found in vagotomized mice, implicating the vagus nerve as a direct line of communication between the gut bacteria and the brain62.
Another study has implicated the gut microbiome in pain perception. Certain strains of Lactobacillus induce increased expression of μ-opioid and cannabinoid receptors in intestinal epithelial cells, mimicking the analgesic effects of morphine63.
Similarly, the brain can affect the composition of the gut microbiota. These effects on the microbiota can be indirect, through changes in motility and secretion, or direct, through signaling molecules released into the gastrointestinal tract via enterochromaffin cells, neurons, and immune cells64. The autonomic nervous system (ANS) affects motility as well as mucus secretion into the gut lumen, both of which can alter the gastrointestinal environment, thereby changing the bacteria that are present65, 66. The ANS can also affect epithelial mechanisms involved in immune activation of the gut64. For example, exposure to stressful stimuli has been shown to increase permeability of the epithelium, allowing bacterial antigens to cross the epithelium and stimulate an immune response in the mucosa, which in turn alters the microbiome67. This increased permeability is secondary to mast cell degranulation, overproduction of interferon-γ, and decreased expression of mRNA encoding tight junction proteins68.
DYSBIOSIS AND DISEASE
While it is clear that the maintenance of the microbiome is vital for preservation of health, imbalances in the microbiome can shift the microbiome-host relationship from symbiotic to pathogenic. Below, we discuss some examples of communicable and noncommunicable disorders that are associated with a dysbiotic microbiome.
Clostridium difficile Infection
Clostridium difficile infection (CDI) is the leading nosocomial infection in the United States, affecting more than 500,000 people annually69. A complex microbial community in the intestine is vitally important to providing colonization resistance to CDI. Therefore, alterations to the microbiota increases the risk of infection from C. difficile. When comparing patients with CDI to diarrheal and nondiarrheal controls, Schubert et al6 determined that Ruminococcaceae, Lachnospiraceae, Bacteroides and Porphyromonadaceae were absent in patients with CDI, but highly associated with nondiarrheal controls. These compositional changes are even more pronounced in patients with recurrent CDI who have been exposed to multiple courses of antbiotics70. Recurrent CDI leads to increased abundance of Proteobacteria, with decreased abundances of Bacteroides and Firmicutes compared to healthy controls71.
Antibiotic use is the most common risk factor for the development of CDI, with often longlasting changes to the microbiota72. These antibiotic-related changes included decreased taxonomic and functional diversity of the gut microbiome as well as a decreased colonization resistance against invading pathogens73. Additionally, Denève et al74 discovered that exposure to subinhibitory concentrations of certain antibiotics upregulated the expression of genes encoding colonization factors in C. difficile, and increased the adherence of C. difficile to cultured cells.
Proton pump inhibitor (PPI) use also increases the risk of CDI75. PPI use has been shown to alter the gut microbiota by decreasing microbial diversity, and decreasing the abundances of commensal microorganisms76, 77. Chronic PPI use leads to decreased abundances of Bacteroidetes and increased abundances of Firmicutes at the phylum level, which may predispose patients to CDI78. PPIs also increase fecal Enterococcaceae and Streptococcaceae, taxa which have been associated with CDI77, 79.
Therapies aimed at correcting and restoring health-associated complex microbial communities have yielded successful outcomes in treating CDI. Fecal microbiota transplantation (FMT) is around 90% effective at curing recurrent CDI with one or more infusions80. The rationale behind FMT for CDI is that restoration of the fecal community structure could also restore function, including colonization resistance81. Weingarden et al71 demonstrated that FMT normalizes both bacterial community composition and metabolic capacity. Pre-FMT fecal samples had greater concentrations of primary bile acids and bile salts, while post-FMT samples contained mostly secondary bile acids71. Patients with recurrent CDI yielded disrupted abilities to convert primary bile acids to secondary bile acids. Primary bile acids have been shown to promote germination and growth of C. difficile, while secondary bile acids inhibit this growth82, 83. Therefore, it is possible that the correction of bile acid metabolism, and not just restoration of community structure, is a crucial mechanistic element in the efficacy of FMT against CDI71.
Inflammatory bowel disease
Inflammatory bowel disease (IBD), including Crohn’s disease and ulcerative colitis, are also associated with a dysbiotic microbiome7, 8. However, it is unclear if this dysbiosis plays a role in the etiology of the disease, is a result of the disease, or both. Patients with IBD demonstrated exaggerated immune responses against commensal intestinal microbes, which may be essential for the development of intestinal inflammation84, 85. However, others theorize that an aberrant intestinal microbiota is the primary driver of inflammation in IBD86. In support of this theory, studies have shown that treatment with antibiotics can substantially decrease intestinal inflammation and improve IBD symptoms87. Also, many microbes which are enriched in IBD may be able to potentiate disease88. For example, Ohkusa et al89 demonstrated that the abundance of Fusobacterium species is increased in patients with ulcerative colitis compared with healthy controls. In a separate study, Ohkusa et al90 showed that when given as an enema, the isolated human strain of Fusobacterium varium caused UC-like colonic ulcerations in mice, indicating that this bacterium may play a role in the pathogenesis of ulcerative colitis.
Patients with IBD have a variety of changes in the fecal microbiota including decreased abundances of Bacteroides, Firmicutes, Clostridia, Bifidobacterium and Lactobacillus, as well as increased abundances of Fusobacterium and adherent-invasive Escherichia coli88, 91, 92. Patients with IBD have reduced diversity of the microbiome, which becomes more pronounced in inflamed as compared to noninflamed tissue93. Additionally, the functional composition of the gut microbiota is altered in IBD, with one study showing an alteration of 12% of pathways analyzed compared to 2% of genera between IBD and healthy individuals94. Functional alterations in IBD patients include diminished carbohydrate metabolism and decreased production of butyrate and other short chain fatty acids88, 94, 95.
Given the importance of the microbiome in IBD, therapies aimed at manipulating the microbiome have gained popularity. While there are some promising studies demonstrating the efficacy of certain antibiotic combinations in treating IBD, more controlled trials are needed96. Similarly, while studies exploring the use of probiotics in IBD have yielded encouraging results, results vary across studies and more randomized controlled trials are needed97–99. Finally, fecal microbiota transplantation (FMT) is a possible therapy for IBD, given its success in treating recurrent C. difficile infection100. Few randomized controlled trials have been conducted in patients with IBD, with varying results101–104, and more controlled trials are needed to fully understand the therapeutic potential of FMT for the treatment of IBD.
Irritable Bowel Syndrome
Evidence for the microbiome’s influence on disease is found in irritable bowel syndrome (IBS)9, 10. IBS is diagnosed based on the presence of chronic recurrent abdominal pain related to defecation or associated with changes in frequency or form of stool, without accompanying warning signs105. Multiple studies have demonstrated differences in the fecal microbial communities of patients with IBS compared to healthy controls106. Patient with IBS show reductions in the relative abundance of Bifidobacterium and Lactobacillus, as well as increased abundances in the Firmicutes:Bacteroidetes ratio9, 107. Similarly, a small number of studies have shown shifts in the small intestinal microbiota in patients with IBS108, including evidence that small intestinal bacterial overgrowth may play a role109. While the majority of work has been done in adults with IBS, Saulnier et al110 confirmed differences in microbiome signatures in pediatric IBS compared to healthy controls. Additionally, they were able to classify different subtypes of IBS using a limited set of discriminatory species, with a success rate of 98.5%110.
Further support for the role of gut microbiome perturbations in the development of IBS is the persistence of IBS-like symptoms following confirmed bacterial or viral gastroenteritis, a term called post-infectious IBS111. Proposed mechanisms for post-infectious IBS include enteroendocrine cell hyperplasia, elevated T-lymphocytes, and increased gut permeability following infection112, 113.
To further elucidate the role of the intestinal microbiota in IBS, multiple studies have examined the effects of probiotic supplementation in this disorder. Based on several meta-analyses and systematic reviews, probiotic use for treatment of IBS appears more effective than placebo114–116. However, many of these studies vary in their specific conclusions, likely due to inadequate sample sizes, weak study design, and the use of various probiotic strains making comparisons difficult117. In an effort to clarify which organisms were potentially effective in improving IBS symptoms, Ortiz-Lucas et al118specifically examined 10 randomized controlled trials. They found that probiotic combinations containing Bifidobacterium breve, Bifidobacterium longum, or Lactobacillus acidophilus improved pain scores118. Meanwhile, probiotics containing Bifidobacterium breve, Bifidobacterium longum, Lactobacillus casei, or Lactobacillus plantarum improved distension scores118.
Another therapeutic avenue for the treatment of IBS has included dietary changes. Fermentable carbohydrates can be difficult to absorb and have been shown to contribute to symptoms in IBS. Consistent with this finding, a low FODMAP (fermentable oligosaccharides, disaccharides, monosaccharides and polyols) diet has been shown to decrease symptoms in adults with IBS119, 120. Chumpitazi et al121 confirmed that this response was also true in pediatric IBS. Additionally, they demonstrated that specific microbial signatures were associated with the efficacy of the FODMAP diet121. Specifically, FONDMAP responders had baseline microbiomes enriched with taxa with greater saccharolytic metabolic capacity and metabolic pathways related to carbohydrate metabolism121.
Allergy and Asthma
The development of asthma and allergies has been associated with deviations in the developing microbiota. For example, infants colonized with Escherichia coli were at an increased risk of developing eczema, while infants colonized with Clostridium difficile were at increased risk of all atopic outcomes (eczema, recurrent wheeze, and allergic sensitization)122. Similarly, Clostridium difficile colonization at 1 month of age was associated with asthma at 6 to 7 years of age123. Antibiotic exposure in the first year of life has also been associated with an increased risk for the development of asthma in children, with this risk increasing in parallel with the number of courses of antibiotics prescribed11.
These human findings have been further explored using mouse models of allergy and atopy. Allergic germ-free mice developed more severe disease than conventionally housed controls124. Importantly, this phenotype could be reversed by recolonization of the germ-free mice with conventional microbiota, demonstrating the important and influential role of the microbiota in allergic conditions124. Furthermore, in a mouse model of allergic airway inflammation (asthma), symptoms could be attenuated by exposure to Lactobacillus reuteri, but not Lactobacillus salivarius125, signifying the importance of the bacterial species and strains that are present. In line with these findings, Russell et al126 demonstrated that exposure to vancomycin, but not streptomycin, increased the severity of murine allergic asthma.
Obesity
Obesity has become a major global health problem, with increasing prevalence127. The patterns of maturation of microbial communities in infancy can affect the relative risk of becoming overweight and obese in later childhood. A recent longitudinal study of more than 900 infants found that mode of delivery and infant gut microbiota (specifically belonging to the Lachnospiraceae family) mediated the association between prepregnancy maternal overweight status and overweight status of children at 1 and 3 years of age128. Another study found that low levels of Bifidobacterium spp. and increased Staphlococcus aureas in infancy was associated with being overweight by age seven12.
Further support for the microbiome’s role in obesity was demonstrated by Cho et al129. In this study low dose antibiotic exposure in young mice led to increased adiposity, metabolic hormone levels, and SCFA levels, as well as changes to the hepatic metabolism of lipids and cholesterol129. Additional work by Cox et al130 found that low dose penicillin given at birth can induce sustained effects on body composition and enhance high fat diet-induced obesity in mice. Furthermore, the obese phenotype was transferable to germ-free mice by transfer of low-dose penicillin microbiome130, implicating the microbiome as the driver of this phenotype as opposed to antibiotics.
Autism Spectrum Disorder
While the underlying etiology of autism or autism spectrum disorder is not well understood, the intestinal microbiota is proposed to play a role in the development of autism. Children with autism have dysbiotic fecal microbiota, with greater abundances of Bacteroidetes and lesser abundances of Firmicutes compared to controls13. Luna et al131 compared the mucosal microbiome of autistic children with functional abdominal pain to neurotypical children with function abdominal pain, and found distinct microbial signatures in autistic children that correlate with cytokine quantities and tryptophan homeostasis.
Children with regressive (late-onset) autism have increased numbers of fecal clostridial species, as well as the presence of non-spore-forming anaerobes and microaerophilic bacteria, which were absent in control children132. Due to frequent parental reports of an antecedent antibiotic exposure followed by chronic diarrhea in regressive autism, Sandler et al133 hypothesized that, in some children, antibiotic-induced disruption of the microbiome may facilitate colonization by autism-promoting bacterial species. They tested this hypothesis by treating 10 autistic children with minimally absorbed oral vancomycin, and found that 8 of 10 children had short-term improvement in autistic symptoms133. While the improvements were not long-lasting, this report indicates a potential role for the gut microbiota in the symptomatology of autism spectrum disorder and thus warrants further investigation.
Murine studies have also supported a role of the microbiome in autism. Buffington et al134 demonstrated impaired social behavior in the offspring of dams fed a high-fat diet, which were mediated by changes in the offspring’s microbiota. While these pups’ microbiota were notable for a significant reduction in Lactobacillus reuteri, supplementation with this bacterium reversed the observed social deficits134. deTheije et al135 found that in utero valproic acid exposure resulted in decreased social behavior scores and impacted the gut microbiota of mice, with specific changes in Bacteroidetes and Firmicutes, similar to human autism studies. Together these results establish that in murine models of autism, behavioral alterations have been associated with altered microbial colonization.
CONCLUSIONS
In this review, we summarized the current understanding of the development of the pediatric microbiome, the impact of the microbiome on the developing brain and brain function through the gut-brain axis, and the impact of dysbiosis on disease development. The intestinal microbiome is an important factor in human growth and development, and the appropriate balance of microbes throughout life plays a crucial role in the both health and disease. As emerging technology allows us to understand more about the microbiome and its many important functions, we in turn begin to understand the disease processes that the microbes impact. With this deeper knowledge and understanding comes the hope of new therapeutic targets and avenues through which to treat these diseases and promote human health across life stages and ages.
Acknowledgments
Funding Sources Statement: This work was supported by the National Institutes of Health (U01 CA170930), Texas Medical Center Digestive Disease Center (P30 DK56338), and unrestricted research support from BioGaia AB (Stockholm, Sweden) (J.V.).
Footnotes
Conflict of Interest Statement: FDI has no conflicts to disclose. J.V. receives unrestricted research support from BioGaia AB.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
REFRENCES
- 1.Human Microbiome Project C Structure, function and diversity of the healthy human microbiome. Nature 2012;486:207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Flint HJ, Scott KP, Duncan SH, et al. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 2012;3:289–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.LeBlanc JG, Milani C, de Giori GS, et al. Bacteria as vitamin suppliers to their host: a gut microbiota perspective. Curr Opin Biotechnol 2013;24:160–8. [DOI] [PubMed] [Google Scholar]
- 4.Russell WR, Hoyles L, Flint HJ, et al. Colonic bacterial metabolites and human health. Curr Opin Microbiol 2013;16:246–54. [DOI] [PubMed] [Google Scholar]
- 5.Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 2009;9:313–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Schubert AM, Rogers MA, Ring C, et al. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. MBio 2014;5:e01021–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sartor RB. Key questions to guide a better understanding of host-commensal microbiota interactions in intestinal inflammation. Mucosal Immunol 2011;4:127–32. [DOI] [PubMed] [Google Scholar]
- 8.Frank DN, Robertson CE, Hamm CM, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm Bowel Dis 2011;17:179–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mayer EA, Savidge T, Shulman RJ. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology 2014;146:1500–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Grenham S, Clarke G, Cryan JF, et al. Brain-gut-microbe communication in health and disease. Front Physiol 2011;2:94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Marra F, Marra CA, Richardson K, et al. Antibiotic use in children is associated with increased risk of asthma. Pediatrics 2009;123:1003–10. [DOI] [PubMed] [Google Scholar]
- 12.Kalliomaki M, Collado MC, Salminen S, et al. Early differences in fecal microbiota composition in children may predict overweight. Am J Clin Nutr 2008;87:534–8. [DOI] [PubMed] [Google Scholar]
- 13.Finegold SM, Dowd SE, Gontcharova V, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010;16:444–53. [DOI] [PubMed] [Google Scholar]
- 14.Gosalbes MJ, Llop S, Valles Y, et al. Meconium microbiota types dominated by lactic acid or enteric bacteria are differentially associated with maternal eczema and respiratory problems in infants. Clin Exp Allergy 2013;43:198–211. [DOI] [PubMed] [Google Scholar]
- 15.Jimenez E, Marin ML, Martin R, et al. Is meconium from healthy newborns actually sterile? Res Microbiol 2008;159:187–93. [DOI] [PubMed] [Google Scholar]
- 16.Perez-Munoz ME, Arrieta MC, Ramer-Tait AE, et al. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017;5:48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Aagaard K, Ma J, Antony KM, et al. The placenta harbors a unique microbiome. Sci Transl Med 2014;6:237ra65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Zheng J, Xiao X, Zhang Q, et al. The Placental Microbiome Varies in Association with Low Birth Weight in Full-Term Neonates. Nutrients 2015;7:6924–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Rautava S, Collado MC, Salminen S, et al. Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial. Neonatology 2012;102:178–84. [DOI] [PubMed] [Google Scholar]
- 20.DiGiulio DB, Romero R, Amogan HP, et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One 2008;3:e3056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jimenez E, Fernandez L, Marin ML, et al. Isolation of commensal bacteria from umbilical cord blood of healthy neonates born by cesarean section. Curr Microbiol 2005;51:270–4. [DOI] [PubMed] [Google Scholar]
- 22.Collado MC, Rautava S, Aakko J, et al. Human gut colonisation may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep 2016;6:23129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Arboleya S, Sanchez B, Milani C, et al. Intestinal microbiota development in preterm neonates and effect of perinatal antibiotics. J Pediatr 2015;166:538–44. [DOI] [PubMed] [Google Scholar]
- 24.Patel AL, Mutlu EA, Sun Y, et al. Longitudinal Survey of Microbiota in Hospitalized Preterm VeryLow-Birth-Weight Infants. J Pediatr Gastroenterol Nutr 2016;62:292–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Korpela K, Blakstad EW, Moltu SJ, et al. Intestinal microbiota development and gestational age in preterm neonates. Sci Rep 2018;8:2453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kramer MS, Demissie K, Yang H, et al. The contribution of mild and moderate preterm birth to infant mortality. Fetal and Infant Health Study Group of the Canadian Perinatal Surveillance System. JAMA 2000;284:843–9. [DOI] [PubMed] [Google Scholar]
- 27.Penders J, Thijs C, Vink C, et al. Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics 2006;118:511–21. [DOI] [PubMed] [Google Scholar]
- 28.Pammi M, Cope J, Tarr PI, et al. Intestinal dysbiosis in preterm infants preceding necrotizing enterocolitis: a systematic review and meta-analysis. Microbiome 2017;5:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dobbler PT, Procianoy RS, Mai V, et al. Low Microbial Diversity and Abnormal Microbial Succession Is Associated with Necrotizing Enterocolitis in Preterm Infants. Front Microbiol 2017;8:2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Stewart CJ, Embleton ND, Marrs ECL, et al. Longitudinal development of the gut microbiome and metabolome in preterm neonates with late onset sepsis and healthy controls. Microbiome 2017;5:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Mai V, Torrazza RM, Ukhanova M, et al. Distortions in development of intestinal microbiota associated with late onset sepsis in preterm infants. PLoS One 2013;8:e52876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Backhed F, Roswall J, Peng Y, et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015;17:852. [DOI] [PubMed] [Google Scholar]
- 33.Heikkila MP, Saris PE. Inhibition of Staphylococcus aureus by the commensal bacteria of human milk. J Appl Microbiol 2003;95:471–8. [DOI] [PubMed] [Google Scholar]
- 34.O’Sullivan A, Farver M, Smilowitz JT. The Influence of Early Infant-Feeding Practices on the Intestinal Microbiome and Body Composition in Infants. Nutr Metab Insights 2015;8:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ward RE, Ninonuevo M, Mills DA, et al. In vitro fermentation of breast milk oligosaccharides by Bifidobacterium infantis and Lactobacillus gasseri. Appl Environ Microbiol 2006;72:4497–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Lin AE, Autran CA, Szyszka A, et al. Human milk oligosaccharides inhibit growth of group B Streptococcus. J Biol Chem 2017;292:11243–11249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gonia S, Tuepker M, Heisel T, et al. Human Milk Oligosaccharides Inhibit Candida albicans Invasion of Human Premature Intestinal Epithelial Cells. J Nutr 2015;145:1992–8. [DOI] [PubMed] [Google Scholar]
- 38.Ruiz-Palacios GM, Cervantes LE, Ramos P, et al. Campylobacter jejuni binds intestinal H(O) antigen (Fuc alpha 1, 2Gal beta 1, 4GlcNAc), and fucosyloligosaccharides of human milk inhibit its binding and infection. J Biol Chem 2003;278:14112–20. [DOI] [PubMed] [Google Scholar]
- 39.Praveen P, Jordan F, Priami C, et al. The role of breast-feeding in infant immune system: a systems perspective on the intestinal microbiome. Microbiome 2015;3:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Palmer C, Bik EM, DiGiulio DB, et al. Development of the human infant intestinal microbiota. PLoS Biol 2007;5:e177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Koenig JE, Spor A, Scalfone N, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci U S A 2011;108 Suppl 1:4578–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Martin R, Makino H, Cetinyurek Yavuz A, et al. Early-Life Events, Including Mode of Delivery and Type of Feeding, Siblings and Gender, Shape the Developing Gut Microbiota. PLoS One 2016;11:e0158498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Azad MB, Konya T, Maughan H, et al. Infant gut microbiota and the hygiene hypothesis of allergic disease: impact of household pets and siblings on microbiota composition and diversity. Allergy Asthma Clin Immunol 2013;9:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Laursen MF, Zachariassen G, Bahl MI, et al. Having older siblings is associated with gut microbiota development during early childhood. BMC Microbiol 2015;15:154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Smith MI, Yatsunenko T, Manary MJ, et al. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science 2013;339:548–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Hollister EB, Riehle K, Luna RA, et al. Structure and function of the healthy pre-adolescent pediatric gut microbiome. Microbiome 2015;3:36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ringel-Kulka T, Cheng J, Ringel Y, et al. Intestinal microbiota in healthy U.S. young children and adults--a high throughput microarray analysis. PLoS One 2013;8:e64315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Agans R, Rigsbee L, Kenche H, et al. Distal gut microbiota of adolescent children is different from that of adults. FEMS Microbiol Ecol 2011;77:404–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 2012;150:470–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Migliarini S, Pacini G, Pelosi B, et al. Lack of brain serotonin affects postnatal development and serotonergic neuronal circuitry formation. Mol Psychiatry 2013;18:1106–18. [DOI] [PubMed] [Google Scholar]
- 52.Diaz Heijtz R, Wang S, Anuar F, et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A 2011;108:3047–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Clarke G, Grenham S, Scully P, et al. The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 2013;18:666–73. [DOI] [PubMed] [Google Scholar]
- 54.Desbonnet L, Garrett L, Clarke G, et al. The probiotic Bifidobacteria infantis: An assessment of potential antidepressant properties in the rat. J Psychiatr Res 2008;43:164–74. [DOI] [PubMed] [Google Scholar]
- 55.O’Mahony SM, Clarke G, Borre YE, et al. Serotonin, tryptophan metabolism and the brain-gutmicrobiome axis. Behav Brain Res 2015;277:32–48. [DOI] [PubMed] [Google Scholar]
- 56.Raboni S, Bettati S, Mozzarelli A. Tryptophan synthase: a mine for enzymologists. Cell Mol Life Sci 2009;66:2391–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Yanofsky C RNA-based regulation of genes of tryptophan synthesis and degradation, in bacteria. RNA 2007;13:1141–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Li G, Young KD. Indole production by the tryptophanase TnaA in Escherichia coli is determined by the amount of exogenous tryptophan. Microbiology 2013;159:402–10. [DOI] [PubMed] [Google Scholar]
- 59.Erny D, Hrabe de Angelis AL, Jaitin D, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci 2015;18:965–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization programs the hypothalamicpituitary-adrenal system for stress response in mice. J Physiol 2004;558:263–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.O’Mahony SM, Hyland NP, Dinan TG, et al. Maternal separation as a model of brain-gut axis dysfunction. Psychopharmacology (Berl) 2011;214:71–88. [DOI] [PubMed] [Google Scholar]
- 62.Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A 2011;108:16050–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Rousseaux C, Thuru X, Gelot A, et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med 2007;13:35–7. [DOI] [PubMed] [Google Scholar]
- 64.Rhee SH, Pothoulakis C, Mayer EA. Principles and clinical implications of the brain-gut-enteric microbiota axis. Nat Rev Gastroenterol Hepatol 2009;6:306–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Van Felius ID, Akkermans LM, Bosscha K, et al. Interdigestive small bowel motility and duodenal bacterial overgrowth in experimental acute pancreatitis. Neurogastroenterol Motil 2003;15:26776. [DOI] [PubMed] [Google Scholar]
- 66.Macfarlane S, Dillon JF. Microbial biofilms in the human gastrointestinal tract. J Appl Microbiol 2007;102:1187–96. [DOI] [PubMed] [Google Scholar]
- 67.Soderholm JD, Yates DA, Gareau MG, et al. Neonatal maternal separation predisposes adult rats to colonic barrier dysfunction in response to mild stress. Am J Physiol Gastrointest Liver Physiol 2002;283:G1257–63. [DOI] [PubMed] [Google Scholar]
- 68.Demaude J, Salvador-Cartier C, Fioramonti J, et al. Phenotypic changes in colonocytes following acute stress or activation of mast cells in mice: implications for delayed epithelial barrier dysfunction. Gut 2006;55:655–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Lessa FC, Mu Y, Bamberg WM, et al. Burden of Clostridium difficile infection in the United States. N Engl J Med 2015;372:825–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Chang JY, Antonopoulos DA, Kalra A, et al. Decreased diversity of the fecal Microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis 2008;197:435–8. [DOI] [PubMed] [Google Scholar]
- 71.Weingarden AR, Chen C, Bobr A, et al. Microbiota transplantation restores normal fecal bile acid composition in recurrent Clostridium difficile infection. Am J Physiol Gastrointest Liver Physiol 2014;306:G310–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jernberg C, Lofmark S, Edlund C, et al. Long-term ecological impacts of antibiotic administration on the human intestinal microbiota. ISME J 2007;1:56–66. [DOI] [PubMed] [Google Scholar]
- 73.Lange K, Buerger M, Stallmach A, et al. Effects of Antibiotics on Gut Microbiota. Dig Dis 2016;34:260–8. [DOI] [PubMed] [Google Scholar]
- 74.Deneve C, Delomenie C, Barc MC, et al. Antibiotics involved in Clostridium difficile-associated disease increase colonization factor gene expression. J Med Microbiol 2008;57:732–8. [DOI] [PubMed] [Google Scholar]
- 75.Janarthanan S, Ditah I, Adler DG, et al. Clostridium difficile-associated diarrhea and proton pump inhibitor therapy: a meta-analysis. Am J Gastroenterol 2012;107:1001–10. [DOI] [PubMed] [Google Scholar]
- 76.Jackson MA, Goodrich JK, Maxan ME, et al. Proton pump inhibitors alter the composition of the gut microbiota. Gut 2016;65:749–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Imhann F, Bonder MJ, Vich Vila A, et al. Proton pump inhibitors affect the gut microbiome. Gut 2016;65:740–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Clooney AG, Bernstein CN, Leslie WD, et al. A comparison of the gut microbiome between longterm users and non-users of proton pump inhibitors. Aliment Pharmacol Ther 2016;43:974–84. [DOI] [PubMed] [Google Scholar]
- 79.Freedberg DE, Toussaint NC, Chen SP, et al. Proton Pump Inhibitors Alter Specific Taxa in the Human Gastrointestinal Microbiome: A Crossover Trial. Gastroenterology 2015;149:883–5 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cammarota G, Ianiro G, Gasbarrini A. Fecal microbiota transplantation for the treatment of Clostridium difficile infection: a systematic review. J Clin Gastroenterol 2014;48:693–702. [DOI] [PubMed] [Google Scholar]
- 81.Khanna S Microbiota Replacement Therapies: Innovation in Gastrointestinal Care. Clin Pharmacol Ther 2018;103:102–111. [DOI] [PubMed] [Google Scholar]
- 82.Giel JL, Sorg JA, Sonenshein AL, et al. Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PLoS One 2010;5:e8740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sorg JA, Sonenshein AL. Inhibiting the initiation of Clostridium difficile spore germination using analogs of chenodeoxycholic acid, a bile acid. J Bacteriol 2010;192:4983–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest 2007;117:514–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Edwards LA, Lucas M, Edwards EA, et al. Aberrant response to commensal Bacteroides thetaiotaomicron in Crohn’s disease: an ex vivo human organ culture study. Inflamm Bowel Dis 2011;17:1201–8. [DOI] [PubMed] [Google Scholar]
- 86.Sartor RB. Microbial influences in inflammatory bowel diseases. Gastroenterology 2008;134:577–94. [DOI] [PubMed] [Google Scholar]
- 87.Sartor RB. Therapeutic manipulation of the enteric microflora in inflammatory bowel diseases: antibiotics, probiotics, and prebiotics. Gastroenterology 2004;126:1620–33. [DOI] [PubMed] [Google Scholar]
- 88.Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014;146:1489–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Ohkusa T, Sato N, Ogihara T, et al. Fusobacterium varium localized in the colonic mucosa of patients with ulcerative colitis stimulates species-specific antibody. J Gastroenterol Hepatol 2002;17:849–53. [DOI] [PubMed] [Google Scholar]
- 90.Ohkusa T, Okayasu I, Ogihara T, et al. Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis. Gut 2003;52:79–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Manichanh C, Rigottier-Gois L, Bonnaud E, et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006;55:205–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Dicksved J, Halfvarson J, Rosenquist M, et al. Molecular analysis of the gut microbiota of identical twins with Crohn’s disease. ISME J 2008;2:716–27. [DOI] [PubMed] [Google Scholar]
- 93.Sepehri S, Kotlowski R, Bernstein CN, et al. Microbial diversity of inflamed and noninflamed gut biopsy tissues in inflammatory bowel disease. Inflamm Bowel Dis 2007;13:675–83. [DOI] [PubMed] [Google Scholar]
- 94.Morgan XC, Tickle TL, Sokol H, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol 2012;13:R79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Erickson AR, Cantarel BL, Lamendella R, et al. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS One 2012;7:e49138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Khan KJ, Ullman TA, Ford AC, et al. Antibiotic therapy in inflammatory bowel disease: a systematic review and meta-analysis. Am J Gastroenterol 2011;106:661–73. [DOI] [PubMed] [Google Scholar]
- 97.Kruis W, Fric P, Pokrotnieks J, et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 2004;53:1617–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sood A, Midha V, Makharia GK, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol 2009;7:1202–9, 1209 e1. [DOI] [PubMed] [Google Scholar]
- 99.Tursi A, Brandimarte G, Papa A, et al. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study. Am J Gastroenterol 2010;105:2218–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.van Nood E, Dijkgraaf MG, Keller JJ. Duodenal infusion of feces for recurrent Clostridium difficile. N Engl J Med 2013;368:2145. [DOI] [PubMed] [Google Scholar]
- 101.Rossen NG, Fuentes S, van der Spek MJ, et al. Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. Gastroenterology 2015;149:110–118 e4. [DOI] [PubMed] [Google Scholar]
- 102.Moayyedi P, Surette M, Wolfe M, et al. A randomized, placebo controlled trial of fecal microbiota therapy in active ulcerative colitis. DDW 2014;Abstract 929c. [Google Scholar]
- 103.Moayyedi P, Surette MG, Kim PT, et al. Fecal Microbiota Transplantation Induces Remission in Patients with Active Ulcerative Colitis in a Randomized, Controlled Trial. Gastroenterology 2015. [DOI] [PubMed] [Google Scholar]
- 104.Paramsothy S, Kamm MA, Kaakoush NO, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet 2017;389:1218–1228. [DOI] [PubMed] [Google Scholar]
- 105.Mearin F, Lacy BE, Chang L, et al. Bowel Disorders. Gastroenterology 2016. [DOI] [PubMed] [Google Scholar]
- 106.Simren M, Barbara G, Flint HJ, et al. Intestinal microbiota in functional bowel disorders: a Rome foundation report. Gut 2013;62:159–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Rajilic-Stojanovic M, Biagi E, Heilig HG, et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology 2011;141:1792–801. [DOI] [PubMed] [Google Scholar]
- 108.Kerckhoffs AP, Ben-Amor K, Samsom M, et al. Molecular analysis of faecal and duodenal samples reveals significantly higher prevalence and numbers of Pseudomonas aeruginosa in irritable bowel syndrome. J Med Microbiol 2011;60:236–45. [DOI] [PubMed] [Google Scholar]
- 109.Giamarellos-Bourboulis EJ, Pyleris E, Barbatzas C, et al. Small intestinal bacterial overgrowth is associated with irritable bowel syndrome and is independent of proton pump inhibitor usage. BMC Gastroenterol 2016;16:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Saulnier DM, Riehle K, Mistretta TA, et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology 2011;141:1782–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lee YY, Annamalai C, Rao SSC. Post-Infectious Irritable Bowel Syndrome. Curr Gastroenterol Rep 2017;19:56. [DOI] [PubMed] [Google Scholar]
- 112.Dunlop SP, Jenkins D, Neal KR, et al. Relative importance of enterochromaffin cell hyperplasia, anxiety, and depression in postinfectious IBS. Gastroenterology 2003;125:1651–9. [DOI] [PubMed] [Google Scholar]
- 113.Spiller RC, Jenkins D, Thornley JP, et al. Increased rectal mucosal enteroendocrine cells, T lymphocytes, and increased gut permeability following acute Campylobacter enteritis and in post-dysenteric irritable bowel syndrome. Gut 2000;47:804–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Moayyedi P, Ford AC, Talley NJ, et al. The efficacy of probiotics in the treatment of irritable bowel syndrome: a systematic review. Gut 2010;59:325–32. [DOI] [PubMed] [Google Scholar]
- 115.Clarke G, Cryan JF, Dinan TG, et al. Review article: probiotics for the treatment of irritable bowel syndrome--focus on lactic acid bacteria. Aliment Pharmacol Ther 2012;35:403–13. [DOI] [PubMed] [Google Scholar]
- 116.Brenner DM, Moeller MJ, Chey WD, et al. The utility of probiotics in the treatment of irritable bowel syndrome: a systematic review. Am J Gastroenterol 2009;104:1033–49; quiz 1050. [DOI] [PubMed] [Google Scholar]
- 117.Sanders ME, Guarner F, Guerrant R, et al. An update on the use and investigation of probiotics in health and disease. Gut 2013;62:787–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ortiz-Lucas M, Tobias A, Saz P, et al. Effect of probiotic species on irritable bowel syndrome symptoms: A bring up to date meta-analysis. Rev Esp Enferm Dig 2013;105:19–36. [DOI] [PubMed] [Google Scholar]
- 119.Halmos EP, Power VA, Shepherd SJ, et al. A diet low in FODMAPs reduces symptoms of irritable bowel syndrome. Gastroenterology 2014;146:67–75 e5. [DOI] [PubMed] [Google Scholar]
- 120.Bohn L, Storsrud S, Liljebo T, et al. Diet low in FODMAPs reduces symptoms of irritable bowel syndrome as well as traditional dietary advice: a randomized controlled trial. Gastroenterology 2015;149:1399–1407 e2. [DOI] [PubMed] [Google Scholar]
- 121.Chumpitazi BP, Cope JL, Hollister EB, et al. Randomised clinical trial: gut microbiome biomarkers are associated with clinical response to a low FODMAP diet in children with the irritable bowel syndrome. Aliment Pharmacol Ther 2015;42:418–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Penders J, Thijs C, van den Brandt PA, et al. Gut microbiota composition and development of atopic manifestations in infancy: the KOALA Birth Cohort Study. Gut 2007;56:661–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.van Nimwegen FA, Penders J, Stobberingh EE, et al. Mode and place of delivery, gastrointestinal microbiota, and their influence on asthma and atopy. J Allergy Clin Immunol 2011;128:948–55 e1–3. [DOI] [PubMed] [Google Scholar]
- 124.Herbst T, Sichelstiel A, Schar C, et al. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am J Respir Crit Care Med 2011;184:198–205. [DOI] [PubMed] [Google Scholar]
- 125.Forsythe P, Inman MD, Bienenstock J. Oral treatment with live Lactobacillus reuteri inhibits the allergic airway response in mice. Am J Respir Crit Care Med 2007;175:561–9. [DOI] [PubMed] [Google Scholar]
- 126.Russell SL, Gold MJ, Hartmann M, et al. Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep 2012;13:440–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ng M, Fleming T, Robinson M, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 2014;384:766–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Tun HM, Bridgman SL, Chari R, et al. Roles of Birth Mode and Infant Gut Microbiota in Intergenerational Transmission of Overweight and Obesity From Mother to Offspring. JAMA Pediatr 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Cho I, Yamanishi S, Cox L, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 2012;488:621–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Cox LM, Yamanishi S, Sohn J, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014;158:705–721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Luna RA, Oezguen N, Balderas M, et al. Distinct Microbiome-Neuroimmune Signatures Correlate With Functional Abdominal Pain in Children With Autism Spectrum Disorder. Cell Mol Gastroenterol Hepatol 2017;3:218–230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Finegold SM, Molitoris D, Song Y, et al. Gastrointestinal microflora studies in late-onset autism. Clin Infect Dis 2002;35:S6–S16. [DOI] [PubMed] [Google Scholar]
- 133.Sandler RH, Finegold SM, Bolte ER, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol 2000;15:429–35. [DOI] [PubMed] [Google Scholar]
- 134.Buffington SA, Di Prisco GV, Auchtung TA, et al. Microbial Reconstitution Reverses Maternal Diet-Induced Social and Synaptic Deficits in Offspring. Cell 2016;165:1762–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.de Theije CG, Wopereis H, Ramadan M, et al. Altered gut microbiota and activity in a murine model of autism spectrum disorders. Brain Behav Immun 2014;37:197–206. [DOI] [PubMed] [Google Scholar]
- 136.Collins SM, Surette M, Bercik P. The interplay between the intestinal microbiota and the brain. Nat Rev Microbiol 2012;10:735–42. [DOI] [PubMed] [Google Scholar]