Role of the intestinal microbiota in the pathogenesis of multiple sclerosis. Part 2. Gut microbiota as a predisposition factor for the multiple sclerosis development

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

This part of the review focuses on the proposed involvement of the gut microbiota in the realization of the genetic risk of multiple sclerosis, the formation of the intestinal microbiome in early life, and provides data supporting the hypothesis that aberrant formation of the intestinal microbiota in early life may be a predisposing factor to multiple sclerosis.

Full Text

Introduction

For most immune-mediated diseases, the gut microbiota is assumed to be involved in the realization of genetic risk; the disease develops in genetically predisposed individuals against the background of a certain composition of the gut microbiota. For example, certain risk alleles associated with inflammatory bowel diseases are only manifested when they are activated by the gut microbiome [1, 2]. Given that immunologically and metabolically the gut microbiota is integrated with the host [3] and the number of bacterial genes is hundreds of times greater than the number of host genes [4], its contribution to the genetic risk of diseases appears quite logical.

On the contrary, the gut microbiota is considered a complex polygenic factor influenced by combinations of host genomic loci and environmental factors [5]. While the main emphasis was previously placed on the influence of environmental factors, recently more attention has been paid to the genetic control of the composition of intestinal microbiota. Approximately 2%–8% of the intestinal microbiota is inherited [6], particularly families Christensenellaceae and Methanobacteriaceae [7] and bacterial taxa such as Faecalibacterium, an unclassified genus of families Ruminococcaceae, Coprococcus, Bifidobacterium, Parabacteroides, and Bacteroides [8–13]. In addition, associations of characteristic changes in the intestinal microbiome with disease risk genes in celiac disease, inflammatory bowel disease, rheumatoid arthritis, systemic lupus erythematosus, and others have been identified [1, 2, 14–16].

Among the host candidate genes that can control the composition of the microbiome, immune response genes, genes regulating gut functions, and genes involved in metabolic processes are considered [17], which ensure an effective symbiosis between the host and microbiota and act as genetic risk factors for various diseases.

The following sections will present data on genes related to risk factors of multiple sclerosis (MS) or involved in the pathogenesis of the disease that is thought to be involved in controlling the composition of the gut microbiota.

Intestinal microbiota as a factor of genetic risk realization

Genetic control of intestinal microbiota composition

Over the past decade, research has demonstrated a close link between the host gut microbiota and health, which raises the question of how the gut microbiota is formed and whether host genes play a role in this process.

The influence of host genetic factors on intestinal microbial populations was demonstrated in experiments with reciprocal colonization of germ-free (GF) danio rerio or GF mice with intestinal microbiota from pathogen-free mice and fish species, respectively. It turned out that after 14 days, the taxa characteristic of the recipient species prevailed in the transplanted bacterial community [18].

The role of the genome in the control of the gut microbiome is also indicated by studies of monozygotic and dizygotic twins that have revealed greater similarity of microbial taxa among monozygotic twins than among dizygotic twins and twins compared with unrelated individuals [7, 19–23]. The gut microbiomes of twins differ if they live and eat in different households [7, 21].

How and to what extent the host controls the composition of the intestinal microbiota are currently unknown. It is assumed that the influence on the bacterial community inhabiting the gut is exerted through specific genetic loci controlling individual microbial species or a group of related taxa or through genetic loci exerting a pleiotropic effect on groups of relatively distant microorganisms, promoting the spread of those of them that benefit the host [5]. Host gene mutations can influence their interaction with the compositional and functional diversity of the microbiome, potentially modulating human susceptibility to disease [24].

Genes of innate immunity receptors, toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), are considered the most likely candidate genes because microorganisms are recognized through them and genes involved in TLR- and NLR-mediated signal transduction pathways (Myd88, IRAK, and others) [25, 26].

The activity of TLR2, which recognizes Gram-positive bacteria, correlates with the abundance of Coriobacteriaceae (p_Actinobacteria) and Lactococcus (p_Firmicutes) [5]. An abnormal increase in Enterobacteriaceae (p_Proteobacteria) is observed in mice lacking TLR5, which recognizes flagellin [27]. Moreover, comparative experiments on microbiota transfer in newborn and adult Tlr5-deficient mice show that the neonatal expression of TLR5 affects the microbiota composition throughout life [28].

NOD2 loci are also associated with the abundance of Enterobacteriaceae [2]. In addition, the abundance of the representatives of Bacteroidetes increases in NOD2-/- mice [26, 29, 30], the abundance of Alistipes (f_Rikenellaceae) and Bacteroides (f_Bacteroidaceae) increased at the genus level, and the abundance of Prevotella (f_Prevotellaceae) decreased [30]. In another study in NOD2-/- mice, the abundance of Faecalibacterium prausnitzii decreased in the ileum and cecum [31].

When the adaptor protein MyD88, which mediates signal transduction from all TLRs except TLR3, was deficient, the Firmicutes/Bacteroidetes ratio decreased, and the abundance of Lactobacillaceae, Rikenellaceae, and Porphyromonadaceae increased in mouse intestines [32].

In addition to TLRs and NLRs, other receptors for innate immunity – C-type lectins and RIG-I-like receptors, are barely studied in relation to microbiome control.

Genes encoding antimicrobial peptides can also control intestinal bacterial populations [33]. The adaptive immune system evolved in vertebrates to regulate the abundance of beneficial microorganisms [34]; thus, the genes of the adaptive immune response may be also involved in the genetic control of the diversity of the colonized microbiota, for example, major histocompatibility complex II (MHCII) or human leucocyte antigen (HLA-DR) genes, which are responsible for the presentation of antigens to T cells, and genes controlling the production and secretion of immunoglobulins into the intestinal lumen [30, 35–37].

To identify the genetic factors controlling the composition and functional diversity of the microbiome, microbiome genome-wide association studies (mGWAS) have been used recently. In these studies, many “immune” and “metabolic” host genes contributed to beta diversity (interindividual variability) and variability of the composition and functional composition of the intestinal microbiome. Forty-two host genetic loci that influence the beta diversity of the intestinal microbiome were identified, and the association of polymorphisms in several genes with the composition of the intestinal microbiome was revealed [38]. In particular, polymorphisms of LCT encoding lactase and FUT2 encoding galactoside-2-alpha-L-fucosyltransferase 2 were found to be associated with the abundance and diversity of Bifidobacterium (p_Actionbacteria) [39, 40] and polymorphisms of vitamin D receptor (VDR) with the abundance of Parabacteroides (p_Bacteroidetes). VDR-/- mice had an increased abundance of Parabacteroides [38] and Helicobacter hepaticus (p_Proteobacteria) [41] and decreased abundance Akkermansia muciniphila (p_Verrucomicrobia) [41] compared with normal mice. mGWAS are discussed in detail in reviews [42, 43].

Although the data presented do not provide clear insights into the role of host genetic factors in controlling the composition of the gut microbiome, human genes and bacterial taxa play a role in the risk of the development or pathogenesis of MS [44].

Genome–microbiome interactions as a potential predisposition factor for MS

mGWAS have identified associations of genome–microbiome interactions in many diseases [45, 46], supporting the view that the microbiome is critical to host susceptibility/resistance to disease and, possibly, treatment response. Genome–microbiome associations have been identified for some autoimmune diseases, such as inflammatory bowel disease [2], rheumatoid arthritis [14]. However, no studies have been performed for MS. Nevertheless, evidence reveals the presence of this association in MS, since the genetically determined propensity for inflammatory processes can create a favorable environment for autoreactive T cells to initiate a specific immune response against autoantigens.

This assumption is supported by data [47] that female TNF2 receptor-deficient mice (Tnfr2-/- 2D2) harbor in the fecal microbiome several Bacteroides spp., Bacteroides uniformis, and Parabacteroides spp., which predisposes them to the development of autoimmune demyelination of the central nervous system, compared with male mice with a different microbial background (Akkermansia muciniphila, Sutterella sp., Oscillospira spp., Bacteroides acidifaciens, and Anaeroplasma spp.

For other genes, associations with the intestinal microbiome composition have also been shown. For example, variants of certain loci of VDR [48, 49], which are associated with MS risk, are simultaneously associated with the modulation of the composition and metabolism of the gut microbiome [38], VDR expression protects the host from invasive pathogens and supports gut homeostasis, and gut bacteria activate VDR signal transduction [50]. VDR forms a heterodimeric complex with the retinoid X receptor, which mediates the action of several endogenous and exogenous ligands (vitamin D, secondary bile acids, and fatty acids) [51, 52]. In turn, bile acids act as key VDR ligands and regulators of VDR expression [53]. Interestingly, Parabacteroides, whose abundance varies with genetic VDR polymorphisms [38], contain genes involved in secondary bile acid metabolic pathways [54]. The abundance of Parabacteroides varies in several studies [55–58], as well as impaired bile acid metabolism in MS [59]. These findings indicate that these factors may be interrelated, and genome–microbiome interactions, in this case, VDR polymorphisms, bile acid metabolism, Parabacteroides abundance, and their contribution to the pool and regulation of bile acid metabolism may play significant roles in the pathogenesis of MS.

Table presents some associations of genes that control certain bacterial taxa within the intestinal microbiota and are also risk genes (or involved in the pathogenesis) of MS.

 

Genes that control the composition of the gut microbiome and their association with the risk (pathogenesis) of multiple sclerosis

Гены, контролирующие состав кишечного микробиома, и их ассоциация с риском (патогенезом) рассеянного склероза

Gene

Association of the gene with the abundance of bacterial taxa

Changes in genome-controlled bacterial taxa in MS/EAE

Attitude toward risk (pathogenesis) of MS/EAE

Gene

Of a bacterial taxon

TLR2

(toll-like receptor 2)

TLR2 correlates with f_Coriobacteriaceae’s abundance and g_Lactococcus [5]

↑ f_Coriobacteriaceae [55, 63]

  • TLR2 response in MS [64].
  • TLR2 activation in the CNS contributes to inflammation in ER-PS and progression of EAE [65].
  • In female TLR2-/- mice, the severity of EAE ↓ [66].
  • In TLR2-/- mice, the severity of EAE is similar to that in WT mice [67]
  • The negative effect of f_Coriobacteriaceae has been suggested [55, 63].
  • Coriobacteriales more than double the risk of MS activity in children [68].
  • The administration of Hsp65-producing Lactococcus lactis prevents EAE [69]

TLR5

(toll-like receptor 5)

At TLR5-/- ↑ f_Enterobacteriaceae [27]

  • ↑ f_Enterobacteriaceae [55, 70].
  • ↑ opportunistic species (Citrobacter spp., Acinetobacter spp.) f_Enterobacteriaceae [71].
  • ↑ g_ Acinetobacter [72]
  • TLR5 expression is not increased in EAE in C57BL/6 mice [67].
  • ↑ production of IL-12 and IL-6 by monocytes of patients with MS when stimulated by flagellin, a TLR5 ligand [73]
  • Cross-reactive antibodies specific to Acinetobacter species are elevated in the serum of patients with MS [74, 75].
  • Acinetobacter 3-oxoadipate-CoAtransferase has homologous amino acid sequences with MOG and 4-carboxymuconolactondecarboxylase with BMP [76]

TLR4

(toll-like receptor 4)

At TLR4-/-:

Lachnospiraceae_NK4A136_group;

↓ g_Faecalibacterium;

↓ g_Alistipes;

↓ g_Bifidobacterium [77]

  • Lachnospiraceae_NK4A136 [68, 78, 79].
  • ↓ g_Faecalibacterium [56, 57, 79, 80–83].
  • ↑ g_Alistipes [56].
  • ↓ g_Bifidobacterium [57].
  • ↑ g_Bifidobacterium [55, 80, 84, 85]
  • TLR4-/- mice have ↑ the severity of EAE [86].
  • In male C57Bl/6 TLR4-/- mice, ↑ the severity of EAE [66].
  • TLR4 contributes to the infiltration of Th17 cells in the CNS during EAE [87]
  • Lachnospiraceae_NK4A136 has a protective effect on MS in children [68].
  • g_Faecalibacterium has a protective effect on MS [56, 57, 79, 80–83].
  • ↑ g_Alistipes in the chronic phase of encephalomyelitis associated with demyelination in mice infected with the Theiler virus [88].
  • Bifidobacterium levels correlate with disease severity [89].
  • Bifidobacterium adolescentis promotes Th17 differentiation in the intestine of mice [90].
  • Bifidobacterium adolescentis aggravates autoimmune arthritis in mice [90]

NOD1

(Nucleotide-binding oligomerization domain-containing protein 1)

↑ o_Clostiridiales; ↑ g_Bacteroides;

↑ f_Enterobacteriaceae [91, 92]

  • ↑ o_Clostridiales [93].
  • ↑ c_Clostridia [94, 95].
  • ↑ g_Bacteroides [55, 57].
  • ↑ f_Enterobacteriaceae [55, 70]

NOD1-/- mice are EAE-resistant [96]

  • Clostridium perfringens ε-toxin promotes demyelination [97].
  • A course of administration of Bacteroides fragilis reduces the severity of EAE in mice [98].
  • ↑ f_Enterobacteriaceae has a proinflammatory effect [55, 70]

NOD2

(Nucleotide-binding oligomerization domain-containing protein 2)

  • NOD2 gene polymorphism affects:
  • ↑ f_Enterobactericea [2];
  • ↑ g_Bacteroides [26];
  • ↑ g_Dorea [26];
  • ↑ g_Subdoligranulum [26];
  • Faecalibacterium prausnitzii [99]
  • ↑ f_Enterobacteriaceae [55, 70].
  • ↑ g_Bacteroides [55, 57].
  • ↑ g_Dorea [58, 94, 100].
  • ↓ g_Dorea [83].
  • ↓ g_Subdoligranulum [78].
  • Faecalibacterium prausnitzii [56, 57, 80–83]

NOD2-/- mice are EAE-resistant [96]

  • ↑ f_Enterobacteriaceae has a proinflammatory effect [55, 70].
  • g_Bacteroides has a protective effect by promoting Treg differentiation [98].
  • g_Dorea (f_Lachnospiraceae) produces formic acid, inhibits the growth of Enterobacteriaceae [101], and can have a proinflammatory effect by promoting the production of IFNγ [102].
  • g_Subdoligranulum is associated with MS [103].
  • Faecalibacterium prausnitzii has a protective effect by producing butyrate and stimulating Treg differentiation [56, 57, 80–83]

NLRP12

(NLR [NOD-like receptor] family pyrin domain containing 12)

At NLRP12-/-:

  • ↓ o_Bacteroidales;
  • ↓ o_Clostridiales;
  • ↓ f_Lachnospiraceae;
  • ↑ f_Erysipelotrichaceae [104]
  • ↓ in MS:
  • g_Bacteroides [80, 81, 105];
  • c_Clostridia [80];
  • f_Lachnospiraceae [93, 100]
  • ↑ in MS:
  • f_Erysipelotrichaceae;
  • Erysipelotrichacea_unclas [78];
  • g_Holdemania [94];
  • g_Turicibacter [78];
  • g_Catenibacterium [55]
  • A p.Leu972His mutation in the sixth LRR domain of NRLP12 is associated with familial MS [106].
  • In NLRP12-/- mice, ↑ the severity of EAE [107].
  • In NLRP12-/- mice, EAE with atypical symptoms (ataxia and balance disorder) [107–109]
  • g_Bacteroides has a protective effect by promoting Treg differentiation [98].
  • f_Erysipelotrichaceae (g_Holdemania, g_Turicibacter, g_Catenibacterium, Erysipelotrichaceae_unclas) are increased in MS and are considered harmful in MS [55, 78, 94].
  • c_Clostridia has protective butyrate-producing species [80, 94, 95, 105], but there are also pathogenic ones (С. perfringens) [97].
  • f_Lachnospiraceae can have dual effects, both anti-and proinflammatory [110]

NLRP1

(NLR Family Pyrin Domain Containing 1)

In NLRP1-/- mice:

↑ o_Clostridiales;

↑ f_Lachnospiraceae;

↑ g_Oscillospira;

↑ g_Ruminococcus [111]

  • ↑ f_Lachnospiraceae [55].
  • ↑ g_Blautia [55, 83, 94, 100].
  • ↑ g_Dorea [58, 94, 100].
  • ↑ g_Coprococcus [55, 58, 81].
  • ↑ c_Clostridia [94, 95].
  • ↑ o_Clostridiales [93].
  • ↑ g_Oscillospira [55].
  • ↑ g_Ruminococcus [55, 56, 81, 85]
  • Homozygous mutations (p.Ile601Phe or p.Ser1387Ile) ↑ the risk of MS [112], a homozygous missense variant in NLRP1 (Gly587Ser; p) is associated with familial MS [113].
  • Associations of several heterozygous NLRP1 mutations in patients with MS have been identified [114]
  • ↑ c_Clostridia [94, 95].
  • ↓ f_Lachnospiraceae [100].
  • g_Blautia is presumed to have proinflammatory effects on MS, but there are protective species [55, 82, 93].
  • g_Coprococcus is involved in the regulation of intestinal permeability and suppression of TNFα expression [115].
  • Coprococcus butyrate producers show neuroprotective activity in Parkinson’s disease [116].
  • ↑ g_Oscillospira assumed to have a negative effect [55].
  • Mucin-degrading Ruminococcus increases in inflammatory diseases [117]

NLRP3

(NLR Family Pyrin Domain Containing 3)

At NLRP3-/-:

↓ g_Bacteroides;

↑ g_Prevotella; ↑ g_Desulphovibrio;

↑ g_Rumonococcus;

↑ g_Oscillospira [118]

  • ↓ g_Bacteroides [80, 81, 105].
  • ↑ g_Desulphovibrio [55].
  • ↓ g_Prevotella [80, 100].
  • Prevotella copri [55].
  • Prevotella stercorea [55].
  • ↓ g_Prevotella [81, 100].
  • ↑ g_Ruminococcus [55, 56, 81, 85].
  • ↑ g_Oscillospira [55]
  • NLRP3 activation promotes MS/EAE progression [119, 120].
  • NLRP3 is associated with the response to IFN-β treatment in patients with MS with EAE mice [121].
  • NLRP3-/- mice are EAE-resistant [120].
  • In NLRP3-/- mice, ↓ the severity of EAE [122]
  • Bacteroides produce lipid 654, which regulates the innate immune response [123], and propionate, which has an anti-inflammatory effect [124].
  • ↓ g_Bacteroides are associated with inflammation in the gut [125].
  • The negative role of ↑ g_Desulphovibrio is suggested [55, 63, 126].
  • Prevotella histicola attenuates inflammation and reduces the severity of EAE [127].
  • Prevotella copri aggravates inflammation in arthritis [128].
  • Ruminococcus correlates with TNFα, IL-6, and IL-17 and is associated with decreased vitamin D levels [129]

NLRP6

(NLR Family Pyrin Domain Containing 6)

At NLRP6-/-: ↑ Prevotellaceae_g;

↑ g_Prevotella;

↑ p_TM7;

↓ g_Lactobacillus [130]

  • Prevotella stercorea [51].
  • ↓ g_Lactobacillus [74, 100]
  • NLRP6 is an important regulator of homeostasis in the gut [130].
  • NLRP6 can have a protective effect in chronic inflammation and a detrimental effect in acute inflammation [131]
  • Prevotella stercorea in MS, a proinflammatory role is assumed [55].
  • Prevotella copri is associated with the onset of rheumatoid arthritis [132].
  • TM7 ↑ on the Mediterranean diet [133], which is recommended for MS, TM7 ↓ for cancer [133].
  • Lactobacillus has strain-specific protective effects on EAE and MS [134, 135]

Myd88

(Myeloid differentiation primary response 88)

  • Under Myd88-/-:
  • Firmicutes/Bacteroidetes ratio;
  • ↑ f_Lactobacillaceae;
  • ↑ f_Rikenellaceae;
  • ↑ f_Porphyromonadaceae [32];
  • ↑ g_SFB [136]
  • Reduced Firmicutes/Bacteroidetes ratio.
  • ↑ in MS:
  • g_Lactobacillus [55, 56, 105];
  • f_Rikenellaceae MS/EAE [55, 137];
  • f_Porphyromonadaceae [57].
  • ↓ in MS:
  • ↑ f_Rikenellaceae [93]

MyD88-/- mice have ↓ severity of EAE [66, 67, 86]

  • The administration of a mixture of lactobacilli reduced the severity of EAE [134].
  • Alistipes (f_Rikenellaceae) uses tryptophan and exacerbates EAE by stimulating Th17 responses [138].
  • Porphyromonas gingivalis (f_Porphyromonadaceae) is involved in complement-dependent inflammation [139] and impairs epithelial barrier function [140].
  • The administration of SFB to GF mice led to the development of EAE by inducing Th17 cells [141]

HLA MHCII

  • MHC II class variants in mice and fish are associated with different intestinal microbiota [142].
  • In MHC II-/- mice:

↓ g_Lactobacillus;

↑ g_SFB;

↑ g_Helicobacter [143]

  • ↑ g_Lactobacillus.
  • ↓ g_Lactobacillus.
  • Helicobacter pylori is more common in patients with MS than in healthy individuals [144]
  • The presence of the DRB1*15.01 allele increases three times the risk of MS [145].
  • HLA-DRB1*0301 and HLA-DRB1*1303 alleles ↑ the risk of MS [146, 147].
  • The HLA-A*0201 allele is protective [146]
  • HLA polymorphism modulates the course of EAE and gut microbiota [148].
  • Helicobacter pylori-infected patients have a milder course of MS [149, 150].
  • Helicobacter pylori infection reduces the severity of EAE in mice [151].
  • SFB aggravates EAE [141]

LCT

(Lactase)

  • The LCT gene polymorphism rs4988235 is associated with the abundance of Bifidobacterium longum [39].
  • The SNP at the LCT locus correlates with the abundance of g_Bifidobacterium [17, 152, 153].
  • ↑ abundance of Bifidobacterium longum [39]

In MS ↑ g_Bifidobacterium [80, 84, 85, 154]

The rs4988235 (LCT-13910 C > T) variant associated with higher milk intake is associated with ↓ MS risk [155]

  • Bifidobacterium levels correlate with disease severity [89].
  • Bifidobacterium adolescentis promotes Th17 differentiation in the intestine of mice [90].
  • Bifidobacterium adolescentis aggravates autoimmune arthritis in mice [91]
     
     

FUT2

(Fucosyl-transferase 2)

  • FUT2 variants are related:
  • ↓ g_Faecalibacterium and ↑ p_Proteobacteria [156];
  • Blautia and ↑ Rikenellaceae, Peptostreptococcaceae, Clostridiales and Turicibacter [157];
  • With the abundance of Ruminococcus torques [153]
  • ↑ p_Proteobacteria [158].
  • ↓ p_Proteobacteria [85, 93].
  • Sutterella (p_Proteobacteria) [159].
  • Ruminococcus torques [43]

  • Psuedomonas, Mycoplana, Haemophilus, and Acinetobacter (p_Proteobacteria) contribute to inflammation [92].
  • Transplantation of microbiota from patients with MS with decreased Sutterella into GF mice caused a more severe course of EAE than microbiota transplantation from healthy mice [159].
  • Ruminococcus torques negatively correlate with the abundance of Treg cells in RA [160]

VDR

(vitamin D receptor)

  • VDR gene variants are associated with β-diversity [38].
  • VDR gene polymorphism is associated with Parabacterioides abundance [38].
  • At VDR-/- Parabacteroides [38]

↓ g_Parabacteroides [72, 100, 161, 162]

The ApaI polymorphism of VDR may confer different susceptibility to MS in different populations [49, 163]

  • g_Parabacteroides are involved in the metabolism of phytoestrogens and bile acids, which are VDR ligands and regulate VDR expression [72].
  • g_Parabacteroides stimulate Treg differentiation [72]

PLD1

(Phospholipasa D1)

SNP in the PLD1 gene in humans are associated with Akkermansia muciniphila levels [8]

Akkermansia muciniphila

[57, 72, 137]

  • PLD1 levels ↓ in the plasma of patients with RR-PS [164].
  • In PDL-/- mice the severity of EAE ↓ [165].
  • PLD1 expression in rat CNS at the peak of EAE and ↑ abundance of PLD1-positive cells in EAE lesions [166]
  • Akkermansia muciniphila stimulates the Th1-type immune response [72].
  • Can have both pro- and anti-inflammatory effects [167]

Note: CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; ER-PS, secondary-progressive type of multiple sclerosis; RR-PS, remitting–relapsing type of multiple sclerosis; ↑, increase; ↓, decrease.

 

Note: CNS, central nervous system; EAE, experimental autoimmune encephalomyelitis; ER-PS, secondary-progressive type of multiple sclerosis; RR-PS, remitting–relapsing type of multiple sclerosis; ↑, increase; ↓, decrease.

Characteristically, these genes control the colonization of bacterial taxa whose abundance is altered in patients with MS. On the contrary, the altered composition of the gut microbiome affects the type of immune response that is involved in modifying the gut microbiome [60, 61]. All this presents a combined effect of host genes and gut microbiome in both increasing the predisposition to MS and realizing the risk of MS development. That is, the intestinal microbiome acts as an environmental factor that directly interacts with the host genes to form the disease phenotype and a genetically determined factor that is shaped by the host and interacts with it [62].

As shown in the table, the abundance of the same bacterial taxa can be affected by variations in different genes but with different vectors of changes or differently affect representatives belonging to the same phylum. For example, the abundance of Bacteroidetes changes when many NLRs (NOD1, NOD2, NLRP12, NLRP3, and NLRP6) are knocked out, and the abundance of Bacteroides increases when NOD1 and NOD2 are knocked out but decreases when NLRP3 is knocked out. NLRP3 knockout decreases the abundance of Bacteroides and increases the abundance of Prevotella. TLR4 knockout decreases the abundance of Alistipes (p_Bacteroidetes f_Rikenellaceae), and Myd88 knockout, which mediates signal transduction pathways from TLRs, increases the abundance of representatives of this phylum, Rikenellaceae, and Porphyromonadaceae. The abundance of Parabacteroides (p_Bacteroidetes) increases when VDR is knocked out.

The associations listed in Table 1 do not exhaust all the genome–microbiome interactions relevant to MS; future studies are likely to identify new associations and confirm or refute the significance of those already identified. For example, a recent study in patients with MS found a correlation between BTF3L4, a homolog of BTF3, a regulator of apoptosis, and abundance of Lawsonella (p_Actinobacteria; o_Mycobacteriales; suborder Corynebacterineae) [168]. L. clevelandensis was first isolated from abscesses [169] and found in amyotrophic lateral sclerosis in the nervous tissue [170]. Associations of genes involved in the synthesis and metabolism of B vitamins with the composition of the gut microbiome may be associated with the risk of MS development, especially in children [171].

Finally, the first study evaluating the interaction of the host genome with the intestinal microbiome in a model of experimental autoimmune encephalomyelitis (EAE) was recently published [172]. The influence of the host genome on the susceptibility to EAE and bacterial composition of the gut microbiome before disease onset has been demonstrated. In mice with 29 unique genotypes, the authors identified specific gut bacteria and their metabolic functions associated with lower or higher susceptibility to EAE in mice of several genotypes and showed that short-chain fatty acid metabolism was a key factor, with the ability of the commensal species Lactobacillus reuteri to exacerbate EAE [172]. These results demonstrate the existence of complex interactions between the host genome and gut microbiota that modulate the susceptibility to autoimmune diseases of the central nervous system. Understanding these interactions will provide future opportunities to develop strategies for modulating the gut microbiome and reducing the risk of developing not only MS but also other autoimmune diseases.

We also observed that the background initial microbiota predetermine the course of EAE in animals, and high levels of indigenous Enterococcus spp. is a necessary but not sufficient condition for resistance to disease development [71]. Thus, the reciprocal influence of the host genome on the microbiome composition and the regulatory influence of the microbiota inhabiting the gut on host gene activity may be a risk factor (predisposition) for MS.

Although the host genotype contributes to the formation of the gut microbial community, environmental influences at different periods can alter the microbiota profile [173]. The most sensitive and important period for the action of adverse factors may be the stage of gut microbiota formation.

Disruption of gut microbiota formation as an MS-predisposing factor

Formation of intestinal microbiota

The formation of the intestinal microbial community occurs during the first few years of life, a time that corresponds to the critical period of immune and nervous system development; therefore, the future health of an individual largely depends on which microorganisms will colonize various niches of the body during this period. The colonization of the intestine with microbiota is a dynamic process that occurs in a certain sequence according to the law of succession: facultative anaerobes such as enterobacteria (coliform bacteria) and lactobacilli colonized first, followed by anaerobic species such as bifidobacteria, bacteroides, clostridia, and eubacteria [174–176]. The first microorganisms are transmitted from the mother, a mechanism that ensures the acquisition of the “right” microorganisms necessary for the development of the infant’s body. When exactly this process begins is currently under active research.

For many decades, the fetal gut was believed to be sterile, and the first microorganism-colonize the infant’s body at birth from the mother and environment [177, 178].

Based on these ideas and the determination of microbiota using culturing methods, in 1983, Cooperstock and Zedd [179] distinguished four phases of early colonization of the infant gut: phase I, during the first 2 weeks after birth; phase II, breastfeeding period; phase III, from the beginning of complementary feeding until complete cessation of breastfeeding; and phase IV, until complete complementary feeding, and introduction to adult diet. However, results of sequencing technologies to identify microorganisms show that intestinal microbiome formation can occur in utero due to maternal microorganisms in the placenta and amniotic fluid [180–183]. Although not yet universally accepted and debated, these findings demonstrate the importance of the maternal microbiome for the initial stages of infant gut microbiome formation [184].

Bacteria have been detected in the umbilical cord blood [185], amniotic fluid [186–188], and fetal membranes [188, 189] of healthy women who have given birth without signs of inflammation. For example, Proteobacteria (Ralstonia insidiosa), Firmicutes (Lactobacillus rhamnosus, Lactobacillus crispatus, Lactobacillus iners), and Actinobacteria (Bifidobacterium spp.) were found in the placenta and amniotic fluid [180–183].

How microorganisms enter the placenta and amniotic fluid is uncertain. They may be translocated from the mother’s oral cavity and intestines through the bloodstream or with dendritic cells, which absorb bacteria from the intestinal lumen and transport them throughout the body during migration to lymphoid organs [190, 191]. In particular, a similar mechanism has been described in mice [192]. However, whether these microorganisms establish themselves in the fetal intestine or whether their presence is transient is unclear.

The meconium (first stool after birth) of infants defines a complex community of microbes, although less diverse than adult feces [185, 193]. The presence of typical gastrointestinal representatives such as Enterococcus spp. and Escherichia coli in the meconium [185, 193] indicates that these microorganisms enter the infant’s intestine intrauterus.

Immunological tolerance is required for the successful colonization of the infant’s gut with microorganisms, which is also provided by the mother through the preferential induction of regulatory T lymphocytes [194]. The maternal gut microbiome changes during pregnancy, adapting to the stage of fetal development [195]: In the first trimester, the abundance of Faecalibacterium prausnitzii increases, promoting the formation of T-regulatory cells (Tregs), which provide immunological tolerance to the fetus; in contrast, in the third trimester, the abundance of Enterobacteriaceae, Enterococcaceae, and Streptococcaceae, facultative anaerobic microorganisms that are transmitted to the infant either intrauterine or at birth and dominate the gut microbial community during the first days of its life, increases. Since the infant receives microorganisms at birth when passing through the maternal delivery canal [174, 196–198], the meconium microbiome of newborns is unsurprisingly very similar to the mother’s vaginal microbiome [199]. However, the similarity in the microbial composition of the meconium of children born vaginally and by cesarean section may indicate that these microorganisms may have entered the intestines of the children while they were still in utero.

The second stage of maternal microbiota transmission occurs during breastfeeding [200, 201]. Between 100 and 600 bacterial species belonging to eight genera, namely, Staphylococcus, Streptococcus, Serratia, Pseudomonas, Corynebacterium, Ralstonia, Propionibacterium, Sphingomonas, and the Bradyrhizobiaceae family were found in the breast milk of healthy women [202]. These bacterial taxa constituted approximately half of the entire microbiome and were present in all samples, forming a common part of the microbiome. The remaining part of the microbiome (variable) was represented by microorganisms found in individual breast milk samples in different combinations [202].

During the lactation period, the bacterial composition of breast milk changes. Milk produced immediately after delivery (colostrum) contains more lactic acid bacteria along with staphylococci, streptococci, and lactococci. After 6 months of lactation, the abundance of species colonizing the oral cavity (Veillonella spp., Leptotrichia spp., and Prevotella spp.) increases in breast milk, possibly to prepare the baby for the transition to solid food [203].

In general, the gut microbiota of newborns is characterized by an unstable structure and has a limited set of bacterial taxa, which becomes more complex with increasing food diversity [204, 205]. Thus, the introduction of additional food to breast milk is associated with an increase in the diversity of the bacterial community and a change in the predominant bacterial phyla. At 6 months of age, Bacteroidetes and Firmicutes begin to dominate the human fecal microbiota, and Verrucomicrobia appear; by contrast, Proteobacteria and aerobic Gram-negative bacteria significantly decreased [206].

The composition of the intestinal microbiota stabilizes by the end of the first year of life, acquiring the features of the adult gut microbiome [206–208]. During this period, nutritional and environmental factors become more important in maintaining the diversity of the child’s gut microbiome composition than those of the mother.

Although the gut microbiota in 3-year-old children is already similar to that in adults [175, 206, 209], it reaches its maximum diversity only in adolescence [210].

Intestinal microbiocenosis is unique in each person, and under normal (eubiotic) conditions, it is a complex balanced ecosystem in which microorganisms are in a symbiotic relationship with the host. However, some factors can affect intestinal microbiome formation. These factors include premature birth, cesarean section, artificial feeding, early weaning, antibiotic use, stress, and infections [211, 212].

The phases of intestinal microbiome formation and factors that affect this process are summarized in the figure.

 

Figure. Phases of early infant colonization (modified by M.S. Cooperstock and A.J. Zedd [179]) and factors influencing this process. CP, colonization phase; CS, caesarean section; AF, artificial feeding. * Early colonization phases

Рисунок. Фазы ранней колонизации кишечника младенца (модификация M.S. Cooperstock и A.J. Zedd [179]) и влияющие на этот процесс факторы. ФК — фаза колонизации; КС — кесарево сечение; ИВ — искусственное вскармливание. * Фазы ранней колонизации

 

The aberrant formation of the intestinal microbiome in the early stages of development is thought to result in changes in its functional properties, which can lead to long-term epigenetic changes, metabolic, and immunological dysregulation, structural and functional changes in the immune and nervous systems, and disorders of the intestinal and blood–brain barrier [213, 214] and can further contribute to increased susceptibility to various diseases [215, 216] including MS.

Environmental factors affecting the formation of gut microbiota and their relationship to MS risk

Among environmental factors that have the greatest influence on the composition of the gut microbial community, exposures in early life (maternal factors, mode of birth, gestational age, feeding type, weaning age, and antibiotic use), diet, and micronutrients, stress, and lifestyle (see figure) are discussed.

Relatively few studies have reported the influence of maternal and perinatal factors. In particular, a potential association between gestational diabetes and maternal overweight/obesity with MS before pregnancy was noted [217]. Metabolic disorders are accompanied by changes in gut microbiome composition [218]; diet, antibiotic intake, high glucocorticoid levels due to stress, infectious or immune-mediated exposures during pregnancy, or lactation can also lead to gut microbiome dysbiosis and affect the formation of gut microbiota composition in infants [218]. In turn, impaired gut microbiome formation in early life is associated with an increased risk of autoimmune disease development later in life [219–222].

Emerging evidence shows that the influence of the maternal microbiome on the development of the immune system of offspring begins in utero. In this respect, of interest are the studies that showed the long-term effects of gut bacteria, limited to the gestation period, on the immune profiles of cubs when GF-pregnant females were colonized with a genetically modified strain (HA107) of E. coli, which did not replicate and was eventually eliminated from the gut [223]. The maternal microbiota controls the expression of several genes in the intestinal mucosa, prepares the intestines of calves for postnatal microbial colonization, and affects the innate but not the adaptive link of immunity. Previously, long-term immune changes are believed to be associated with the action of the newborn’s microbiota in the postnatal period [224]. That is, the maternal microbiome forms both the composition of the intestinal microbiome and immune functions in the offspring at an early age.

Although the mechanisms of these effects are currently not fully established, the long-term consequences of adverse perinatal influences on the immune system and increased risk for immune-mediated diseases may be associated with epigenetic changes [225], and embryogenesis is a particularly vulnerable period for DNA modifications [226, 227]. Since the gut microbiota is involved in the epigenetic regulation of Th17/Treg balance [228], the absence or excess of certain microorganisms in the maternal microbiome during this period can lead to the dysregulation of gene expression/activity.

In the EAE model reproducing the pathological processes and symptoms characteristic of MS, a more severe disease course was observed in the adult offspring of mice that were exposed to pathogens during pregnancy. These mice showed increased production of Th17 cells and proinflammatory cytokines, which proves the long-term effect of gestational exposure on immune responses in the offspring [229, 230]. Systemic administration of high doses of lipopolysaccharide to pregnant females led to similar effects [231]. In another study [232], pregnant rats received a mixture of antibiotics added to drink water 2 weeks before delivery and 4 weeks after delivery. This exposure altered the microbial profile in the offspring of antibiotic-treated rats and exacerbated the EAE course when the rats reached 3 months of age. Evidence shows that the administration of antibiotics to adult animals reduces the severity of EAE symptoms [233–235].

A pilot study [236] in children with MS revealed an association between perinatal exposure to pesticides and an increased risk of MS development. Pesticides are thought to have the strongest effect on the development of lymphoid organs during embryogenesis or early childhood [237, 238], which leads to long-term immune dysfunction and may contribute to the early development of MS.

At different gestational periods, the composition of the microbiome (intestinal and vaginal) of the mother changes; therefore, in the case of premature birth, the microbial composition of the mother’s microbiome is not quite ready for transmission, which is confirmed by significant differences in the microbiota of preterm and term-born infants. Thus, premature infants lacked two major bacterial genera in the gut microbiota observed in term-born infants: Bifidobacterium and Lactobacillus, with compensatory dominance of Proteobacteria. In addition, Clostridium difficile, Bacillus, and Staphylococcus bacteria were found [239, 240]. A higher incidence of MS was noted in people born prematurely compared with those born at term [241], although another study [242] did not find such a pattern.

Several studies link the risk of MS development to impaired intestinal microbiocenosis formation due to factors such as cesarean section [243], artificial feeding [244], and exposure to antibiotics in early life [245, 246].

The normal dynamics of bacterial colonization of the gut are disrupted in cesarean births, which can lead to an abnormal immune system [247]. The gut microbial composition of infants born by cesarean section resembles that of the maternal skin microbiota, with the dominance of Staphylococcus, Corynebacterium, and Propionibacterium, whereas that of naturally born babies is dominated by Lactobacillus, Prevotella, and Sneathia [198]. Differences in the structure of the bacterial community in the intestine are noted not only within a few months [198, 248, 249] or a year [174, 248, 249] but persist into the 7th year [250] and even into adulthood [249, 251].

Women who underwent cesarean sections usually receive prophylactic antibiotics before delivery; thus, microbiota disturbances in babies born by cesarean section reflect the cumulative effect of both the altered mode of delivery and antibiotic exposure [252].

Conflicting data present the association of the mode of delivery (vaginally or cesarean section) with the risk of MS development. Some studies have shown that cesarean section may be a risk factor for both early onset (in childhood) of MS [253] and an increased risk for MS [236, 243]. Other studies have found no risk [243, 254] or even observed a reduced risk of childhood MS [236]. A study of children with MS reported significant differences in the type and abundance of bacterial taxa in the gut compared with healthy children. In particular, Christensenellaceae, which is thought to be an inherited bacterial taxon [7], may be relevant to the mode of birth and affect MS risk [55, 63, 93]; however, no study has revealed as to which genes control the abundance of this family.

The feeding type during the neonatal period also affects the gut microbial composition of infants, which is a possible risk factor for MS. Breast milk is the first dietary exposure in infancy, contains various protective compounds including carbohydrates, nucleotides, fatty acids, vitamins, immunoglobulins, cytokines, immune cells, lysozyme, lactoferrin, and other immunomodulatory factors [255, 256], and is a rich source of bacteria including lactic acid bacteria, propionic bacteria, bifidobacteria [257]. Breast milk oligosaccharides stimulate specific intestinal microbiota, block pathogen adhesion sites in the gut, and/or act as analogs of soluble pathogen receptors [258, 259]. Breast milk bacteria – Bifidobacterium, Lactobacillus, and Enterococcus – are an important source of gut bacteria for infants, and they can contribute to the normal development of the immune system by competitive exclusion of pathogenic bacteria, increased production of antimicrobial peptides, and improved function of the gut barrier [257]. The intestinal microbiota of breastfed infants has more Bifidobacteria and Lactobacilli, whereas Bacteroides spp., Clostridium, Streptococcus, Enterobacter, Citrobacter, and Veillonella predominate in formula-fed infants [260–263]. The significance of the presence of Bifidobacterium during this period for MS resistance is confirmed by the fact that the administration of Bifidobacterium animalis to rats during lactation contributed to a milder course of adult-induced EAE [264]. Interestingly, Lactobacillus casei Shirota, also administered during lactation, did not have a protective effect and increased the disease duration [265], which once again emphasizes the importance of the presence of bifidobacteria in this period of ontogenesis.

Evidence shows that breastfeeding reduces MS risk in children and adults [266, 267], whereas reducing the duration of breastfeeding and artificial feeding increases MS risk, including in children [253, 266, 268, 269]. However, Graves et al. [236] have not confirmed the protective effect of breastfeeding on MS development.

After birth, the development of the immune and nervous systems continues for the first 2–3 years of life [270], and the gut microbiota plays an important role in these processes as it stimulates the development of immune responses, which in turn inhibit microbiota growth [271]. Because naïve T cells (Th0) differentiate into subsets of Th1 (supporting cellular immune responses), Th2 (supporting humoral and allergic responses), and Th17 (involved in autoimmune processes) [270], impaired microbiota composition early in life can affect host immune status and can be a risk factor for disease, including MS [272, 273].

Lipopolysaccharides of different bacterial species have different structures and immunogenic properties, and children from areas with a higher prevalence of autoimmune diseases have dominance of bacterial species that produce less immunogenic lipopolysaccharides, which may affect the “training” of the immune system in early life, increasing disease susceptibility [274]. The injection of lipopolysaccharides in the neonatal period (P3 and P5 rats or P15 mice) promoted an easier course of EAE in later life, in contrast to prenatal exposure [231], which was accompanied by increased corticosterone levels [275] or more Tregs [276]. The administration of subpyrogenic doses of the proinflammatory cytokine IL-1β to rats at different periods of early postnatal ontogenesis (at 1, 2, 3, or 4 weeks of life) differently affects the EAE course in adults. Thus, the administration of IL-1β at P1–P7 and P22–P28 worsened the EAE course, and at P8–P14, and P15–P21, it attenuated disease severity compared with the corresponding control group [277]. The neonatal administration of dexamethasone (at P1, P2, and P3) to rats aggravated the EAE course in adult rats [278], as did neonatal stress [279, 280]. Early age stress had a long-term effect on immune functions and exacerbated the EAE course in adult rats [280–282] and mice [283], and more pronounced effects were observed in males than in females.

Although the aforementioned studies did not investigate the composition of the intestinal microbiota, prenatal stress, or increased glucocorticoids during this period affects the composition of the offspring microbiota. Thus, in prenatally stressed animals, the abundance of Lactobacillus was reduced and those of Oscillibacter, Anaerotruncus, and Peptococcus increased [284]. Infants of mothers with high cortisol levels during pregnancy were characterized by a lower abundance of lactic acid bacteria and a higher abundance of Proteobacteria, among which other pathogens are present (Serratia, Citrobacter, and Enterobacter) [285], and their abundance increases in MS [44]. In female rhesus macaque cubs that experienced acoustic stress during pregnancy or separation stress in early life, intestinal colonization with microbiota was impaired [286, 287].

Early antibiotic treatment delays microbiota maturation in infancy [288]. In a population-based study of over 776,000 newborns in Denmark, antibiotic use before and during pregnancy correlated with an increased risk of offspring susceptibility to infection in childhood [289]. The administration of antibiotics during the neonatal period (P7–P23) altered the gut microbiota profile and myelin structures in adult mice [290].

Epidemiological evidence presents that sunlight exposure and vitamin D (VitD) intake during pregnancy and early childhood may influence MS risk [291]. Although VitD deficiency in females during pregnancy resulted in reduced EAE severity in the offspring [292], the effect of gestational VitD deficiency on the EAE course was evident in the second generation of mice, which developed a more severe disease course [293]. The importance of normal VitD levels in early life has been demonstrated in experiments with VitD supplementation in rats from birth to weaning (before puberty) in which EAE severity was reduced in later life [294]. Moreover, VitD supplementation precisely at an early age, and not during pregnancy, or as an adult, contributed to the suppression of EAE [295], which indicates the existence of a critical period for the effects of VitD on MS.

The deficiency of B vitamins (folic acid and cyanocobalamin) during the critical window (1000 days) can affect the maturation of the gut microbiota and its interaction with the host, with consequences on adolescence and adulthood [296]. The supplementation of pregnant mice with methyl donors (betaine, choline, folic acid, and vitamin B12) resulted in significant differences in the composition of the postnatal gut microbiota compared with the microbiota of the offspring of mice without supplementation [297]. The addition of methyl donors (folic acid and vitamin B12) increases the pool of single-carbon fragments and changes the methylation of an individual gene locus in mice [298] and humans [299], affecting epigenetic regulation. Importantly, some microorganisms can synthesize various vitamins; however, the significance of this vitamin pool in the formation of the intestinal microbiota and interactions with the host is currently unknown [300].

Essentially, many factors can influence the formation of the intestinal microbiome and its properties in the prenatal and early postnatal period, forming a predisposition to disease in later life.

Conclusion

MS is a multifactorial disease in which genetic predisposition and environmental factors play a role. To date, how risk factors act during development has not been understood. Importantly, the critical age was 0–6 months, which is not only a period of vulnerability but also the most effective “window” for manipulating the composition of the microbiota to maintain and improve effective immune responses [300] and reduce disease risk. The studies mentioned in the review demonstrate that impaired gut microbiota formation has long-term consequences and can increase susceptibility to MS, and correction of gut microbiota composition early in life can be a strategy to reduce MS risk.

Additional information

Funding sources. The work was supported by the RSF and St. Petersburg Science Foundation grant No. 22-25-20191.

Competing interests. The author declare the absence of obvious and potential conflicts of interest related to the publication of this article.

Authors̕ contribution. The author made a significant contribution to the development of the concept and preparation of the article, read and approved the final version before publication.

Дополнительная информация

Источник финансирования. Работа поддержана грантом РНФ и Санкт-Петербургского научного фонда № 22-25-20191.

Конфликт интересов. Автор заявляет об отсутствии конфликта интересов, связанного с подготовкой и публикацией статьи.

Вклад автора. Автор внесла существенный вклад в разработку концепции и подготовку статьи, прочла и одобрила финальную версию перед публикацией.

List of abbreviations

EAE, experimental autoimmune encephalomyelitis; MS, multiple sclerosis

×

About the authors

Irina N. Abdurasulova

Institute of Experimental Medicine

Author for correspondence.
Email: i_abdurasulova@mail.ru
ORCID iD: 0000-0003-1010-6768
SPIN-code: 5019-3940
Scopus Author ID: 22233604700

Cand. Sci. (Biol.), Head of the Pavlov Department of Physiology

Russian Federation, Sаint Petersburg

References

  1. Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–124. doi: 10.1038/nature11582
  2. Knights D, Silverberg MS, Weersma RK, et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014;6(12):107. doi: 10.1186/s13073-014-0107-1
  3. Brestoff JR, Artis D. Commensal bacteria at the interface of host metabolism and the immune system. Nat Immunol. 2013;14(7):676–684. doi: 10.1038/ni.2640
  4. Grise EA, Serge JA. The human microbiome: our second genome. Annu Rev Genomics Hum Genet. 2012;13:151–170. doi: 10.1146/annurev-genom-090711-163814
  5. Benson AK, Kelly SA, Legge R, et al. Individuality in gut microbiota composition is a complex polygenic trait shaped by multiple environmental and host genetic factors. Proc Natl Acad Sci USA. 2010;107(44):18933–18938. doi: 10.1073/pnas.1007028107
  6. Rothschild D, Weissbrod O, Barkan E, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555(7695):210–215. doi: 10.1038/nature25973
  7. Goodrich JK, Waters JL, Poole AC, et al. Human genetics shape the gut microbiome. Cell. 2014;159(4):789–799. doi: 10.1016/j.cell.2014.09.053
  8. Davenport ER, Cusanovich DA, Michelini K, et al. Genome-wide association studies of the human gut microbiota. PLoS One. 2015;10(11):e0140301. doi: 10.1371/journal.pone.0140301
  9. Goodrich JK, Davenport ER, Waters JL, et al. Cross-species comparisons of host genetic associations with the microbiome. Science. 2016;352(6285):532–535. doi: 10.1126/science.aad9379
  10. Goodrich JK, Davenport ER, Beaumont M, et al. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe. 2016;19(5):731–743. doi: 10.1016/j.chom.2016.04.017
  11. Goodrich JK, Davenport ER, Clark AG, Ley RE. The relationship between the human genome and microbiome comes into view. Annu Rev Genet. 2017;51:413–433. doi: 10.1146/annurev-genet-110711-155532
  12. Turpin W, Espin-Garcia O, Xu W, et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat Genet. 2016;48(11):1413–1417. doi: 10.1038/ng.3693
  13. Lim MY, You HJ, Yoon HS, et al. The effect of heritability and host genetics on the gut microbiota and metabolic syndrome. Gut. 2017;66(6):1031–1038. doi: 10.1136/gutjnl-2015-311326
  14. Wells PM, Williams FMK, Matey-Hernandez ML, et al. RA and the Microbiome: Do host genetic factors provide the link? J Autoimmun. 2019;99:104–115. doi: 10.1016/j.jaut.2019.02.004
  15. He Z, Shao T, Li H, et al. Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus. Gut Pathog. 2016;8:64. doi: 10.1186/s13099-016-0146-9
  16. Kwon Y-C, Chun S, Kim K, Mak A. Update on the genetics of systemic lupus erythematosus: genome-wide association studies and beyond. Cells. 2019;8(10):1180. doi: 10.3390/cells8101180
  17. Blekhman R, Goodrich JK, Huang K, et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015;16(1):191. doi: 10.1186/s13059-015-0759-1
  18. Rawls JF, Mahowald MA, Ley RE, Gordon JI. Reciprocal gut microbiota transplants from zebrafish and mice to germ-free recipients reveal host habitat selection. Cell. 2006;127(2):423–433. doi: 10.1016/j.cell.2006.08.043
  19. Zoetendal EG, Akkermans ADL, Akkermans-van Vliet WM, et al. The host genotype affects the bacterial community in the human gastrointestinal tract. Microb Ecol Health Dis. 2001;13(3):129–134. doi: 10.1080/089106001750462669
  20. Stewart JA, Chadwick VS, Murray A. Investigations into the influence of host genetics on the predominant eubacteria in the faecal microflora of children. J Med Microbiol. 2005;54(Pt 12):1239–1242. doi: 10.1099/jmm.0.46189-0
  21. Xie H, Guo R, Zhong H, et al. Shotgun Metagenomics of 250 Adult Twins Reveals Genetic and Environmental Impacts on the Gut Microbiome. Cell Syst. 2016;3(6):572–584. doi: 10.1016/j.cels.2016.10.004
  22. 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(7):716–727. doi: 10.1038/ismej.2008.37
  23. Turnbaugh PJ, Ridaura VK, Faith JJ, et al. The effect of diet on the human gut microbiome: A metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1(6):6–14. doi: 10.1126/scitranslmed.3000322
  24. Sandoval-Motta S, Aldana M, Martínez-Romero E, Frank A. The human microbiome and the missing heritability problem. Front Genet. 2017;8:80. doi: 10.3389/fgene.2017.00080
  25. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, et al. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell. 2004;118(2):229–241. doi: 10.1016/j.cell.2004.07.002
  26. Petnicki-Ocwieja T, Hrncir T, Liu YJ, et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc Natl Acad Sci USA. 2009;106(37):15813–15818. doi: 10.1073/pnas.0907722106
  27. Carvalho FA, Koren O, Goodrich JK, et al. Transient inability to manage Proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell Host Microbe. 2012;12(2):139–152. doi: 10.1016/j.chom.2012.07.004
  28. Fulde M, Sommer F, Chassaing B, et al. Neonatal selection by Toll-like receptor 5 influences long-term gut microbiota composition. Nature. 2018;560(7719):489–493. doi: 10.1038/s41586-018-0395-5
  29. Rehman A, Sina C, Gavrilova O, et al. Nod2 is essential for temporal development of intestinal microbial communities. Gut. 2011;60(10):1354–1362. doi: 10.1136/gut.2010.216259
  30. Mondot S, Barreau F, Al Nabhani Z, et al. Altered gut microbiota composition in immune-impaired Nod2(-/-) mice. Gut. 2012;61(4):634–635. doi: 10.1136/gutjnl-2011- 300478
  31. Gulati AS, Kruek L, Sartor RB. Influence of NOD 2 on the protective intestinal commensal bacterium Faecalibacterium prausnitzii. Gastroenterology. 2010;138(5):S–14. doi: 10.1016/s0016-5085(10)60064-9
  32. Wen L, Ley RE, Volchkov PY, et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 2008;455(7216):1109–1113. doi: 10.1038/nature07336
  33. Salzman NH, Hung K, Haribhai D, et al. Enteric defensins are essential regulators of intestinal microbial ecology. Nat Immunol. 2010;11(1):76–83. doi: 10.1038/ni.1825
  34. McFall-Ngai M. Adaptive immunity: care for the community. Nature. 2007;445(7124):153. doi: 10.1038/445153a
  35. De Palma G, Capilla A, Nadal I, et al. Interplay between Human Leukocyte Antigen genes and the microbial colonization process of the newborn intestine. Curr Issues Mol Biol. 2010;12(1):1–10. doi: 10.2174/1871528113666140330201056
  36. Vijay-Kumar M, Aitken JD, Carvalho FA, et al. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science. 2010;328(5975):228–231. doi: 10.1126/science.1179721
  37. Shulzhenko N, Morgun A, Hsiao W, et al. Crosstalk between B lymphocytes, microbiota and the intestinal epithelium governs immunity versus metabolism in the gut. Nat Med. 2011;17(12):1585–1593. doi: 10.1038/nm.2505
  38. Wang J, Thingholm LB., Skiecevièienë J. et al. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet. 2016;48(11):1396–1406. doi: 10.1038/ng.3695
  39. Kolde R, Franzosa EA, Rahnavard G, et al. Host genetic variation and its microbiome interactions within the Human Microbiome Project. Genome Med. 2018;10(1):6. doi: 10.1186/s13073-018-0515-8
  40. Wacklin P, Mäkivuokko H, Alakulppi N, et al. Secretor genotype (FUT2 gene) is strongly associated with the composition of Bifidobacteria in the human intestine. PLoS One. 2011;6(5):e20113. doi: 10.1371/journal.pone.0020113
  41. Su D, Nie Y, Zhu A, et al. Vitamin D signaling through induction of paneth cell defensins maintains gut microbiota and improves metabolic disorders and hepatic steatosis in animal models. Front Physiol. 2016;7:498. doi: 10.3389/fphys.2016.00498
  42. Awany D, Allali I, Dalvie S, et al. Host and microbiome genome-wide association studies: current state and challenges. Front Genet. 2019;9:637. doi: 10.3389/fgene.2018.00637
  43. Maglione A, Zuccalà M, Tosi M, et al. Host genetics and gut microbiome: perspectives for multiple sclerosis. Genes (Basel). 2021;12(8):1181. doi: 10.3390/genes12081181
  44. Abdurasulova IN. Role of the intestinal microbiota in the pathogenesis of multiple sclerosis. Part 1. Clinical and experimental evidence for the involvement of the gut microbiota in the development of multiple sclerosis. Medical Academic Journal. 2022;22(2):9–36. (In Russ.) doi: 10.17816/MAJ108241
  45. Hall AB, Tolonen AC, Xavier RJ. Human genetic variation and the gut microbiome in disease. Nat Rev Genet. 2017;18(11):690–699. doi: 10.1038/nrg.2017.63
  46. Imhann F, Vich Vila A, Bonder MJ, et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut. 2018;67(1):108–119. doi: 10.1136/gutjnl-2016-312135
  47. Miller PG, Bonn MB, Franklin CL, et al. TNFR2 deficiency acts in concert with gut microbiota to precipitate spontaneous sex-biased central nervous system demyelinating autoimmune disease. J Immunol. 2015;195(10):4668–4684. doi: 10.4049/jimmunol.1501664
  48. Abdollahzadeh R, Fard MS, Rahmani F, et al. Predisposing role of vitamin D receptor (VDR) polymorphisms in the development of multiple sclerosis: A case-control study. J Neurol Sci. 2016;367:148–151. doi: 10.1016/j.jns.2016.05.053
  49. Imani D, Razi B, Motallebnezhad M, Rezaei R. Association between vitamin D receptor (VDR) polymorphisms and the risk of multiple sclerosis (MS): an updated meta-analysis. BMC Neurol. 2019;19(1):339. doi: 10.1186/s12883-019-1577-y
  50. Bakke D, Sun J. Ancient Nuclear Receptor VDR with new functions: microbiome and inflammation. Inflamm Bowel Dis. 2018;24(6):1149–1154. doi: 10.1093/ibd/izy092
  51. Haussler MR, Haussler CA, Bartik L, et al. Vitamin D receptor: molecular signaling and actions of nutritional ligands in disease prevention. Nutr Rev. 2008;66(Suppl. 2):S98–S112. doi: 10.1111/j.1753-4887.2008.00093.x
  52. Makishima M, Lu TT, Xie W, et al. Vitamin D receptor as an intestinal bile acid sensor. Science. 2002;296(5571):1313–1316. doi: 10.1126/science.1070477
  53. Han S, Li T, Ellis E, et al. A novel bile acid-activated vitamin D receptor signaling in human hepatocytes. Mol Endocrinol. 2010;24(6):1151–1164. doi: 10.1210/me.2009-0482
  54. Wang K, Liao M, Zhou N, et al. Parabacteroides distasonis alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids. Cell Reports. 2019;25:222–235. doi: 10.1016/j.celrep.2018.12.028
  55. Tremlett H, Fadrosh DW, Faruqi AA, et al. Gut microbiota in early pediatric multiple sclerosis: a case-control study. Eur J Neurol. 2016;23(8):1308–1321. doi: 10.1111/ene.13026
  56. Reynders T, Devolder L, Valles-Colomer M, et al. Gut microbiome variation is associated to Multiple Sclerosis phenotypic subtypes. Ann Clin Transl Neurol. 2020;7(4):406–419. doi: 10.1002/acn3.51004
  57. Pellizoni FP, Leite AZ, de Campos Rodrigues N, et al. Detection of dysbiosis and increased intestinal permeability in Brazilian patients with relapsing-remitting multiple sclerosis. Int J Environ Res Public Health. 2021;18(9):4621. doi: 10.3390/ijerph18094621
  58. Oezguen N, Yalcinkaya N, Kücükali CI, et al. Microbiota stratification identifies disease-specific alterations in neuro-Behçet’s disease and multiple sclerosis. Clin Exp Rheumatol. 2019;37 Suppl 121(6):58–66.
  59. Bhargava P, Smith MD, Mische L, et al. Bile acid metabolism is altered in multiple sclerosis and supplementation ameliorates neuroinflammation. J Clin Invest. 2020;130(7):3467–3482. doi: 10.1172/JCI129401
  60. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157(1):121–141. doi: 10.1016/j.cell.2014.03.011
  61. Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167(4):1125–1136. doi: 10.1016/j.cell.2016.10.020
  62. Bevins CL, Salzman NH. The potter’s wheel: the host’s role in sculpting its microbiota. Cell Mol Life Sci. 2011;68(22):3675–3685. doi: 10.1007/s00018-011-0830-3
  63. Kozhieva M, Naumova N, Alikina T, et al. Primary progressive multiple sclerosis in a Russian cohort: relationship with gut bacterial diversity. BMC Microbiol. 2019;19(1):309. doi: 10.1186/s12866-019-1685-2
  64. Fujiwara M, Anstadt EJ, Flynn B, et al. Enhanced TLR2 responses in multiple sclerosis. Clin Exp Immunol. 2018;193(3):313–326. doi: 10.1111/cei.13150
  65. Farez MF, Quintana FJ, Gandi R, et al. Toll-like receptor 2 and poly(ADP-ribose) polymerase 1 promote central nervous system neuroinflammation in progressive EAE. Nat Immunol. 2009;10(9):958–964. doi: 10.1038/ni.1775
  66. Miranda-Hernandez S, Gerlach N, Fletcher JM, et al. Role for MyD88, TLR2 and TLR9 but not TLR1, TLR4 or TLR6 in experimental autoimmune encephalomyelitis. J Immunol. 2011;187(2):791–804. doi: 10.4049/jimmunol.1001992
  67. Prinz M, Garbe F, Schmidt H, et al. Innate immunity mediated by TLR9 modulates pathogenicity in an animal model of multiple sclerosis. J Clin Invest. 2006;116(2):456–464. doi: 10.1172/JCI26078
  68. Horton MK, McCauley K, Fadrosh D, et al. Gut microbiome is associated with multiple sclerosis activity in children. Ann Clin Transl Neurol. 2021;8(9):1867–1883. doi: 10.1002/acn3.51441
  69. Rezende RM, Oliveira RP, Medeiros SR, et al. Hsp65-producing Lactococcus lactis prevents experimental autoimmune encephalomyelitis in mice by inducing CD4+LAP+ regulatory T cells. J Autoimmun. 2013;40:45–57. doi: 10.1016/j.jaut.2012.07.012
  70. Cox LM, Maghzi AH, Liu S, et al. The gut microbiome in progressive multiple sclerosis. Ann Neurol. 2021;89(6):1195–1211. doi: 10.1002/ana.26084
  71. Abdurasulova IN, Tarasova EA, Ermolenko EI, et al. Multiple sclerosis is associated with altered quantitative and qualitative composition of intestinal microbiota. Medical Academic Journal. 2015;15(3):55–67. (In Russ.) doi: 10.17816/MAJ15355-67
  72. Cekanaviciute E, Yoo BB, Runia TF, et al. Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models. Proc Natl Acad Sci USA. 2017;114(40):10713–10718. doi: 10.1073/pnas.1711235114
  73. Chiurchiù V, Leuti A, Cencioni M, et al. Modulation of monocytes by bioactive lipid anandamide in multiple sclerosis involves distinct Toll-like receptors. Pharmacol Res. 2016;113(Pt A):313–319. doi: 10.1016/j.phrs.2016.09.003
  74. Hughes L, Smith P, Bonell S, et al. Cross-reactivity between related sequences found in Acinetobacter sp., Pseudomonas aeruginosa, myelin basic protein and myelin oligodendrocyte glycoprotein in multiple sclerosis. J Neuroimmunol. 2003;144(1–2):105–115. doi: 10.1016/s0165-5728(03)00274-1
  75. Ebringer A, Hughes L, Rashid T, Wilson C. Acinetobacter immune responses in multiple sclerosis etiopathogenetic role and its possible use as a diagnostic marker. Arch Neurol. 2005;62(1):33–36. doi: 10.1001/archneur.62.1.33
  76. Ebringer A, Rashid T, Wilson C. The role of Acinetobacter in the pathogenesis of multiple sclerosis examined by using Popper sequences. Med Hypotheses. 2012;78(6):763–769. doi: 10.1016/j.mehy.2012.02.026
  77. Cuesta CM, Pascual M, Pérez-Moraga R, et al. TLR4 deficiency affects the microbiome and reduces intestinal dysfunctions and inflammation in chronic alcohol-fed mice. Int J Mol Sci. 2021;22(23):12830. doi: 10.3390/ijms222312830
  78. Forbes JD, Chen C-Y, Knox NC, et al. A comparative study of the gut microbiota in immune-mediated inflammatory diseases – does a common dysbiosis exist? Microbiome. 2018;6(1):221. doi: 10.1186/s40168-018-0603-4
  79. Cantoni С, Lin Q, Dorsett Y, et al. Alterations of host-gut microbiome interactions in multiple sclerosis. EBioMedicine. 2022;76:103798. doi: 10.1016/j.ebiom.2021.103798
  80. Miyake S, Kim S, Suda W, et al. Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belondind to Clostridia XIVa and IV clusters. PLoS One. 2015;10(9):e0137429. doi: 10.1371/journal.pone.0137429
  81. Cantarel BL, Waubant E, Chehoud C, et al. Gut microbiota in multiple sclerosis: possible influence of immunomodulators. J Investig Med. 2015;63(5):729–734. doi: 10.1097/JIM.0000000000000192
  82. Storm-Larsen C, Myhr K-M, Farbu E, et al. Gut microbiota composition during a 12-week intervention with delayed-release dimethyl fumarate in multiple sclerosis – a pilot trial. Mult Scler J Exp Transl Clin. 2019;5(4):2055217319888767. doi: 10.1177/2055217319888767
  83. Ling Z, Cheng Y, Yan X, et al. Alterations of the fecal microbiota in Chinese patients with multiple sclerosis. Front Immunol. 2020;11:590783. doi: 10.3389/fimmu.2020.590783
  84. Takewaki D, Suda W, Sato W, et al. Alterations of the gut ecological and functional microenvironment in different stages of multiple sclerosis. PNAS. 2020;117(36):22402–22412. doi: 10.1073/pnas.2011703117
  85. Castillo-Álvarez F, Pérez-Matute P, Oteo JA, Marzo-Sola ME. The influence of interferon β-1b on gut microbiota composition in patients with multiple sclerosis. Neurologia (Engl Ed). 2021;36(7):495–503. doi: 10.1016/j.nrleng.2020.05.006
  86. Marta M, Andersson A, Isaksson M, et al. Unexpected regulatory roles of TLR4 and TLR9 in experimental autoimmune encephalomyelitis. Eur J Immunol. 2008;38(2):565–575. doi: 10.1002/eji.200737187
  87. Zhang Y, Han J, Wu M, et al. Toll-like receptor 4 promotes Th17 lymphocyte infiltration via CCL25/CCR9 in pathogenesis of experimental autoimmune encephalomyelitis. J Neuroimmune Pharmacol. 2019;14(3):493–502. doi: 10.1007/s11481-019-09854-1
  88. Carrillo-Salinas FJ, Mestre L, Mecha M, et al. Gut dysbiosis and neuroimmune responses to brain infection with Theiler’s murine encephalomyelitis virus. Sci Rep. 2017. Vol. 7. P. 44377. doi: 10.1038/srep44377
  89. Abdurasulova IN, Tarasova EA, Matsulevich AV, et al. Influence of Bifidobacteria in the composition of the intestinal microbiota on the multiple sclerosis course. Problems Med Mycol. 2022;24(2):38. (In Russ.)
  90. Tan TG, Sefik E, Geva-Zatorsky N, et al. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc Natl Acad Sci USA. 2016;113(50):E8141–E8150. doi: 10.1073/pnas.1617460113
  91. Bouskra D, Brézillon C, Bérard M, et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nat Lett. 2008;456(7221):507–510. doi: 10.1038/nature07450
  92. Chen GY, Núñez G. Gut Immunity: a NOD to the commensals. Curr Biol. 2009;19(4):R171–174. doi: 10.1016/j.cub.2008.12.027
  93. Galluzzo P, Capri FC, Vecchioni L, et al. Comparison of the intestinal microbiome of Italian patients with multiple sclerosis and their household relatives. Life (Basel). 2021;11(7):620. doi: 10.3390/life11070620
  94. Ventura RE, Iizumi1 T, Battaglia T, et al. Gut microbiome of treatment-naïve MS patients of different ethnicities early in disease course. Sci Rep. 2019;9(1):16396. doi: 10.1038/s41598-019-52894-z
  95. Cekanaviciute E., Pröbstel A.-K., Thomann A. et al. Multiple sclerosis-associated changes in the composition and immune functions of spore-forming bacteria. mSystems. 2018;3(6):e00083–18. doi: 10.1128/mSystems.00083-18
  96. Shaw PJ, Barr MJ, Lukens JR, et al. Signaling via the RIP2 adaptor protein in central nervous system-infiltrating dendritic cells promotes inflammation and autoimmunity. Immunity. 2011;34(1):75–84. doi: 10.1016/j.immuni.2010.12.015
  97. Rumah KR, Linden J, Fischetti VA, Vartanian T. Isolation of clostridium perfringens type B in an individual at first clinical presentation of multiple sclerosis provides clues for environmental triggers of the disease. PLoS One. 2013;8(10):e76359. doi: 10.1371/journal.pone.0076359
  98. Ochoa-Repáraz J, Mielcarz DW, Wang Y, et al. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 2010;3(5):487–495. doi: 10.1038/mi.2010.29
  99. Gulati AS, Kreuk L, Sartor RB. 69 Influence of NOD2 on the protective intestinal commensal bacterium faecalibacterium prausnitzii. Gastroenterology. 2010;138(5):S–14. doi: 10.1016/S0016-5085(10)60064-9
  100. Chen J, Chia N, Kalari KR, et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci Rep. 2016;6:28484. doi: 10.1038/srep28484
  101. Taras D, Simmering R, Collins MD, et al. Reclassification of Eubacterium formicigenerans Holdeman and Moore 1974 as Dorea formicigenerans gen. nov., comb. nov., and description of Dorea longicatena sp. nov., isolated from human faeces. Int J Syst Evol Microbiol. 2002;52(Pt 2):423–428. doi: 10.1099/00207713-52-2-423
  102. Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell. 2016;167(4):1897. doi: 10.1016/j.cell.2016.10.020
  103. Al KF, Craven LJ, Gibbons S, et al. Fecal microbiota transplantation is safe and tolerable in patients with multiple sclerosis: A pilot randomized controlled trial. Mult Scler J Exp Transl Clin. 2022;8(2):20552173221086662. doi: 10.1177/20552173221086662
  104. Chen L, Wilson JE, Koenigsknecht MJ, et al. NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat Immunol. 2017;18(5):541–551. doi: 10.1038/ni.3690
  105. Swidsinski A, Dörffel Y, Loening-Baucke V, et al. Reduced mass and diversity of the colonic microbiome in patients with multiple sclerosis and their improvement with ketogenic diet. Front Microbiol. 2017;8:1141. doi: 10.3389/fmicb.2017.01141
  106. Vilariño-Güell C, Zimprich A, Martinelli-Boneschi F, et al. Exome sequencing in multiple sclerosis families identifies 12 candidate genes and nominates biological pathways for the genesis of disease. PLoS Genet. 2019;15(6):e1008180. doi: 10.1371/journal.pgen.1008180
  107. Gharagozloo M, Mahvelati TM, Imbeault E, et al. The nod-like receptor, Nlrp12, plays an anti-inflammatory role in experimental autoimmune encephalomyelitis. J Neuroinflammation. 2015;12:198. doi: 10.1186/s12974-015-0414-5
  108. Gharagozloo M, Mahmoud S, Simard C, et al. The dual immunoregulatory function of Nlrp12 in T cell-mediated immune response: lessons from experimental autoimmune encephalomyelitis. Cells. 2018;7(9):119. doi: 10.3390/cells7090119
  109. Lukens JR, Gurung P, Shaw PJ, et al. The NLRP12 sensor negatively regulates autoinflammatory disease by modulating interleukin-4 production in T cells. Immunity. 2015;42(4):654–664. doi: 10.1016/j.immuni.2015.03.006
  110. Vacca M, Celano G, Calabrese FM, et al. The controversial role of human gut Lachnospiraceae. Microorganisms. 2020;8(4):573. doi: 10.3390/microorganisms8040573
  111. Tye H, Yu C-H, Simms LA, et al. NLRP1 restricts butyrate producing commensals to exacerbate inflammatory bowel disease. Nat Commun. 2018;9(1):3728. doi: 10.1038/s41467-018-06125-0
  112. Popplewell LF, Encarnacion M, Bernales CQ, et al. Genetic analysis of nucleotide-binding leucine-rich repeat (NLR) receptors in multiple sclerosis. Immunogenetics. 2020;72(6–7):381–385. doi: 10.1007/s00251-020-01170-w
  113. Maver A, Lavtar P, Ristić S, et al. Identification of rare genetic variation of NLRP1 gene in familial multiple sclerosis. Sci Rep. 2017;7(1):3715. doi: 10.1038/s41598-017-03536-9
  114. Bernales CQ, Encarnacion M, Criscuoli MG, et al. Analysis of NOD-like receptor NLRP1 in multiple sclerosis families. Immunogenetics. 2017;70(3):205–207. doi: 10.1007/s00251-017-1034-2
  115. Venkatesh M, Mukherjee S, Wang H, et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity. 2014;41(2):296–310. doi: 10.1016/j.immuni.2014.06.014
  116. Vascellari S, Palmas V, Melis M, et al. Gut microbiota and metabolome alterations associated with Parkinson’s disease. mSystems. 2020;15(5):e00561–20. doi: 10.1128/mSystems.00561-20
  117. Bell A, Brunt J, Crost E, et al. Elucidation of a sialic macid metabolism pathway in mucus-foraging Ruminococcus gnavus unravels mechanisms of bacterial adaptation to the gut. Nat Microbiol. 2019;4(12):2393–2404. doi: 10.1038/s41564-019-0590-7
  118. Zhang Y, Huang R, Cheng M, et al. Gut microbiota from NLRP3-deficient mice ameliorates depressive-like behaviors by regulating astrocyte dysfunction via circHIPK2. Microbiome. 2019;7(1):116. doi: 10.1186/s40168-019-0733-3
  119. Jha S, Srivastava SY, Brickey WJ, et al. The inflammasome sensor, NLRP3, regulates CNS inflammation and demyelination via caspase-1 and interleukin-18. J Neurosci. 2010;30(47):15811–15820. doi: 10.1523/JNEUROSCI.4088-10.2010
  120. Inoue M, Williams KL, Gunn MD, Shinohara ML. NLRP3 inflammasome induces chemotactic immune cell migration to the CNS in experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2012;109(26):10480–10485. doi: 10.1073/pnas.1201836109
  121. Malhotra S, Rio J, Urcelay E, et al. NLRP3 inflammasome is associated with the response to IFN-β in patients with multiple sclerosis. Brain. 2015;138(3):644–652. doi: 10.1093/brain/awu388
  122. Gris D, Ye Z, Iocca HA, et al. NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses. J Immunol. 2010;185(2):974–981. doi: 10.4049/jimmunol.0904145
  123. Farrokhi V, Nemati R, Nichols FC. Bacterial lipodipeptide, Lipid 654, is a microbiome-associated biomarker for multiple sclerosis. Clin Transl Immunol. 2013;2(11):e8. doi: 10.1038/cti.2013.11
  124. Haghikia A, Jörg S, Duscha A, et al. Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine. Immunity. 2015;43(4):817–829. doi: 10.1016/j.immuni.2015.09.007
  125. Nomura K, Ishikawa D, Okahara K, et al. Bacteroidetes species are correlated with disease activity in ulcerative colitis. J Clin Med. 2021;10(8):1749. doi: 10.3390/jcm10081749
  126. Elgendy SG, Abd-Elhameed R, Daef E, et al. Gut microbiota in forty cases of egyptian relapsing remitting multiple sclerosis. Iran J Microbiol. 2021;13(5):632–641. doi: 10.18502/ijm.v13i5.7428
  127. Shahi SK, Jensen SN, Murra AC, et al. Human commensal Prevotella histicola ameliorates disease as effectively as interferon-beta in the experimental autoimmune encephalomyelitis. Front Immunol. 2020;11:578648. doi: 10.3389/fimmu.2020.578648
  128. Scher JU, Sczesnak A, Longman RS, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202. doi: 10.7554/eLife.01202
  129. Ghaly S, Kaakoush NO, Lloyd F, et al. Ultraviolet irradiation of skin alters the faecal microbiome independently of vitamin D in mice. Nutrients. 2018;10(8):1069. doi: 10.3390/nu10081069
  130. Elinav E, Strowig T, Kau AL, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell. 2011;145(5):745–757. doi: 10.1016/j.cell.2011.04.022
  131. Ratsimandresy RA, Dorfleutner A, Stehlik C. An update on PYRIN domain-containing pattern recognition receptors: from immunity to pathology. Front Immunol. 2013;4(440):153–171. doi: 10.3389/fimmu.2013.00440
  132. Bernard NJ. Rheumatoid arthritis: Prevotella copri associated with new-onset untreated RA. Nat Rev Rheumatol. 2014;10(1):2. doi: 10.1038/nrrheum.2013.187
  133. Illescas O, Rodriguez-Sosa M, Gariboldi M. Mediterranean diet to prevent the development of colon diseases: a meta-analysis of gut microbiota studies. Nutrients. 2021;13(7):2234. doi: 10.3390/nu13072234
  134. Lavasani S, Dzhambazov B, Nouri M, et al. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One. 2010;5(2):e9009. doi: 10.1371/journal.pone.0009009
  135. Yamashita M., Ukibe K., Matsubara Y. et al. Lactobacillus helveticus SBT2171 attenuates experimental autoimmune encephalomyelitis in mice. Front Microbiol. 2018;8:2596. doi: 10.3389/fmicb.2017.02596
  136. Larsson E, Tremaroli V, Lee YS, et al. Analysis of gut microbial regulation of host gene expression along the length of the gut and regulation of gut microbial ecology through MyD88. Gut. 2012;61(8):1124–1131. doi: 10.1136/gutjnl-2011-301104
  137. Gandy K, Zhang J, Nagarkatti P, Nagarkatti M. The role of gut microbiota in shaping the relapse-remitting and chronic-progressive forms of multiple sclerosis in mouse models. Sci Rep. 2019;9(1):6923. doi: 10.1038/s41598-019-43356-7
  138. Lin X, Singh A, Shan X, et al. Akkermansia muciniphila-mediated degradation of host mucin expands the tryptophan utilizer alistipes and exacerbates autoimmunity by promoting Th17 immune responses. Cell Press. 2022. doi: 10.2139/ssrn.4065073
  139. Olsen I, Lambris JD, Hajishengallis G. Porphyromonas gingivalis disturbs host–commensal homeostasis by changing complement function. J Oral Microbiol. 2017;9(1):1340085. doi: 10.1080/20002297.2017.1340085
  140. Amano A. Disruption of epithelial barrier and impairment of cellular function by Porphyromonas gingivalis. Front Biosci. 2007;12:3965–3974. doi: 10.2741/2363
  141. Lee Y-K, Menezes JS, Umesaki Y, Mazmanian SK. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4615–4622. doi: 10.1073/pnas.1000082107
  142. Toivanen P, Vaahtovuo J, Eerola E. Influence of major histocompatibility complex on bacterial composition of fecal flora. Infect Immun. 2001;69(4):2372–2377. doi: 10.1128/IAI.69.4.2372-2377.2001
  143. Kubinak JL, Zac Stephens W, Soto R, et al. MHC variation sculpts individualized microbial communities that control susceptibility to enteric infection. Nat Commun. 2015;6:8642. doi: 10.1038/ncomms9642
  144. Gavalas E, Kountouras J, Boziki M, et al. Relationship between Helicobacter pylori infection and multiple sclerosis. Ann Gastroenterol. 2015;28(3):353–356.
  145. Lincoln MR, Montpetit A, Cader MZ, et al. A predominant role for the HLA class II region in the association of the MHC region with multiple sclerosis. Nat Genet. 2005;37(10):1108–1112. doi: 10.1038/ng1647
  146. Goris A, Pauwels I, Dubois B. Progress in multiple sclerosis genetics. Curr Genom. 2012;13(8):646–663. DOI: 10.2174/ 138920212803759695
  147. Alcina A, Abad-Grau Mdel M, Fedetz M, et al. Multiple sclerosis risk variant HLA-DRB1*1501 associates with high expression of DRB1 gene in different human populations. PLoS One. 2012;7(1):e29819. doi: 10.1371/journal.pone.0029819
  148. Shahi SK, Soham A, Jaime CM, et al. HLA class II polymorphisms modulate gut microbiota and EAE phenotype. Immunohorizons. 2022;5(8):627–646. doi: 10.4049/immunohorizons.2100024
  149. Li W, Minohara M, Su JJ, et al. Helicobacter pylori infection is a potential protective factor against conventional multiple sclerosis in the Japanese population. J Neuroimmunol. 2007;184(1–2):227–231. doi: 10.1016/j.jneuroim.2006.12.010
  150. Pedrini MJ, Seewann A, Bennett K.A. et al. Helicobacter pylori infection as a protective factor against multiple sclerosis risk in females. J Neurol Neurosurg Psychiatry. 2015;86(6):603–607. doi: 10.1136/jnnp-2014-309495
  151. Cook KW, Crooks J, Hussain K, et al. Helicobacter pylori infection reduces disease severity in an experimental model of multiple sclerosis. Front Microbiol. 2015;6:52. doi: 10.3389/fmicb.2015.00052
  152. Bonder MJ, Kurilshikov A, Tigchelaar EF, et al. The effect of host genetics on the gut microbiome. Nat Genet. 2016;48(11):1407–1412. doi: 10.1038/ng.3663
  153. Kurilshikov A, Medina-Gomez C, Bacigalupe R, et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat Genet. 2021;53(2):156–165. doi: 10.1038/s41588-020-00763-1
  154. Abdurasulova IN, Tarasova EA, Kudryavtsev IV, et al. Intestinal microbiota composition and peripheral blood Th cell subsets in patients with multiple sclerosis. Infect. Immun. 2019;9(3-4):504–522. (In Russ.) doi: 10.15789/2220-7619-2019-3-4-504-522
  155. Zhang Z, Wang M, Yuan S, et al. Genetically predicted milk intake and risk of neurodegenerative diseases. Nutrients. 2021;13(8):2893. doi: 10.3390/nu13082893
  156. Hall AB, Tolonen AC, Xavier RJ. Human genetic variation and the gut microbiome in disease. Nat Rev Genet. 2017;18(11):690–699. doi: 10.1038/nrg.2017.63
  157. Gampa A, Engen PA, Shobar R, Mutli EA. Relationships between gastrointestinal microbiota and blood group antigens. Physiol Genomics. 2017;49(9):473–483. doi: 10.1152/physiolgenomics.00043.2017
  158. Cosorich I, Dalla-Costa G, Sorini C. et al. High frequency of intestinal TH17 cells correlates with microbiota alterations and disease activity in multiple sclerosis. Sci Adv. 2017;3(7):e1700492. doi: 10.1126/sciadv.1700492
  159. Berer K, Gerdes LA, Cekanaviciute E, et al. Gut Microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalitis in mice. Proc Natl Acad Sci USA. 2017;114(40):10719–10724. doi: 10.1073/pnas.1711233114
  160. Wu R, An J, Ding T et al. The level of peripheral regulatory T cells is associated with the changes of intestinal microbiota in patients with rheumatoid arthritis. Ann Rheumatic Dis. 2021;80(Suppl 1):427. POS0396. doi: 10.1136/annrheumdis-2021-eular.2783
  161. Shahi SK, Freedman SN, Mangalam AK. Gut microbiome in multiple sclerosis: The players involved and the roles they play. Gut Microbes. 2017;8(6):607–615. doi: 10.1080/19490976.2017.1349041
  162. Saresella M, Marventano I, Barone M, et al. Alterations in circulating fatty acid are associated with gut microbiota dysbiosis and inflammation in multiple sclerosis. Front Immunol. 2020;11:1390. doi: 10.3389/fimmu.2020.01390
  163. Zhang Y-J, Zhang L, Chen S-Y, et al. Association between VDR polymorphisms and multiple sclerosis: systematic review and updated meta-analysis of case-control studies. Neurol Sci. 2018;39(2):225–234. doi: 10.1007/s10072-017-3175-3
  164. Eftekharian MM, Azimi T, Ghafouri-Fard S, et al. Phospholipase D1 expression analysis in relapsing-remitting multiple sclerosis patients. Neurol Sci. 2017;38(5):865–872. doi: 10.1007/s10072-017-2857-1
  165. Göbel K, Schuhmann MK, Pankratz S, et al. Phospholipase D1 mediates lymphocyte adhesion and migration in experimental autoimmune encephalomyelitis. Eur J Immunol. 2014;44(8):2295–2305. doi: 10.1002/eji.201344107
  166. Ahn M, Min DS, Kang J, et al. Increased expression of phospholipase D1 in the spinal cords of rats with experimental autoimmune encephalomyelitis. Neurosci Lett. 2001;316(2):95–98. doi: 10.1016/s0304-3940(01)02383-7
  167. Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol. 2004;54:1469–1476. doi: 10.1099/ijs.0.02873-0
  168. Levi I, Gurevich M, Perlman G, et al. Potential role of indolelactate and butirate in multiple sclerosis revealed by integrated microbiome-metabolome analysis. Cell Rep Med. 2021;2(4):100246. doi: 10.1016/j.xcrm.2021.100246
  169. Bell ME, Bernard KA, Harrington SM, et al. Lawsonella clevelandensis gen. nov., sp. nov., a new member of the suborder Corynebacterineae isolated from human abscesses. Int J Evol Microbiol. 2016;66(8):2929–2935. doi: 10.1099/ijsem.0.001122
  170. Alonso R, Pisa D, Carrasco K. Searching for bacteria in neural tissue from amyotrophic lateral sclerosis. Front Neurosci. 2019;13:171. doi: 10.3389/fnins.2019.00171
  171. Abdurasulova IN, Dmitriev AV. Group B vitamins: From homeostasis to pathogenesis and treatment of multiple sclerosis Adv Physiol Sci. 2023;54(1). (In Russ.) doi: 10.31857/S0301179823010034
  172. Montgomery TL, Künstner A, Kennedy JJ et al. Interactions between host genetics and gut microbiota determine susceptibility to CNS autoimmunity. Proc Natl Acad Sci USA. 2020;117(44):27516–27527. doi: 10.1073/pnas.2002817117
  173. Rodríguez JM, Murphy K, Stranton CS, et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Health Dis. 2015;26(1):26050. doi: 10.3402/mehd.v26.26050
  174. Bäckhed 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(5):690–703. doi: 10.1016/j.chom.2015.04.004
  175. Köenig JE, Spor A, Scalfone N, et al. Succession of microbial consortia in the developing infant gut microbiom. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4578–4585. doi: 10.1073/pnas.1000081107
  176. La Rosa PS, Warner BB, Zhou Y, et al. Patterned progression of bacterial populations in the premature infant gut. Proc Natl Acad Sci USA. 2014;111(34):12522–12527. doi: 10.1073/pnas.1409497111
  177. Falk PG, Hooper LV, Midtverd T, Gordon JI. Creating and maintaining the gastrointestinal ecosystem: what we know and need to know from gnotobiology. Microbiol Mol Biol Rev. 1998;62(4):1157–1170. doi: 10.1128/MMBR.62.4.1157-1170.1998
  178. Perez-Muñoz ME, Arrieta M-C, Ramer-Tait AE, Walter J. A critical assessment of the sterile womb and in utero colonization hypotheses: implications for research on the pioneer infant microbiome. Microbiome. 2017;5(1):48. doi: 10.1186/s40168-017-0268-4
  179. Cooperstock MSZ, Zedd AJ. Intestinal flora of infants. In: Human intestinal microflora in health and disease. Ed. by D.J. Hentges. 1983. Chapter 4. P. 79–99. doi: 10.1016/B978-0-12-341280-5.50010-0
  180. Aagaard K, Ma J, Antony KM, et al. The placenta harbors a unique microbiome. Sci Transl Med. 2014;6(237):237ra65. doi: 10.1126/scitranslmed.3008599
  181. Collado MC, Rautava S, Aakko J, et al. Human gut colonization may be initiated in utero by distinct microbial communities in the placenta and amniotic fluid. Sci Rep. 2016;6:23129. doi: 10.1038/srep23129
  182. Satokari R, Gronroos T, Laitinen K, et al. Bifidobacterium and Lactobacillus DNA in the human placenta. Lett Appl Microbiol. 2009;48(1):8–12. doi: 10.1111/j.1472-765X.2008.02475.x
  183. Parnell LA, Briggs CM, Cao B, et al. Microbial communities in placentas from term normal pregnancy exhibit spatially variable profiles. Sci Rep. 2017;7(1):11200. doi: 10.1038/s41598-017-11514-4
  184. Mueller NT, Bakacs E, Combellick J, et al. The infant microbiome development: Mom matters. Trends Mol Med. 2015;21(2):109–117. doi: 10.1016/j.molmed.2014.12.002
  185. 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(4):270–274. doi: 10.1007/s00284-005-0020-3
  186. Bearfield C, Davenport ES, Sivapathasundaram V, Allaker RP. Possible association between amniotic fluid microorganism infection and microflora in the mouth. BJOG. 2002;109(5):527–533. doi: 10.1111/j.1471-0528.2002.01349.x
  187. DiGiulio DB. Diversity of microbes in amniotic fluid. Semin Fetal Neonatal Med. 2012;17(1):2–11. doi: 10.1016/j.siny.2011.10.001
  188. Rautava S, Collado MC, Salminen S, Isolauri E. Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial. Neonatology. 2012;102(3):178–184. doi: 10.1159/000339182
  189. Steel JH, Malatos S, Kennea N, et al. Bacteria and inflammatory cells in fetal membranes do not always cause preterm labor. Pediatr Res. 2005;57(3):404–411. doi: 10.1203/01.PDR.0000153869.96337.90
  190. Vazquez-Torres A, Jones-Carson J, Baumler AJ et al. Extraintestinal dissemination of Salmonella by CD18-expressing phagocytes. Nature. 1999;401(6755):804–808. doi: 10.1038/44593
  191. Rescigno M, Rotta G, Valzasina B, Ricciardi-Castagnoli P. Dendritic cells shuttle microbes across gut epithelial monolayers. Immunobiology. 2001;204(5):572–581. doi: 10.1078/0171-2985-00094
  192. Perez PF, Dore J, Leclerc M, et al. Bacterial imprinting of the neonatal immune system: lessons from maternal cells? Pediatrics. 2007;119(3):e724–e732. doi: 10.1542/peds.2006-1649
  193. Gosalbes MJ, Abellan JJ, Durbán A, et al. Metagenomics of human microbiome: beyond 16s rDNA. Clin Microbiol Infect. 2012;18 Suppl(4):47–49. doi: 10.1111/j.1469-0691.2012.03865.x
  194. Mold JE, Michaëlsson J, Burt TD, et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science. 2008;322(5907):1562–1565. doi: 10.1126/science.1164511
  195. Koren O, Goodrich JK, Cullender TC, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150(3):470–480. doi: 10.1016/j.cell.2012.07.008
  196. Donnet-Hughes A, Perez PF, Doré J, et al. Potential role of the intestinal microbiota of the mother in neonatal immune education. Proc Nutr Soc. 2010;69(3):407–415. doi: 10.1017/S0029665110001898
  197. Collado MC, Laitinen K, Salminen S, Isolauri E. Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr Res. 2012;72(1):77–85. doi: 10.1038/pr.2012.42
  198. Matamoros S, Gras-Leguen C, Le Vacon F, et al. Development of intestinal microbiota in infants and its impact on health.Trends Microbiol. 2013;21(4):167–173. doi: 10.1016/j.tim.2012. 12.001
  199. Dominguez-Bello MG, Costello EK, Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc Natl Acad Sci USA. 2010;107(26):11971–11975. doi: 10.1073/pnas.1002601107
  200. Fernandez L, Langa S, Martin V, et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol Res. 2013;69:1–10. doi: 10.1016/j.phrs.2012.09.001
  201. Sanz Y. Gut microbiota and probiotics in maternal and infant health. Am J Clin Nutr. 2011;94(Suppl 6):2000S–2005S. doi: 10.3945/ajcn.110.001172
  202. Hunt KM, Foster JA, Forney LJ, et al. Characterization of the diversity and temporal stability of bacterial communities in human milk. PLoS One. 2011;6(6):e21313. doi: 10.1371/journal.pone.0021313
  203. Cabrera-Rubio R, Collado MC, Laitinen K, et al. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am J Clin Nutr. 2012;96(3):544–551. doi: 10.3945/ajcn.112.037382
  204. Hyman RW, Fukushima M, Diamond L, et al. Microbes on the human vaginal epithelium. Proc Natl Acad Sci USA. 2005;102(22):7952–7957. doi: 10.1073/pnas.0503236102
  205. Zhou X, Brown CJ, Abdo Z, et al. Differences in the composition of vaginal microbial communities found in healthy Caucasian and black women. ISME J. 2007;1(2):121–133. doi: 10.1038/ismej.2007.12
  206. Palmer C, Bik EM, Di Giulio DB, et al. Development of the human infant intestinal microbiota. PLoS Biol. 2007;5(7):e177. doi: 10.1371/journal.pbio.0050177
  207. Vael C, Desager K. The importance of the development of the intestinal microbiota in infancy. Curr Opin Pediatr. 2009;21(6):794–800. doi: 10.1097/MOP.0b013e328332351b
  208. Quigley EMM. Gut bacteria in health and disease. Gastroenterol Hepatol (NY). 2013;9(9):560–569.
  209. Mackie RI., Sghir A, Gaskins HR. Developmental microbial ecology of the neonatal gastrointestinal tract. Am J Clin Nutr. 1999;69(5):1035S–1045S. doi: 10.1093/ajcn/69.5.1035s
  210. O’Toole PW, Claesson MJ. Gut microbiota: changes throughout the lifespan from infancy to elderly. Int Dairy J. 2010;20(4):281–291. doi: 10.1016/j.idairyj.2009.11.010
  211. Balmer SE, Hanvey LS, Wharton BA. Diet and faecal flora in the newborn: nucleotides. Arch Dis Child Fetal Neonatal Ed. 1994;70(2):F137–F140. doi: 10.1136/fn.70.2.f137
  212. Bennet R, Nord CE. Development of the faecal anaerobic microflora after caesarean section and treatment with antibiotics in newborn infants. Infection. 1987;15(5):332–336. doi: 10.1007/bf01647733
  213. Ruiz VE, Battaglia T, Kurtz ZD, et al. A single early-in-life macrolide course has lasting effects on murine microbial network topology and immunity. Nat Commun. 2017;8:518. doi: 10.1038/s41467-017-00531-6
  214. Lynn MA, Tumes DJ, Choo JM et al. Early-life antibiotic-driven dysbiosis leads to dysregulated vaccine immune responses in mice. Cell Host Microbe. 2018;23:653–660.e5. doi: 10.1016/j.chom.2018.04.009
  215. Dinan TG, Cryan JF. Gut instincts: Microbiota as a key regulator of brain development, ageing and neurodegeneration. J Physiol. 2017;595(2):489–503. doi: 10.1113/JP273106
  216. Korpela K, Salonen A, Virta LJ, et al. Intestinal microbiome is related to lifetime antibiotic use in finnish pre-school children. Nat Commun. 2016;7:10410. doi: 10.1038/ncomms10410
  217. Maghzi AH, Ghazavi H, Ahsan M, et al. Increasing female preponderance of multiple sclerosis in Isfahan, Iran: a population-based study. Mult Scler. 2010;16(3):359–361. doi: 10.1177/1352458509358092
  218. Di Giulio 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(8):e3056. doi: 10.1371/journal.pone.0003056
  219. Wahlberg J, Fredriksson J, Nikolic E, et al. Environmental factors related to the induction of beta cell autoantibodies in 1-yr-old healthy children. Pediatr Diabetes. 2005;6(4):199–205. doi: 10.1111/j.1399-543X.2005.00129.x
  220. Beijers R, Jansen J, Riksen-Walraven M, de Weerth C. Maternal prenatal anxiety and stress predict infant illnesses and health complaints. Pediatrics. 2010;12(2):e401–e409. doi: 10.1542/peds.2009-3226
  221. Aoyama K, Seaward PG, Lapinsky SE. Fetal outcome in the critically ill pregnant woman. Crit Care. 2014;18(3):307. doi: 10.1186/cc13895
  222. Mor G, Cardenas I. The immune system in pregnancy: a unique complexity. Am J Reprod Immunol. 2010;63(6):425–433. doi: 10.1111/j.1600-0897.2010.00836.x
  223. Gomes de Agüero M, Ganal-Vonarburg SC, Fuhrer T, et al. The maternal microbiota drives early postnatal innate immune development. Science. 2016;361(6279):1296–1302. doi: 10.1126/science.aad2571
  224. Kabat AM, Srinivasan N, Maloy KJ. Modulation of immune development and function by intestinal microbiota. Trends Immunol. 2014;35(11):507–517. doi: 10.1016/j.it.2014.07.010
  225. Cortessis VK, Thomas DC, Levine AJ, et al. Environmental epigenetics: prospects for studying epigenetic mediation of exposure-response relationships. Hum Genet. 2012;131(10):1565–1589. doi: 10.1007/s00439-012-1189-8
  226. Jirtle RL, Skinner MK. Environmental epigenomics and disease susceptibility. Nat Rew Gen. 2007;8(4):253–262. doi: 10.1038/nrg2045
  227. Perera F, Herbstman J. Prenatal environmental exposures, epigenetics, and disease. Reprod Toxicol. 2011;31(3):363–373. doi: 10.1016/j.reprotox.2010.12.055
  228. Luo A, Leach ST, Barres R, et al. The microbiota and epigenetic regulation of T helper 17 / regulatory T cells: in search of a balanced immune system. Front Immunol. 2017;8:417. doi: 10.3389/fimmu.2017.00417
  229. Zager A, Peron JP, Mennecier G, et al. Maternal immune activation in late gestation increases neuroinflammation and aggravates experimental autoimmune encephalomyelitis in the offspring. Brain Behav Immun. 2015;43:159–171. doi: 10.1016/j.bbi.2014.07.021
  230. Mandal M, Donnelly R, Elkabes S, et al. Maternal immune stimulation during pregnancy shapes the immunological phenotype of offspring. Brain Behav Immun. 2013;33:33–45. doi: 10.1016/j.bbi.2013.04.012
  231. Solati J, Asiaei M, Hoseini MH. Using experimental autoimmune encephalomyelitis as a model to study the effect of prenatal stress on fetal programming. Neurol Res. 2012;34(5):478–483. doi: 10.1179/1743132812Y.0000000032
  232. Stanisavljević S, Čepić A, Bojić S, et al. Oral neonatal antibiotic treatment perturbs gut microbiota and aggravates central nervous system autoimmunity in Dark Agouti rats. Sci Rep. 2019;9(1):918. doi: 10.1038/s41598-018-37505-7
  233. Ochoa-Reparaz J, Mielcarz DW, Ditrio LE, et al. Role of gut commensal microfora in the development of experimental autoimmune encephalomyelitis. J Immunol. 2009;183(10):6041–6050. doi: 10.4049/jimmunol.0900747
  234. Ochoa-Reparaz J, Mielcarz DW, Haque-Begum S, Kasper LH. Induction of a regulatory B cell population in experimental allergic encephalomyelitis by alteration of the gut commensal microbiora. Gut Microbes. 2010;1(2):103–108. doi: 10.4161/gmic.1.2.11515
  235. Yokote H, Miyake S, Croxford JL, et al. NKT cell-dependent amelioration of a mouse model of multiple sclerosis by altering gut microflora. Am J Pathol. 2008;173(6):1714–1723. doi: 10.2353/ajpath.2008.080622
  236. Graves JS, Chitnis T, Weinstock-Guttman B, et al. Maternal and perinatal exposures are associated with risk for pediatric-onset multiple sclerosis. Pediatrics. 2017;139(4):e20162838. doi: 10.1542/peds.2016-2838
  237. Corsini E, Sokooti M, Galli CL, et al. Pesticide induced immunotoxicity in humans: a comprehensive review of the existing evidence. Toxicology. 2013;307:123–135. doi: 10.1016/j.tox.2012.10.009
  238. Mokarizadeh A, Faryabi MR, Rezvanfar MA, Abdollahi M. A comprehensive review of pesticides and the immune dysregulation: mechanisms, evidence and consequences. Toxicol Mech Methods. 2015;25(4):258–278. doi: 10.3109/15376516.2015.1020182
  239. Barrett E, Guinane CM, Ryan CA, et al. Microbiota diversity and stability of the preterm neonatal ileum and colon of two infants. Microbiologyopen. 2013;2(2):215–225. doi: 10.1002/mbo3.64
  240. Barrett E, Deshpandey AK, Ryan CA, et al. The neonatal gut harbours distinct bifidobacterial strains. Arch Dis Child Fetal Neonatal Ed. 2015;100(5):F405–F410. doi: 10.1136/archdischild-2014-306110
  241. Goldacre A, Pakpoor J, Goldacre M. Maternal and perinatal characteristics of infants who, later in life, developed multiple sclerosis: Record-linkage study. Mult Scler Relat Disord. 2017;13:98–102. doi: 10.1016/j.msard.2017.02.004
  242. Ramagopalan SV, Valdar W, Dyment DA, et al. Canadian collaborative study group. no effect of preterm birth on the risk of multiple sclerosis: a population based study. BMC Neurol. 2008;8:30. doi: 10.1186/1471-2377-8-30
  243. Maghzi A-H, Etemadifar M, Heshmat-Ghahdarijani K, et al. Cesarean delivery may increase the risk of multiple sclerosis. Mult Scler J. 2012;18(4):468–471. doi: 10.1177/1352458511424904
  244. Conradi S, Malzahn U, Paul F, et al. Breastfeeding is associated with lower risk for multiple sclerosis. Mult Scler. 2013;19(5):553–558. doi: 10.1177/1352458512459683
  245. Norgaard M, Nielsen RB, Jacobsen JB, et al. Use of penicillin and other antibiotics and risk of multiple sclerosis: a population-based case-control study. Am J Epidemiol. 2011;174(8):945–948. doi: 10.1093/aje/kwr201
  246. Zeissig S, Blumberg RS. Life at the beginning: perturbation of the microbiota by antibiotics in early life and its role in health and disease. Nat Immunol. 2014;15(4):307–310. doi: 10.1038/ni.2847
  247. Neu J, Rushing J. Cesarean versus vaginal delivery: long-term infant outcomes and the hygiene hypothesis. Clin Perinatol. 2011;38(2):321–331. doi: 10.1016/j.clp.2011.03.008
  248. Bokulich NA, Chung J, Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med. 2016;8(343):343ra82. doi: 10.1126/scitranslmed.aad7121
  249. Yassour M, Vatanen T, Siljander H, et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci Transl Med. 2016;8(343):343ra81. doi: 10.1126/scitranslmed.aad0917
  250. Salminen S, Gibson G, McCartney A, Isolauri E. Influence of mode of delivery on gut microbiota composition in seven year old children. Gut. 2004;53(9):1388–1389. doi: 10.1136/gut.2004.041640
  251. Goedert JJ, Hua X, Yu G, Shi J. Diversity and composition of the adult fecal microbiome associated with history of cesarean birth or appendectomy: Analysis of the American Gut Project. EBioMedicine. 2014;1(2-3):167–172. doi: 10.1016/j.ebiom.2014.11.004
  252. Blaser MJ, Dominguez-Bello MG. The human microbiome before birth. Cell Host Microbe. 2016;20(5):558–560. doi: 10.1016/j.chom.2016.10.014
  253. Dalla Costa G, Romeo M, Esposito F, et al. Caesarean section and infant formula feeding are associated with an earlier age of onset of multiple sclerosis. Mult Scler Relat Disord. 2019;33:75–77. doi: 10.1016/j.msard.2019.05.010
  254. Nielsen NM, Bager P, Stenager E, et al. Cesarean section and offspring’s risk of multiple sclerosis: a Danish nationwide cohort study. Mult Scler. 2013;19(11):1473–1477. doi: 10.1177/1352458513480010
  255. Boehm G, Moro G. Structural and functional aspects of prebiotics used in infant nutrition. J Nutr. 2008;138(9):1818S–1828S. doi: 10.1093/jn/138.9.1818S
  256. Walker A. Breast milk as the gold standard for protective nutrients. J Pediatr. 2010;156(2 Suppl):S3–S7. doi: 10.1016/j.jpeds.2009.11.021
  257. Fernandez L, Langa S, Martin V, et al. The human milk microbiota: origin and potential roles in health and disease. Pharmacol Res. 2013;69(1):1–10. doi: 10.1016/j.phrs.2012.09.001
  258. Andersson B, Porras O, Hanson LA, et al. Inhibition of attachment of Streptococcus pneumoniae and Haemophilus influenzae by human milk and receptor oligosaccharides. J Infect Dis. 1986;153(2):232–237. doi: 10.1093/infdis/153.2.232
  259. Cravioto A, Tello A, Villafan H, et al. Inhibition of localized adhesion of enteropathogenic Escherichia coli to HEp-2 cells by immunoglobulin and oligosaccharide fractions of human colostrum and breast milk. J Infect Dis. 1991;163(6):1247–1255. doi: 10.1093/infdis/163.6.1247
  260. Gueimonde M, Laitinen K, Salminen S, Isolauri E. Breast milk: a source of bifidobacteria for infant gut development and maturation? Neonatology. 2007;92(1):64–66. doi: 10.1159/000100088
  261. Martín R, Heilig G, Zoetendal E, et al. Diversity of the Lactobacillus group in breast milk and vagina of healthy women and potential role in the colonization of the infant gut. J Appl Microbiol. 2007;103(6):2638–2644. doi: 10.1111/j.1365-2672.2007.03497.x
  262. Penders J, Vink C, Driessen C, et al. Quantification of Bifidobacterium spp., Escherichia coli and Clostridium difficile in faecal samples of breast-fed and formula-fed infants by real-time PCR. FEMS Microbiol Lett. 2005;243(1):141–147. doi: 10.1016/j.femsle.2004.11.052
  263. Bezirtzoglou E, Tsiotsias A, Welling GW. Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH). Anaerobe. 2011;17(6):478–482. doi: 10.1016/j.anaerobe.2011. 03.009
  264. Ezendam J, de Klerk A, Gremmer ER, van Loveren H. Effects of Bifidobacterium animalis administered during lactation on allergic and autoimmune responses in rodents. Clin Exp Immunol. 2008;154(3):424–431. doi: 10.1111/j.1365-2249.2008.03788.x
  265. Ezendam J, van Loveren H. Lactobacillus casei Shirota administered during lactation increases the duration of autoimmunity in rats and enhances lung inflammation in mice. Br J Nutr. 2008;99(1):83–90. doi: 10.1017/S0007114507803412
  266. Conradi S, Malzahn U, Paul F, et al. Breastfeeding is associated with lower risk for multiple sclerosis. Mult Scler. 2013;19(5):553–558. doi: 10.1177/1352458512459683
  267. Brenton JN, Engel CE, Sohn MW, Goldman MD. Breastfeeding during infancy is associated with a lower future risk of pediatric multiple sclerosis. Pediart Neurol. 2017;77:67–72. doi: 10.1016/j.pediatrneurol.2017.09.007
  268. Pisacane A, Impagliazzo N, Russo M, et al. Breast feeding and multiple sclerosis. BMJ. 1994;308(6941):1411–1412. doi: 10.1136/bmj.308.6941.1411
  269. Ragnedda G, Leoni S, Parpinel M, et al. Reduced duration of breastfeeding is associated with a higher risk of multiple sclerosis in both Italian and Norwegian adult males: the EnvI MS study. J Neurol. 2015;262(5):1271–1277. doi: 10.1007/s00415-015-7704-9
  270. Simon AK, Hollander GA, McMichael A. Evolution of the immune system in humans from infancy to old age. Proc Biol Sci. 2015;282(1821):20143085. doi: 10.1098/rspb.2014.3085
  271. Martin R, Nauta AJ, Ben Amor K, et al. Early life: Gut microbiota and immune development in infancy. Benef Microbes. 2010;1(4):367–382. doi: 10.3920/BM2010.0027
  272. Kamada N, Seo S-U, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13(5):321–335. doi: 10.1038/nri3430
  273. Kamada N, Núñez G. Regulation of the immune system by the resident intestinal bacteria. Gastroenterology. 2014;146(6):1477–1488. doi: 10.1053/j.gastro.2014.01.060
  274. Vatanen T, Kostic AD, d’Hennezel E, et al. Variation in microbiome LPS immunogenicity contributes to autoimmunity in humans. Cell. 2016;165(4):842–853. doi: 10.1016/j.cell.2016.04.007
  275. Wang Z-W, Wang P, Lin F-H. Early-life exposure to lipopolysaccharide reduces the severity of experimental autoimmune encephalomyelitis in adulthood and correlated with increased urine corticosterone and apoptotic CD4+ T cells. Neuroscienci. 2011;193:283–290. doi: 10.1016/j.neuroscience.2011.07.047
  276. Ellestad KK, Tsutsui S, Noorbakhsh F, et al. Early life exposure to lipopolysaccharide suppresses experimental autoimmune encephalomyelitis by promoting tolerogenic dendritic cells and regulatory T cells. J Immunol. 2009;183(1):298–309. doi: 10.4049/jimmunol.0803576
  277. Abdurasulova IN, Zubareva OE, Zhitnukhin Yu.L, et al. The course of experimental allergic encephalomyelitis in adult rats after administration of interleukin-1β at different periods in early life. J Neurosci Behav Physiol. 2015;46(7):794–802. doi: 10.1007/s11055-016-0313-y
  278. Bakker JM, Kavelaars A, Kamphuis PJGH, et al. Neonatal dexamethasone treatment increases susceptibility to experimental autoimmune disease in adult rats. J Immunol. 2000;165(10):5932–5937. doi: 10.4049/jimmunol.165.10.5932
  279. Stephan M, Straub RH, Breivik T, et al. Postnatal maternal deprivation aggravates experimental autoimmune encephalomyelitis in adult Lewis rats: reversal by chronic imipramine treatment. Int J Devl Neurosi. 2002;20(2):125–132. doi: 10.1016/s0736-5748(02)00007-2
  280. Teunis MAT, Heijnen CJ, Sluyter F, et al. Maternal deprivation of rat pups increases clinical symptoms of experimental autoimmune encephalomyelitis at adult age. J Neuroimmunol. 2002;133(1-2):30–38. doi: 10.1016/s0165-5728(02)00351-x
  281. Laban O, Dimitrijevic M, von Hoersten S, et al. Experimental allergic encephalomyelitis in adult DA rats subjected to neonatal handling or gentling. Brain Res. 1995;676(1):133–140. doi: 10.1016/0006-8993(95)00106-z
  282. Dimitrijevic M, Laban O, von Hoersten S, et al. Neonatal sound stress and development of experimental allergic encephalomyelitis in Lewis and DA rats. Int J Neurosci. 1994;78(1-2):135–143. doi: 10.3109/00207459408986052
  283. Columba-Cabezas S, Iaffaldano G, Chiarotti F, et al. Early handling increases susceptibility to experimental autoimmune encephalomyelitis (EAE) in C57BL/6 male mice. J Neuroimmunol. 2009;212(1-2):10–16. doi: 10.1016/j.jneuroim.2009.05.007
  284. Golubeva AV, Crampton S, Desbonnet L, et al. Prenatal stress-induced alterations in major physiological systems correlate with gut microbiota composition in adulthood. Psychoneuroendocrinol. 2015;60:58–74. doi: 10.1016/j.psyneuen.2015.06.002
  285. Zijlmans MAC, Korpela K, Riksen-Walravena JM, et al. Maternal prenatal stress is associated with the infant intestinal microbiota. Psychoneuroendocrinol. 2015;53:233–245. doi: 10.1016/j.psyneuen.2015.01.006
  286. Bailey MT, Lubach GR, Coe CL. Prenatal stress alters bacterial colonization of the gut in infant monkeys. J Pediatr Gastroenterol Nutr. 2004;38(4):414–421. doi: 10.1097/00005176-200404000-00009
  287. Bailey MT, Coe CL. Maternal separation disrupts the integrity of the intestinal microflora in infant rhesus monkeys. Dev Psychobiol. 1999;35(2):146–155.
  288. Bokulich NA, Chung J, Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med. 2016;8(343):343ra82. doi: 10.1126/scitranslmed.aad7121
  289. Miller JE, Wu C, Pedersen LH, et al. Maternal antibiotic exposure during pregnancy and hospitalization with infection in offspring: a population-based cohort study. Int J Epidemiol. 2018;47(2):561–571. doi: 10.1093/ije/dyx272
  290. Keogh CE, Kim DHJ, Pusceddu MM, et al. Myelin as a regulator of development of the microbiota – gut – brain axis. Brain Behav Immun. 2021;91:437–450. doi: 10.1016/j.bbi.2020.11.001
  291. Mirzaei F, Michels KB, Munger K, et al. Gestational vitamin D and the risk of multiple sclerosis in offspring. Ann Neurol. 2011;70(1):30–40. doi: 10.1002/ana.22456
  292. Fernandes de Abreu DA, Ibrahim EC, Boucraut J, et al. Severity of experimental autoimmune encephalomyelitis is unexpectedly reduced in mice born to vitamin D-deficient mothers. J Steroid Biochem Mol Biol. 2010;121(1-2):250–253. doi: 10.1016/j.jsbmb.2010.03.006
  293. Fernandes de Abreu DA, Landel V, Barnett AG. Prenatal vitamin D deficiency induces an early and more severe experimental autoimmune encephalomyelitis in the second generation. Int J Mol Sci. 2012;13(9):10911–10919. doi: 10.3390/ijms130910911
  294. Fernandes de Abreu DA, Landel V, Feron F. Seasonal, gestational and postnatal influences on multiple sclerosis: the beneficial role of a vitamin D supplementation during early life. J Neurol Sci. 2011;311(1-2):64–68. doi: 10.1016/j.jns.2011.08.044
  295. Adzemovic MZ, Zeitelhofer M, Hochmeister S, et al. Efficacy of vitamin D in treating multiple sclerosis-like neuroinflammation depends on developmental stage. Exp Neurol. 2013;249:39–48. doi: 10.1016/j.expneurol.2013.08.002
  296. Biesalski HK. Nutrition meets the microbiome: Micronutrients and the microbiota. Ann NY Acad Sci. 2016;1372(1):53–64. doi: 10.1111/nyas.13145
  297. Smith AD, Kim YI, Refsum H. Is folic acid good for everyone? Am J Clin Nutr. 2008;87(3):517–533. doi: 10.1093/ajcn/87.3.517
  298. Nagy-Szakal D, Ross MC, Dowd SE, et al. Maternal micronutrients can modify colonic mucosal microbiota matuartation in murien offspring. Gut Microbes. 2012;3(5):426–433. doi: 10.4161/gmic.20697
  299. Steegers-Theunissen RP, Obermann-Borst SA, Kremer D, et al. Periconceptional maternal folic acid use of 400 g per day is related to increased methylation of the IGF2 gene in the very young child. PLoS One. 2009;4(11):e7845. doi: 10.1371/journal.pone.0007845
  300. Collado MC, Cernada M, Baüerl C, et al. Microbial ecology and host-microbiota interactions during early life stages. Gut Microbes. 2012;3(4):352–365. doi: 10.4161/gmic.21215

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Figure. Phases of early infant colonization (modified by M.S. Cooperstock and A.J. Zedd [179]) and factors influencing this process. CP, colonization phase; CS, caesarean section; AF, artificial feeding. * Early colonization phases

Download (793KB)

Copyright (c) 2023 Eco-Vector



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 74760 от 29.12.2018 г.


This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies