Next Article in Journal
Defective Induction of IL-27-Mediated Immunoregulation by Myeloid DCs in Multiple Sclerosis
Next Article in Special Issue
IL-7 Deficiency Exacerbates Atopic Dermatitis in NC/Nga Mice
Previous Article in Journal
Peptaibol Analogs Show Potent Antibacterial Activity against Multidrug Resistant Opportunistic Pathogens
Previous Article in Special Issue
The Role of New 3D Pathology and Lymphocyte Expression of Interstitial Inflammation in Pediatric-Onset Lupus Nephritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Association between Taxonomic Composition of Gut Microbiota and Host Single Nucleotide Polymorphisms in Crohn’s Disease Patients from Russia

by
Maria Markelova
1,
Anastasia Senina
1,
Dilyara Khusnutdinova
1,
Maria Siniagina
1,
Elena Kupriyanova
1,
Gulnaz Shakirova
2,
Alfiya Odintsova
3,
Rustam Abdulkhakov
4,
Irina Kolesnikova
5,
Olga Shagaleeva
5,
Svetlana Lyamina
6,
Sayar Abdulkhakov
1,
Natalia Zakharzhevskaya
5 and
Tatiana Grigoryeva
1,*
1
Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russia
2
Municipal Polyclinic №21, 420139 Kazan, Russia
3
Republican Clinical Hospital, 420064 Kazan, Russia
4
Hospital Therapy Department, Kazan State Medical University, 420012 Kazan, Russia
5
Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
6
Molecular Pathology of Digestion Laboratory, A.I. Yevdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(9), 7998; https://doi.org/10.3390/ijms24097998
Submission received: 7 April 2023 / Revised: 25 April 2023 / Accepted: 26 April 2023 / Published: 28 April 2023
(This article belongs to the Special Issue Updates in Cell and Molecular Mechanisms of Autoimmune Diseases)

Abstract

:
Crohn’s disease (CD) is a chronic relapsing inflammatory bowel disease of unknown etiology. Genetic predisposition and dysbiotic gut microbiota are important factors in the pathogenesis of CD. In this study, we analyzed the taxonomic composition of the gut microbiota and genotypes of 24 single nucleotide polymorphisms (SNP) associated with the risk of CD. The studied cohorts included 96 CD patients and 24 healthy volunteers from Russia. Statistically significant differences were found in the allele frequencies for 8 SNPs and taxonomic composition of the gut microbiota in CD patients compared with controls. In addition, two types of gut microbiota communities were identified in CD patients. The main distinguishing driver of bacterial families for the first community type are Bacteroidaceae and unclassified members of the Clostridiales order, and the second type is characterized by increased abundance of Streptococcaceae and Enterobacteriaceae. Differences in the allele frequencies of the rs9858542 (BSN), rs3816769 (STAT3), and rs1793004 (NELL1) were also found between groups of CD patients with different types of microbiota communities. These findings confirm the complex multifactorial nature of CD.

1. Introduction

Crohn’s disease (CD) is a chronic relapsing disease characterized by inflammation of various regions of the gastrointestinal tract, mainly the small and large intestines. While the etiology of this disease is still unclear, it is known to be multifactorial. The pathogenesis of CD depends on environmental factors, genetic predisposition, individual immune response, and intestinal microbiota.
Through the development of DNA sequencing technology in recent decades, there is a significant amount of data on the intestinal microbiota in health and disease. It is known that inflammatory bowel diseases (IBD) mainly affect the regions of the gastrointestinal tract with the maximum density of the bacterial population (colon and small intestine). Many studies confirm the association of microbiota composition with IBD [1,2,3,4,5], including in the Russian population [6,7,8,9]. The microbiota of IBD patients is most often characterized by reduced alpha diversity, and a decrease in abundances of Firmicutes and Bacteroidetes, and an increase in Proteobacteria and E. coli, in particular. At the functional level, these changes lead to reduced levels of short-chain fatty acids (SCFAs), especially butyrate—anti-inflammatory metabolites produced by microbiota, and shifts in oxidative stress pathways and the secretion of toxins [10,11]. Many genetic polymorphisms associated with IBD are located in genes related to the host immune response and, in particular, interaction with the microbiota. The most studied is the mutation in the NOD2 gene, which determines the immune response to the peptidoglycan of the bacterial cell wall [12]. Thus, patients with CD carrying a mutation in NOD2 are characterized by an increased amount of adhesive bacteria and decrease in the representation of Faecalibacterium [13,14], and NOD2-deficient mice have an altered microbiome [15]. The NOD2 gene product interacts with the ATG16L1 gene product, mutations in which are also associated with CD [16]. TH1 and TH17 immune responses are increased in mice with the ATG16L1 T300A variant, and the bacterial genera Bacteroides and Escherichia are more prevalent in the intestines of these mice [17,18]. Mutations in the FUT2 and CARD9 genes also affect the gut microbiota of patients with IBD [18,19,20].
It is known that CD is accompanied by disturbed integrity of the intestinal wall due to inflammation [21]. This leads to an increased intestinal permeability and penetration of food and microbial antigens into the bloodstream, which is characterized by elevated serum antibodies against them. The most frequently mentioned immunoglobulins are anti-Saccharomyces cerevisiae antibodies (ASCA), which are significantly increased in the serum of patients with CD, coeliac disease, rheumatoid arthritis, and autoimmune liver diseases [22,23,24]. ASCA is known to be elevated even in unaffected relatives of IBD patients, probably due to a genetic predisposition to abnormal intestinal permeability [25]. In addition, bacteria can affect barrier function by degradation of the mucus layer, regulation of epithelial cell apoptosis, and synthesis of components necessary for tight junctions [26]. Thus, microbiota antigens can interact with the host immune system not only on the intestinal mucosa, but also in the bloodstream due to the impaired barrier function.
However, identifying human genome and microbiome associations in IBD patients remains an urgent task for a deeper understanding of the disease pathogenesis and further personalized treatment selection.

2. Results

2.1. Human Subjects

The present study involved 96 patients with Crohn’s disease (53 female and 43 male, mean age 32.3 ± 11.8 years). CD was diagnosed using standard clinical, endoscopic, and histological criteria. All patients were in the acute stage with varying severity and localization of inflammation (Table 1). The average duration of the CD was 9.3 ± 4.5 years. The control group consisted of 24 healthy volunteers (15 female and 9 male, mean age 35.3 ± 10.0 years).

2.2. Microbiota Analysis

2.2.1. Gut Microbiota of CD Patients and Healthy Volunteers

The number of sequencing read pairs obtained from fecal samples of CD patients and healthy controls ranged from 53,175 to 182,362 (median 91,061). Raw reads were deposited in the NCBI SRA under accession number PRJNA938107 in the fastq format. After merging, quality control, removing of chimeric reads, and rarefying, 20,938 reads per sample remained.
The major bacterial phyla constituting the intestinal microbiota of healthy volunteers and CD patients were Firmicutes (65.2 ± 14.7% and 63.0 ± 16.6%, respectively), Bacteroidetes (22.9 ± 15.5% and 19.8 ± 17.5%), Proteobacteria (2.5 ± 3.2% and 7.3 ± 10.9%), and Actinobacteria (6.8 ± 6.0% and 6.9 ± 8.5%).
Shannon’s alpha diversity index and observed operational taxonomic units (OTUs) were significantly reduced in CD patients compared with controls (Figure 1A). A decreased abundance of the families Clostidiaceae, Coriobacteriaceae, and Rikenellaceae and an increased representation of Lactobacillaceae, Enterococcaceae, Streptococcaceae, and Enterobacteriaceae were also found (Figure 1B).
Depending on the CDAI, differences in the taxonomic composition of CD patients’ microbiota are revealed. The Firmicutes phylum and Micrococcaceae and Enterococcaceae families showed significant positive correlations with CD activity (Figure 2A). Significant negative correlations with the severity of the disease were found for the Bacteroidetes phylum and Eryspelotrichaceae, [Odoribacteraceae], Rikenellaceae, Coriobacteriaceae, Bacteroidaceae, and Porphyromonadaceae families (Figure 2A). When CD patients were divided into three groups according to the activity of the disease, significant differences in the representation of three families were revealed—Micrococcaceae increased with the increase in CD severity, while the abundance of Coriobacteriaceae and Bacteroidaceae decreased (Figure 2B).
Based on the Dirichlet multinomial mixtures method, two types of microbiota can be distinguished according to the taxonomic composition (Figure 3). The first community type (I) included 61 CD patients and all 24 controls, while the second group (II) included 35 CD patients. Thus, the frequency of occurrence of microbiota types in CD patients and controls is significantly different (p = 0.0003, Exact Fisher test). The main driver representatives (the most abundant in these communities) of the first community type are the families Lachnospiraceae, Ruminococcaceae, and Bacteroidaceae, and an unclassified member of the order Clostridiales (Figure 4A), while the second type is determined by Lachnospiraceae, Streptococcaceae, Ruminococcaceae, and Enterobacteriaceae (Figure 4B).

2.2.2. Analysis of Microbiota Community Types in CD Patients

When comparing the two types of communities identified in CD patients, the second type showed a significant decrease in the number of observed OTUs and Shannon’s alpha diversity index, indicating a more prominent dysbiosis (Figure 5A, Table S1). A decrease in the abundance of the Bacteroidetes phylum and an increase in the Proteobacteria, Fusobacteria, and Verrucomicrobia phyla were also observed. Moreover, the abundance of the Bacteroidaceae, Prevotellaceae, Lachnospiraceae, Ruminococcaceae, and Erysipelotrichaceae families and unclassified Clostridiales were significantly declined in the second type of community of CD patients (Figure 5B, Table S1). These bacteria are members of the normal human microbiota and play a role in maintaining intestinal homeostasis. An increased amount of the Verrucomicrobiaceae, Enterococcaceae, Streptococcaceae, and Enterobacteriaceae families was also found (Figure 5B, Table S1). Thus, the microbiome of CD patients with community type II is characterized by prominent dysbiosis, while the microbiome of patients with the first type is more similar to the healthy ones.

2.2.3. Analysis of Clinical Parameters in CD Patients with Different Types of Microbial Communities

When comparing CD patients with different types of microbial communities, no significant differences were found in clinical characteristics—duration of disease, location of inflammation (ileitis, colitis, ileocolitis), disease activity (based on the Crohn’s disease activity index), phenotype of disease (inflammatory, stricturing, fistulizing), or stool frequency (Table 2).

2.3. SNP Analysis

SNP Analysis in CD Patients and Healthy Volunteers
All 24 genetic markers agreed to Hardy–Weinberg equilibrium proportions in the control population (p > 0.05). Allele frequencies of 8 genetic polymorphisms were significantly different between the CD groups and healthy subjects (Table 3). The alleles rs1004819A and rs11209026G of the IL23R gene, as well as rs2241880A (ATG16L1), rs4958847A (IRGM), rs1992662G (PTGER4), rs2274910C (ITLN1), rs6601764T, and rs7807258C were found to be more frequent in patients with CD.

2.4. Analysis of Association between Microbiota and SNPs Allele Frequency in CD Patients

2.4.1. SNP Analysis in CD Patients According to the Type of Microbial Community

In the group of CD patients with the second type of gut microbiota community, the following allele frequencies: A in rs9858542 of the BSN gene, T in rs3816769 of the STAT3 gene, and C in rs1793004 of the NELL1 gene were significantly increased (Table 4). All of these alleles are associated with an increased risk of CD [27,28,29,30,31,32,33].

2.4.2. Correlation between SNP and Taxonomic Composition of Gut Microbiota in CD Patients

Statistically significant negative correlations of rs9858542 (BSN) with the number of observed OTUs and the representation of the Bacteroidetes phylum were revealed using an additive model (Figure 6). Rs3816769 (STAT3) showed a negative correlation with the phylum Bacteroidetes and especially with the family Bacteroidaceae. For rs1793004 (NELL1) a negative correlation was found with the family Ruminococcaceae and a positive correlation with the family Enterococcaceae. Furthermore, significant negative correlations were found between abundance of Bacteroidaceae with rs2274910 (ITLN1), rs2522057 (IRF1-AS1), rs224136 (intergenic), rs6908425 (CDKAL1), and rs12037606 (intergenic) and a positive correlations with rs1992662 (PTGER4), rs1456893 (intergenic), and 13361189 (IRGM). Enterococcaceae and Enterobacteriaceae families showed significant positive correlations with rs224136 (intergenic).

3. Discussion

Changes in the gut microbiota composition and role of genetics in CD patients have been described in a number of studies. However, there is limited data on CD patients in the Russian population. CD prevalence in Russia is estimated to be 3.0–7.88 cases per 100,000 population [34,35], and it rises 8–10% annually [35], but it is still substantially lower than in Western Europe and North America [36]. Patients in our study were recruited from two regions of Russia (the Republic of Tatarstan and Moscow), ensuring that people of different nationalities (mainly Russians and Tatars) were represented.
Our results indicate a decrease in the diversity of the gut microbiota in CD patients compared to healthy volunteers, which has also been found in many other studies [37,38,39]. Changes in the abundance of the families Bacteroidaceae, Prevotellaceae, Clostridiaceae, Lachnospiraceae, Ruminococcaceae, Eryspelotrichaceae, Enterobacteriaceae, Fusobacteriaceae, Lactobacillaceae, Enterococcaceae, and Streptococcaceae are often detected. The families Bacteroidaceae, Prevotellaceae, and Rikenellaceae are members of the phylum Bacteroidetes and perform several important functions in the gut, including metabolizing proteins and carbohydrates [40], producing butyrate [41], and preventing the colonization of the gastrointestinal tract by pathogenic bacteria [42]. In our study, among the most abundant phylum Bacteroidetes in CD patients, only the Rikenellaceae family decreased significantly compared to controls. The functions of this family in the gut microbiota have not yet been studied, but there is evidence of its decrease in patients with IBD and an increase in patients with irritable bowel syndrome [43]. Among the representatives of the phylum Firmicutes, there was a decrease in the proportion of the order Clostridiales and, in particular, of the family Clostridiaceae. They are known to be SCFAs producers and are involved in the metabolism of bile acids. There is a number of conflicting data on this taxon. While some authors observe an increase of Clostidiaceae in healthy controls and a decrease in CD patients [44,45,46,47], the others found an increase of this taxon in IBD patients [48,49]. In our study, we found a decreased abundance of the Coriobacteriaceae family of the Actinobacteria phylum in CD patients, which is consistent with previous studies [50,51,52]. Coriobacteriaceae have important functions in the gut including the conversion of bile salts and steroids and the activation of dietary polyphenols [53].
An increase of the Enterobacteriaceae family members was also found in the microbiota of CD patients. This is consistent with previously reported data in which the increased representation of this family was a marker of dysbiosis in IBD [8,54]. However, no association of any E. coli virulence genes with CD was found in the Russian population [55]. In our study, we found an increase in the proportion of lactic acid producing bacteria from the Lactobacillaceae, Enterococcaceae, and Streptococcaceae families in patients with CD, which is consistent with previous studies [9,56,57,58,59,60]. These bacteria are commensals; however, they can sometimes cause inflammation of various tissues in the respiratory, cardiovascular, and nervous systems [61,62,63,64]. Streptococci are known to provoke intestinal inflammation by inducing a pro-inflammatory response to lipoproteins and other components, as well as to the interaction of subtilisin-like protease (SspA) with Toll-like receptor 2 (TLR2) [65]. The role of enterococci in the pathogenesis of IBD has been described in a study showing that Enterococcus faecalis can cause IBD in the IL-10 knockout mouse model [66]. A pathogenicity island encoding surface aggregating protein (asa1), gelatinase (gelE), cytolysin (cylA), extracellular surface protein (esp), and hyaluronidase (hyl) was also identified as a possible trigger of the host inflammatory response [67]. Whether lactobacilli can provoke IBD or are simply adapted to survive in an inflamed gut is still an open question.
Many studies attempted to identify bacterial taxa that change with IBD activity/severity. Many of them agree that Faecalibacterium prausnitzii is associated with minimal inflammation [68,69,70]. However, the results for other taxa are conflicting. We found an increase in the representation of the Enterococcaceae and Micrococcaceae families in the gut microbiota of patients with more severe CD, which is consistent with the results of other studies [71,72,73]. In addition, we found a decrease in the abundance of the Eryspelotrichaceae and Coriobacteriaceae families with higher disease activity. A similar trend was observed for the Eryspelotrichaceae, while the opposite was found for Coriobacteriaceae by Papa et al. [74]. In our study, similar to Tedjo et al. Bacteroidaceae were increased in patients with mild CD [75], whereas other authors found the opposite [46,75,76]. Therefore, there is no clear understanding of the microbiota composition of IBD patients according to disease severity.
According to our data, the microbiota of CD patients is heterogeneous and two types of communities that can be identified. Thus, patients with a type I microbiota community shared it with control samples. Patients with a type II microbiota community are characterized by a lower diversity of the microbiota and a lower number of observed OTUs compared with the type I, indicating a more severe dysbiosis. In a study by Vieira-Silva et al., a similar method revealed four enterotypes, whose drivers were Ruminococcaceae, Prevotella, and Bacteroides [70]. The microbiota enterotypes of the Japanese, European, and American populations are characterized by the same taxa [77]. Other enterotypes were identified in a model organism study by Barron et al. where the main driver taxa were Lachnospiraceae and Ruminoccoacceae, Enterobacteriaceae and Lactobacillus, Erysipelotrichaceae and Akkermansia [78]. In our study, a number of bacterial families were represented differently in the microbiota community types. Thus, study participants with community type II had an increased abundance of Enterobacteriaceae, Enterococcaceae, and Streptococcaceae families, which, as noted above, are typical characteristics of CD patients’ gut microbiota. In addition, the abundance of Verrucomicrobiaceae, whose role in the pathogenesis of IBD is actively debated, was increased. Some authors noted a decrease of its representation in IBD and even suggested the use of Akkermansia muciniphila as a new generation probiotics [9,78,79,80], while others showed its increase in the microbiota of CD patients and suggested that it degrades the mucin of the intestinal mucosa, thereby provoking its inflammation [8]. There was also a decrease in the abundance of Bacteroidaceae, Prevotellaceae, Lachnospiraceae, Ruminococcaceae, Erysipelotrichaceae, and unclassified Clostridiales in the CD patients’ microbiota community of type II. These bacteria belong to the normal microbiota and are important in keeping the gut healthy. Thus, the second type of microbial community is characterized with more prominent dysbiotic changes in the microbiota of CD patients.
We found no differences in the clinical characteristics of CD (duration of disease, location of inflammation, disease activity, and stool frequency) between the two groups of patients with different types of gut microbiota communities, suggesting the presence of other reasons for this distribution.
As CD is a multifactorial disease, genetic factors may be responsible for differences in the gut microbiota composition. There are 24 single nucleotide polymorphisms studied, which have previously been associated with CD in various populations. Compared with controls, patients with CD have a significantly higher allele frequency of 8 SNPs. For the remaining 16 SNPs, no significant differences were found, probably due to the regional characteristics of the Russian population o the limited size of the cohort. It is known that the representation of some bacterial taxa in the intestinal microbiota is associated with specific alleles of host SNP. Therefore, polymorphisms in the LCT gene determine the percentage of Bifidobacterium in the gut microbiota of healthy individuals [81], which can be explained by bacterial enzymes compensating for lactase deficiency. There are also data on the relationship of representatives of Akkermansia, Anaerostipes, Clostridiaceae, Blautia, Dialister, Bacteroides, Atopobium, etc. with various host genetic loci, but the mechanism of these relationships has not been studied [81,82,83]. In the case of IBD, a high abundance of Enterobacteriaceae was found in the microbiota of NOD2-deficient patients [84]. Certain polymorphisms in the FUT2 gene were associated with decreased SCFAs-producing Faecalibacterium and increased Proteobacteria [85]. It is also known that the ATG16L1 T300A variant is associated with increased abundance of the Bacteroides genus [17]. In our study, rs9858542A allele in the BSN gene was found to be more frequent in CD patients with a second dysbiotic type of microbiota community and negatively correlated with the number of observed OTUs and Bacteroidetes phylum representation. The rs9858542A allele is known to be associated with an increased risk of CD [27,28,29]. The BSN gene encodes Bassoon Presynaptic Cytomatrix Protein, which is involved in organizing the presynaptic cytoskeleton and expressed primarily in brain neurons, although there is an evidence that this protein is also expressed at low levels in enteroendocrine cells in the gastrointestinal tract, including the stomach, duodenum, colon, and rectum [86]. These cells produce gut hormones that control digesting and food absorbtion, insulin secretion, etc. [87]. It is also known that the gut microbiota produce several metabolites (SCFAs, secondary bile acids, indoles, and lipopolysaccharides) that stimulate enteroendocrine cells [88,89,90,91,92]. The mechanism of BSN gene product interaction with intestinal microbiota is still unknown, but probably involves the interplay of microbiota metabolites with host enteroendocrine cells. In our study, we also found that the T allele in rs3816769 of the STAT3 gene is significantly more frequent in CD patients with a second dysbiotic type of intestinal microbiota and negatively correlates with the Bacteroidetes phylum and Bacteroidaceae family in particular. This variant is also known to be associated with CD risk [30,93]. The transcription factor STAT3 (signal transducer and activator of transcription 3) regulates apoptosis, cell growth and inflammation in response to internal and external stimuli. In animal models, STAT3 activation in intestinal epithelial cells is required for wound healing, but also leads to the development of colitis-associated cancer in chronic inflammation [92,94]. Additionally, STAT3-deficient mice have increased sensitivity to bacterial lipopolysaccharide and increased levels of pro-inflammatory cytokines, and are more prone to chronic enterocolitis [95]. Zhao et al. found that microbial SCFAs activate STAT3 in intestinal epithelial cells, while STAT3 knockout resulted in a decrease in SCFA-induced antimicrobial peptide production [96]. Therefore, the STAT3 gene mutation rs3816769T may affect the host-microbiota interaction. The C allele of rs1793004 in the NELL1 gene was significantly more frequent in CD patients with the second dysbiotic microbiota type. Furthermore, a negative correlation of this variant with the Ruminococcaceae family and a positive correlation with Enterococcaceae were found. NELL1 encodes neural epidermal growth factor-like 1, which is expressed at significant levels in epithelial cells of the small and large intestine, including inflamed epithelium [97]. The association of rs1793004C with IBD has been demonstrated by a genome-wide association study in a German population of IBD patients [97]. However, the mechanisms by which the NELL1 gene product interacts with the intestinal microbiota remain unknown.
The findings of this study regarding the association between genetic polymorphisms and intestinal microbiota composition may help in developing personalized therapy for CD patients. Probiotics are considered a promising treatment of various autoimmune diseases–type 1 diabetes [98], multiple sclerosis [99], autoimmune hepatitis [100], rheumatoid arthritis [101], etc. Such therapy may include traditional probiotics (based on lactobacilli and bifidobacteria), next generation probiotics (based on Faecalibacterium prausnitzii [102] or Akkermansia muciniphila [103]), or fecal microbiota transplantation [104].
The limitation of the study is the relatively small number of healthy volunteers. However, differences in the microbiota of CD patients and healthy controls have been described in many previous works, while the variability of the microbiota within a group of CD patients is much less discussed. For this reason, we decided to study a larger number of CD patients for more reliable results. Taking these limitations into account, further investigations on associations of microbiota and genetic markers in both CD patients and healthy controls are required.

4. Materials and Methods

4.1. CD Patient and Controls

Venous blood and stool samples were collected from CD patients admitted to the Republican Clinical Hospital (Kazan, Russia) and clinical department of the Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency (Moscow, Russia) during the period 2017–2021 (43/53 male/female, 32.3 ± 11.8 years old). CD was diagnosed using standard clinical, endoscopic, and histological criteria. The control group consisted of 24 volunteers (9/15 male/female, 35.3 ± 10.0 years old) from the same regions of Russia as the CD patients. Eligibility of patients with CD and healthy volunteers was determined according to specific inclusion/exclusion criteria as listed in Table S2.

4.2. Ethics Statement

Informed consent was obtained from all subjects involved in the study. The study was conducted in accordance with the recommendations of the local ethics committee of the Kazan Federal University, Kazan, Russia (Protocol No. 6, dated 13 October 2017) and Interuniversity ethics committee, Moscow, Russia (Protocol No.8, dated 23 September 2021).

4.3. 16S rRNA Gene-Based Metagenomic Analysis of Stool Samples

Genomic DNA was extracted from fecal samples using the QIAamp DNA Stool Mini Kit (Qiagen, Germantown, MD, USA) in accordance with the manufacturer’s instructions. A 16S rRNA sequencing library was constructed according to the 16S metagenomics sequencing library preparation protocol (Illumina, San Diego, CA, USA) targeting the V3 and V4 hypervariable regions of the 16S rRNA gene. The initial PCR was performed with template DNA using region-specific primers shown to have compatibility with the Illumina index and sequencing adapters (forward primer: 5′-TCGTCGGCAGCGTCAGATGTGTATAAGAGACAGTCGTCGGCAGCGTCAGATGTGTATAAGAGACAGCCTACGGGNGGCWGCAG-3′; reverse primer: 5′-GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGGTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGGACTACHVGGGTATCTAATCC-3′). After purification of PCR products with AMPure XP magnetic beads, the second PCR was performed using primers from a Nextera XT Index Kit (Illumina). Subsequently, purified PCR products were visualized using gel electrophoresis and quantified with a Qubit dsDNA HS Assay Kit (Thermo Scientific, Waltham, MA, USA) on a Qubit 2.0 fluorometer. The sample pool (4 nM) was denatured with 0.2 N NaOH, diluted further to 4 pM, and combined with 20% (v/v) denatured 4 pM PhiX, prepared following Illumina guidelines. Sequencing of 16S rRNA gene V3-V4 variable regions was performed on the Illumina MiSeq platform in 2 × 300 bp mode at the Interdisciplinary Center of Shared Use of Kazan Federal University.
Reads were further processed and analyzed using the QIIME software, version 1.9.1 [105] according to protocols. Before filtering, there were 53,175–182,362 (median 91,061) read pairs per sample. Paired-end reads were initially merged and then processed to remove low quality and chimeric sequence data. The rarefaction step was performed to reduce sequencing depth heterogeneity between samples. After quality filtering, chimera filtering and rarefying, we analyzed on average 20,938 joined read pairs. Sequences were clustered into operational taxonomic units (OTU) based on the 97% identity threshold (open reference-based OTU picking strategy); the SILVA database v.138 [106] was used. To characterize the richness and evenness of the bacterial community, the alpha diversity index was calculated using Shannon’s metrics.

4.4. Genotyping

A total of 24 SNPs were selected based on data indicating their potential association with risk for IBD (Table S3). Genomic DNA from venous blood was isolated and purified using the QIAamp DNA Mini Kit (Qiagen, Germantown, MD, USA) as described by the manufacturer. PCR amplification was performed using the primers listed in Table S3 according to the protocol [107]. Genotyping was performed using MALDI-TOF mass spectrometry as described previously [107].

4.5. Statistical Analysis

The distribution of genotypes for all SNPs was tested for compliance with the Hardy-Weinberg equilibrium using the chi-square test. Analysis of the allele frequencies was done using Fisher’s exact test. The strength of associations was assessed using the odds ratio (OR, (lower 95% confidence interval; upper 95% confidence interval)). Differences in the taxonomic composition of the gut microbiota were assessed using the Kruskal-Wallis test. Correlations between genotypes and gut microbiota composition were analyzed using the R “psych” package [108] based on the Spearman’s rank correlation coefficient using an additive genetic model (depending on the genotype, a higher risk of developing CD corresponds to a higher rank). p < 0.05 values were considered as significant. To determine the types of bacterial communities, the Dirichlet multinomial mixture algorithm was applied to cluster the gut microbiota samples [109].

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms24097998/s1. References [110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148] are cited in the supplementary materials.

Author Contributions

Conceptualization, M.M., T.G. and N.Z.; methodology, A.S., D.K., E.K., I.K., O.S. and S.L.; software, M.M.; validation, M.M., A.S. and D.K.; formal analysis, M.M.; investigation, M.M., A.S., D.K., I.K., O.S. and S.L.; resources, G.S., A.O., R.A. and S.A.; data curation, M.M.; writing—original draft preparation, M.M. and M.S.; writing—review and editing, M.M., A.S., D.K., M.S., G.S., I.K., O.S., S.L., S.A., N.Z. and T.G.; visualization, M.M., supervision, N.Z. and T.G; funding acquisition, N.Z. and T.G. All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by the Kazan Federal University Strategic Academic Leadership Program (PRIORITY-2030) and a subsidy allocated to Kazan Federal University for the state assignment in the sphere of scientific activities (#FZSM-2023-0013).

Institutional Review Board Statement

The study was conducted in accordance with the recommendations of the local ethics committee of the Kazan Federal University, Kazan, Russia (Protocol No. 6, dated 13 October 2017) and Interuniversity ethics committee, Moscow, Russia (Protocol No.8, dated 23 September 2021).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. Raw reads are deposited in the NCBI SRA under accession number PRJNA938107 in the fastq format (https://www.ncbi.nlm.nih.gov/sra/PRJNA938107, accessed on 30 March 2023).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Franzosa, E.A.; Sirota-Madi, A.; Avila-Pacheco, J.; Fornelos, N.; Haiser, H.J.; Reinker, S.; Vatanen, T.; Hall, A.B.; Mallick, H.; McIver, L.J.; et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019, 4, 293–305. [Google Scholar] [CrossRef]
  2. Halfvarson, J.; Brislawn, C.J.; Lamendella, R.; Vázquez-Baeza, Y.; Walters, W.A.; Bramer, L.M.; D’Amato, M.; Bonfiglio, F.; McDonald, D.; Gonzalez, A.; et al. Dynamics of the human gut microbiome in inflammatory bowel disease. Nat. Microbiol. 2017, 2, 17004. [Google Scholar] [CrossRef] [PubMed]
  3. Vrakas, S.; Mountzouris, K.C.; Michalopoulos, G.; Karamanolis, G.; Papatheodoridis, G.; Tzathas, C.; Gazouli, M. Intestinal Bacteria Composition and Translocation of Bacteria in Inflammatory Bowel Disease. PLoS ONE 2017, 12, e0170034. [Google Scholar] [CrossRef]
  4. Andoh, A.; Kuzuoka, H.; Tsujikawa, T.; Nakamura, S.; Hirai, F.; Suzuki, Y.; Matsui, T.; Fujiyama, Y.; Matsumoto, T. Multicenter analysis of fecal microbiota profiles in Japanese patients with Crohn’s disease. J. Gastroenterol. 2012, 47, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
  5. Pascal, V.; Pozuelo, M.; Borruel, N.; Casellas, F.; Campos, D.; Santiago, A.; Martinez, X.; Varela, E.; Sarrabayrouse, G.; Machiels, K.; et al. A microbial signature for Crohn’s disease. Gut 2017, 66, 813–822. [Google Scholar] [CrossRef]
  6. Sitkin, S.I.; Vakhitov, T.Y.; Demyanova, E.V. Microbiome, gut dysbiosis and inflammatory bowel disease: That moment when the function is more important than taxonomy. Almanac. Clin. Med. 2018, 46, 396–425. [Google Scholar] [CrossRef]
  7. Sitkin, S.I.; Vakhitov, T.Y.; Tkachenko, E.I.; Oreshko, L.S.; Zhigalova, T.N.; Radchenko, V.G.; Seliverstov, P.V.; Avalueva, E.B.; Suvorova, M.A.; Komlichenko, E.V. Gut microbiota in ulcerative colitis and cealic disease. Exp. Gastroenterol. 2017, 1, 8–30. [Google Scholar]
  8. Danilova, N.A.; Abdulkhakov, S.R.; Grigoryeva, T.V.; Markelova, M.I.; Vasilyev, I.Y.; Boulygina, E.A.; Ardatskaya, M.D.; Pavlenko, A.V.; Tyakht, A.V.; Odintsova, A.K.; et al. Markers of dysbiosis in patients with ulcerative colitis and Crohn’s disease. Ter. Arkh. 2019, 91, 17–24. [Google Scholar] [CrossRef] [PubMed]
  9. Lo Sasso, G.; Khachatryan, L.; Kondylis, A.; Battey, J.N.D.; Sierro, N.; Danilova, N.A.; Grigoryeva, T.V.; Markelova, M.I.; Khusnutdinova, D.R.; Laikov, A.V.; et al. Inflammatory Bowel Disease-Associated Changes in the Gut: Focus on Kazan Patients. Inflamm. Bowel. Dis. 2021, 27, 418–433. [Google Scholar] [CrossRef]
  10. Erickson, A.R.; Cantarel, B.L.; Lamendella, R.; Darzi, Y.; Mongodin, E.F.; Pan, C.; Shah, M.; Halfvarson, J.; Tysk, C.; Henrissat, B.; et al. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS ONE 2012, 7, e49138. [Google Scholar] [CrossRef]
  11. Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef] [PubMed]
  12. Ogura, Y.; Bonen, D.K.; Inohara, N.; Nicolae, D.L.; Chen, F.F.; Ramos, R.; Britton, H.; Moran, T.; Karaliuskas, R.; Duerr, R.H.; et al. A frameshift mutation in NOD2 associated with susceptibility to Crohn’s disease. Nature 2001, 411, 603–606. [Google Scholar] [CrossRef] [PubMed]
  13. Swidsinski, A.; Ladhoff, A.; Pernthaler, A.; Swidsinski, S.; Loening-Baucke, V.; Ortner, M.; Weber, J.; Hoffmann, U.; Schreiber, S.; Dietel, M.; et al. Mucosal flora in inflammatory bowel disease. Gastroenterology 2002, 122, 44–54. [Google Scholar] [CrossRef]
  14. Li, E.; Hamm, C.M.; Gulati, A.S.; Sartor, R.B.; Chen, H.; Wu, X.; Zhang, T.; Rohlf, F.J.; Zhu, W.; Gu, C.; et al. Inflammatory bowel diseases phenotype, C. difficile and NOD2 genotype are associated with shifts in human ileum associated microbial composition. PLoS ONE 2012, 7, e26284. [Google Scholar] [CrossRef]
  15. Al Nabhani, Z.; Lepage, P.; Mauny, P.; Montcuquet, N.; Roy, M.; Le Roux, K.; Dussaillant, M.; Berrebi, D.; Hugot, J.P.; Barreau, F. Nod2 Deficiency Leads to a Specific and Transmissible Mucosa-associated Microbial Dysbiosis Which Is Independent of the Mucosal Barrier Defect. J. Crohn’s Colitis 2016, 10, 1428–1436. [Google Scholar] [CrossRef]
  16. Travassos, L.H.; Carneiro, L.A.; Ramjeet, M.; Hussey, S.; Kim, Y.G.; Magalhães, J.G.; Yuan, L.; Soares, F.; Chea, E.; Le Bourhis, L.; et al. Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma membrane at the site of bacterial entry. Nat. Immunol. 2010, 11, 55–62. [Google Scholar] [CrossRef]
  17. Lavoie, S.; Conway, K.L.; Lassen, K.G.; Jijon, H.B.; Pan, H.; Chun, E.; Michaud, M.; Lang, J.K.; Gallini Comeau, C.A.; Dreyfuss, J.M.; et al. The Crohn’s disease polymorphism, ATG16L1 T300A, alters the gut microbiota and enhances the local Th1/Th17 response. Elife 2019, 8, e39982. [Google Scholar] [CrossRef] [PubMed]
  18. Frank, D.N.; Robertson, C.E.; Hamm, C.M.; Kpadeh, Z.; Zhang, T.; Chen, H.; Zhu, W.; Sartor, R.B.; Boedeker, E.C.; Harpaz, N.; et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm. Bowel. Dis. 2011, 17, 179–184. [Google Scholar] [CrossRef]
  19. Rausch, P.; Rehman, A.; Künzel, S.; Häsler, R.; Ott, S.J.; Schreiber, S.; Rosenstiel, P.; Franke, A.; Baines, J.F. Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (Secretor) genotype. Proc. Natl. Acad. Sci. USA 2011, 108, 19030–19035. [Google Scholar] [CrossRef]
  20. Lamas, B.; Richard, M.L.; Leducq, V.; Pham, H.P.; Michel, M.L.; Da Costa, G.; Bridonneau, C.; Jegou, S.; Hoffmann, T.W.; Natividad, J.M.; et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 2016, 22, 598–605. [Google Scholar] [CrossRef]
  21. Turpin, W.; Lee, S.H.; Raygoza Garay, J.A.; Madsen, K.L.; Meddings, J.B.; Bedrani, L.; Power, N.; Espin-Garcia, O.; Xu, W.; Smith, M.I.; et al. Increased Intestinal Permeability Is Associated With Later Development of Crohn’s Disease. Gastroenterology 2020, 159, 2092–2100.e5. [Google Scholar] [CrossRef] [PubMed]
  22. Walker, L.J.; Aldhous, M.C.; Drummond, H.E.; Smith, B.R.; Nimmo, E.R.; Arnott, I.D.; Satsangi, J. Anti-Saccharomyces cerevisiae antibodies (ASCA) in Crohn’s disease are associated with disease severity but not NOD2/CARD15 mutations. Clin. Exp. Immunol. 2004, 135, 490–496. [Google Scholar] [CrossRef] [PubMed]
  23. Granito, A.; Zauli, D.; Muratori, P.; Muratori, L.; Grassi, A.; Bortolotti, R.; Petrolini, N.; Veronesi, L.; Gionchetti, P.; Bianchi, F.B.; et al. Anti-Saccharomyces cerevisiae and perinuclear anti-neutrophil cytoplasmic antibodies in coeliac disease before and after gluten-free diet. Aliment. Pharmacol. Ther. 2005, 21, 881–887. [Google Scholar] [CrossRef]
  24. Granito, A.; Muratori, L.; Muratori, P.; Guidi, M.; Lenzi, M.; Bianchi, F.B.; Volta, U. Anti-saccharomyces cerevisiae antibodies (ASCA) in coeliac disease. Gut 2006, 55, 296. [Google Scholar]
  25. Vermeire, S.; Peeters, M.; Vlietinck, R.; Joossens, S.; Den Hond, E.; Bulteel, V.; Bossuyt, X.; Geypens, B.; Rutgeerts, P. Anti-Saccharomyces cerevisiae antibodies (ASCA), phenotypes of IBD, and intestinal permeability: A study in IBD families. Inflamm. Bowel. Dis. 2001, 7, 8–15. [Google Scholar] [CrossRef] [PubMed]
  26. Merga, Y.; Campbell, B.J.; Rhodes, J.M. Mucosal barrier, bacteria and inflammatory bowel disease: Possibilities for therapy. Dig. Dis. 2014, 32, 475–483. [Google Scholar] [CrossRef]
  27. Latiano, A.; Palmieri, O.; Corritore, G.; Valvano, M.R.; Bossa, F.; Cucchiara, S.; Castro, M.; Riegler, G.; De Venuto, D.; D’Incà, R.; et al. Variants at the 3p21 locus influence susceptibility and phenotype both in adults and early-onset patients with inflammatory bowel disease. Inflamm. Bowel. Dis. 2010, 16, 1108–1117. [Google Scholar] [CrossRef]
  28. Márquez, A.; Cénit, M.C.; Núñez, C.; Mendoza, J.L.; Taxonera, C.; Díaz-Rubio, M.; Bartolomé, M.; Arroyo, R.; Fernández-Arquero, M.; de la Concha, E.G.; et al. Effect of BSN-MST1 locus on inflammatory bowel disease and multiple sclerosis susceptibility. Genes Immun. 2009, 10, 631–635. [Google Scholar] [CrossRef]
  29. Yuan, F.; Hung, R.J.; Walsh, N.; Zhang, H.; Platz, E.A.; Wheeler, W.; Song, L.; Arslan, A.A.; Beane Freeman, L.E.; Bracci, P.; et al. Genome-Wide Association Study Data Reveal Genetic Susceptibility to Chronic Inflammatory Intestinal Diseases and Pancreatic Ductal Adenocarcinoma Risk. Cancer Res. 2020, 80, 4004–4013. [Google Scholar] [CrossRef]
  30. Ferguson, L.R.; Han, D.Y.; Fraser, A.G.; Huebner, C.; Lam, W.J.; Morgan, A.R.; Duan, H.; Karunasinghe, N. Genetic factors in chronic inflammation: Single nucleotide polymorphisms in the STAT-JAK pathway, susceptibility to DNA damage and Crohn’s disease in a New Zealand population. Mutat. Res. 2010, 690, 108–115. [Google Scholar] [CrossRef]
  31. Can, G.; Tezel, A.; Gürkan, H.; Tozkır, H.; Ünsal, G.; Soylu, A.R.; Ümit, H.C. Investigation of IL23R, JAK2, and STAT3 gene polymorphisms and gene-gene interactions in Crohn’s disease and ulcerative colitis in a Turkish population. Turk. J. Gastroenterol. 2016, 27, 525–536. [Google Scholar] [CrossRef] [PubMed]
  32. Robinson, P.; Magness, E.; Montoya, K.; Engineer, N.; Eckols, T.K.; Rodriguez, E.; Tweardy, D.J. Genetic and Small-Molecule Modulation of Stat3 in a Mouse Model of Crohn’s Disease. J. Clin. Med. 2022, 11, 7020. [Google Scholar] [CrossRef] [PubMed]
  33. Cheng, X.; Shi, J.; Jia, Z.; Ha, P.; Soo, C.; Ting, K.; James, A.W.; Shi, B.; Zhang, X. NELL-1 in Genome-Wide Association Studies across Human Diseases. Am. J. Pathol. 2022, 192, 395–405. [Google Scholar] [CrossRef] [PubMed]
  34. Veselov, A.V. Inflammatory Bowel Diseases in the Russian Federation: Problems of the Regulatory Framework and Their Solutions. Materials of the On-Site Meeting of the Expert Council on Healthcare, the Committee of the Federation Council on Social Policy on the Topic “Regulatory and Legal Improvement in the Provision of Medical Care to Patients with Inflammatory Bowel Diseases”. 2018. Available online: http://social.council.gov.ru/activity/activities/expert_activities/94070/ (accessed on 25 February 2023).
  35. Bezdenezhnykh, T.P.; Fedyayev, D.V.; Khachatryan, G.R.; Arutyunov, G.G.; Gerasimova, K.V. Economic assessment of optimization of medical care for patients with inflammatory bowel diseases on the example of the Republic of Tatarstan. Farmakoekonomika. Sovrem. Farmakoekon. Farm. 2019, 12, 14–26. [Google Scholar] [CrossRef]
  36. Ng, S.C.; Shi, H.Y.; Hamidi, N.; Underwood, F.E.; Tang, W.; Benchimol, E.I.; Panaccione, R.; Ghosh, S.; Wu, J.C.Y.; Chan, F.K.L.; et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. Lancet 2017, 390, 2769–2778. [Google Scholar] [CrossRef]
  37. Manichanh, C.; Rigottier-Gois, L.; Bonnaud, E.; Gloux, K.; Pelletier, E.; Frangeul, L.; Nalin, R.; Jarrin, C.; Chardon, P.; Marteau, P.; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–211. [Google Scholar] [CrossRef]
  38. Mosca, A.; Leclerc, M.; Hugot, J.P. Gut Microbiota Diversity and Human Diseases: Should We Reintroduce Key Predators in Our Ecosystem? Front. Microbiol. 2016, 7, 455. [Google Scholar] [CrossRef]
  39. Sartor, R.B.; Mazmanian, S.K. Intestinal Microbes in Inflammatory Bowel Diseases. Am. J. Gastroenterol. Suppl. 2012, 1, 15–21. [Google Scholar] [CrossRef]
  40. Thomas, F.; Hehemann, J.H.; Rebuffet, E.; Czjzek, M.; Michel, G. Environmental and gut bacteroidetes: The food connection. Front. Microbiol. 2011, 2, 93. [Google Scholar] [CrossRef]
  41. Kim, Y.S.; Milner, J.A. Dietary modulation of colon cancer risk. J. Nutr. 2007, 137 (Suppl. 11), 2576S–2579S. [Google Scholar] [CrossRef]
  42. Mazmanian, S.K.; Round, J.L.; Kasper, D.L. A microbial symbiosis factor prevents intestinal inflammatory disease. Nature 2008, 453, 620–625. [Google Scholar] [CrossRef] [PubMed]
  43. Lo Presti, A.; Zorzi, F.; Del Chierico, F.; Altomare, A.; Cocca, S.; Avola, A.; De Biasio, F.; Russo, A.; Cella, E.; Reddel, S.; et al. Fecal and Mucosal Microbiota Profiling in Irritable Bowel Syndrome and Inflammatory Bowel Disease. Front. Microbiol. 2019, 10, 1655. [Google Scholar] [CrossRef] [PubMed]
  44. Labbé, A.; Ganopolsky, J.G.; Martoni, C.J.; Prakash, S.; Jones, M.L. Bacterial bile metabolising gene abundance in Crohn’s, ulcerative colitis and type 2 diabetes metagenomes. PLoS ONE 2014, 9, e115175. [Google Scholar] [CrossRef] [PubMed]
  45. Hedin, C.R.; McCarthy, N.E.; Louis, P.; Farquharson, F.M.; McCartney, S.; Taylor, K.; Prescott, N.J.; Murrells, T.; Stagg, A.J.; Whelan, K.; et al. Altered intestinal microbiota and blood T cell phenotype are shared by patients with Crohn’s disease and their unaffected siblings. Gut 2014, 63, 1578–1586. [Google Scholar] [CrossRef]
  46. Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef]
  47. Wlodarska, M.; Willing, B.P.; Bravo, D.M.; Finlay, B.B. Phytonutrient diet supplementation promotes beneficial Clostridia species and intestinal mucus secretion resulting in protection against enteric infection. Sci. Rep. 2015, 5, 9253. [Google Scholar] [CrossRef]
  48. Muñiz Pedrogo, D.A.; Chen, J.; Hillmann, B.; Jeraldo, P.; Al-Ghalith, G.; Taneja, V.; Davis, J.M.; Knights, D.; Nelson, H.; Faubion, W.A.; et al. An Increased Abundance of Clostridiaceae Characterizes Arthritis in Inflammatory Bowel Disease and Rheumatoid Arthritis: A Cross-sectional Study. Inflamm. Bowel. Dis. 2019, 25, 902–913. [Google Scholar] [CrossRef]
  49. Scarpa, M.; Grillo, A.; Faggian, D.; Ruffolo, C.; Bonello, E.; D’Incà, R.; Castagliuolo, I.; Angriman, I. Relationship between mucosa-associated microbiota and inflammatory parameters in the ileal pouch after restorative proctocolectomy for ulcerative colitis. Surgery 2011, 150, 56–67. [Google Scholar] [CrossRef]
  50. Imhann, F.; Vich Vila, A.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
  51. Maukonen, J.; Kolho, K.L.; Paasela, M.; Honkanen, J.; Klemetti, P.; Vaarala, O.; Saarela, M. Altered Fecal Microbiota in Paediatric Inflammatory Bowel Disease. J. Crohn’s Colitis 2015, 9, 1088–1095. [Google Scholar] [CrossRef]
  52. Pittayanon, R.; Lau, J.T.; Leontiadis, G.I.; Tse, F.; Yuan, Y.; Surette, M.; Moayyedi, P. Differences in Gut Microbiota in Patients With vs Without Inflammatory Bowel Diseases: A Systematic Review. Gastroenterology 2020, 158, 930–946.e931. [Google Scholar] [CrossRef] [PubMed]
  53. Lee, T.; Clavel, T.; Smirnov, K.; Schmidt, A.; Lagkouvardos, I.; Walker, A.; Lucio, M.; Michalke, B.; Schmitt-Kopplin, P.; Fedorak, R.; et al. Oral versus intravenous iron replacement therapy distinctly alters the gut microbiota and metabolome in patients with IBD. Gut 2017, 66, 863–871. [Google Scholar] [CrossRef] [PubMed]
  54. Baldelli, V.; Scaldaferri, F.; Putignani, L.; Del Chierico, F. The Role of Enterobacteriaceae in Gut Microbiota Dysbiosis in Inflammatory Bowel Diseases. Microorganisms 2021, 9, 697. [Google Scholar] [CrossRef] [PubMed]
  55. Siniagina, M.N.; Markelova, M.I.; Boulygina, E.A.; Laikov, A.V.; Khusnutdinova, D.R.; Abdulkhakov, S.R.; Danilova, N.A.; Odintsova, A.H.; Abdulkhakov, R.A.; Grigoryeva, T.V. Diversity and Adaptations of Escherichia coli Strains: Exploring the Intestinal Community in Crohn’s Disease Patients and Healthy Individuals. Microorganisms 2021, 9, 1299. [Google Scholar] [CrossRef]
  56. Heidarian, F.; Noormohammadi, Z.; Aghdaei, H.A.; Alebouyeh, M. Relative abundance of streptococcus spp. and its association with disease activity in inflammatory bowel disease patients compared with controls. Arch. Clin. Infect. Dis. 2017, 12, e57291. [Google Scholar] [CrossRef]
  57. Vich Vila, A.; Imhann, F.; Collij, V.; Jankipersadsing, S.A.; Gurry, T.; Mujagic, Z.; Kurilshikov, A.; Bonder, M.J.; Jiang, X.; Tigchelaar, E.F.; et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome. Sci. Transl. Med. 2018, 10, eaap8914. [Google Scholar] [CrossRef]
  58. Shiga, H.; Kajiura, T.; Shinozaki, J.; Takagi, S.; Kinouchi, Y.; Takahashi, S.; Negoro, K.; Endo, K.; Kakuta, Y.; Suzuki, M.; et al. Changes of faecal microbiota in patients with Crohn’s disease treated with an elemental diet and total parenteral nutrition. Dig. Liver Dis. 2012, 44, 736–742. [Google Scholar] [CrossRef]
  59. Lewis, J.D.; Chen, E.Z.; Baldassano, R.N.; Otley, A.R.; Griffiths, A.M.; Lee, D.; Bittinger, K.; Bailey, A.; Friedman, E.S.; Hoffmann, C.; et al. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn’s Disease. Cell Host Microbe 2015, 18, 489–500. [Google Scholar] [CrossRef]
  60. Wang, W.; Chen, L.; Zhou, R.; Wang, X.; Song, L.; Huang, S.; Wang, G.; Xia, B. Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J. Clin. Microbiol. 2014, 52, 398–406. [Google Scholar] [CrossRef]
  61. Dinis, M.; Plainvert, C.; Kovarik, P.; Longo, M.; Fouet, A.; Poyart, C. The innate immune response elicited by Group A Streptococcus is highly variable among clinical isolates and correlates with the emm type. PLoS ONE 2014, 9, e101464. [Google Scholar] [CrossRef]
  62. Chirouze, C.; Athan, E.; Alla, F.; Chu, V.H.; Ralph Corey, G.; Selton-Suty, C.; Erpelding, M.L.; Miro, J.M.; Olaison, L.; Hoen, B.; et al. Enterococcal endocarditis in the beginning of the 21st century: Analysis from the International Collaboration on Endocarditis-Prospective Cohort Study. Clin. Microbiol. Infect. 2013, 19, 1140–1147. [Google Scholar] [CrossRef] [PubMed]
  63. Khanum, I.; Anwar, S.; Farooque, A. Enterococcal Meningitis/Ventriculitis: A Tertiary Care Experience. Asian J. Neurosurg. 2019, 14, 102–105. [Google Scholar] [CrossRef] [PubMed]
  64. Tang, Q.; Hao, Y.; Wang, L.; Lu, C.; Li, M.; Si, Z.; Wu, X.; Lu, Z. Characterization of a bacterial strain Lactobacillus paracasei LP10266 recovered from an endocarditis patient in Shandong, China. BMC Microbiol. 2021, 21, 183. [Google Scholar] [CrossRef] [PubMed]
  65. Frolova, L.; Drastich, P.; Rossmann, P.; Klimesova, K.; Tlaskalova-Hogenova, H. Expression of Toll-like receptor 2 (TLR2), TLR4, and CD14 in biopsy samples of patients with inflammatory bowel diseases: Upregulated expression of TLR2 in terminal ileum of patients with ulcerative colitis. J. Histochem. Cytochem. 2008, 56, 267–274. [Google Scholar] [CrossRef]
  66. Balish, E.; Warner, T. Enterococcus faecalis induces inflammatory bowel disease in interleukin-10 knockout mice. Am. J. Pathol. 2002, 160, 2253–2257. [Google Scholar] [CrossRef]
  67. Golińska, E.; Tomusiak, A.; Gosiewski, T.; Więcek, G.; Machul, A.; Mikołajczyk, D.; Bulanda, M.; Heczko, P.B.; Strus, M. Virulence factors of Enterococcus strains isolated from patients with inflammatory bowel disease. World J. Gastroenterol. 2013, 19, 3562–3572. [Google Scholar] [CrossRef]
  68. Swidsinski, A.; Loening-Baucke, V.; Vaneechoutte, M.; Doerffel, Y. Active Crohn’s disease and ulcerative colitis can be specifically diagnosed and monitored based on the biostructure of the fecal flora. Inflamm. Bowel. Dis. 2008, 14, 147–161. [Google Scholar] [CrossRef]
  69. Sokol, H.; Seksik, P.; Furet, J.P.; Firmesse, O.; Nion-Larmurier, I.; Beaugerie, L.; Cosnes, J.; Corthier, G.; Marteau, P.; Doré, J. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm. Bowel. Dis. 2009, 15, 1183–1189. [Google Scholar] [CrossRef]
  70. Vieira-Silva, S.; Sabino, J.; Valles-Colomer, M.; Falony, G.; Kathagen, G.; Caenepeel, C.; Cleynen, I.; van der Merwe, S.; Vermeire, S.; Raes, J. Quantitative microbiome profiling disentangles inflammation- and bile duct obstruction-associated microbiota alterations across PSC/IBD diagnoses. Nat. Microbiol. 2019, 4, 1826–1831. [Google Scholar] [CrossRef]
  71. Buffet-Bataillon, S.; Bouguen, G.; Fleury, F.; Cattoir, V.; Le Cunff, Y. Gut microbiota analysis for prediction of clinical relapse in Crohn’s disease. Sci. Rep. 2022, 12, 19929. [Google Scholar] [CrossRef]
  72. Zhou, Y.; Xu, Z.Z.; He, Y.; Yang, Y.; Liu, L.; Lin, Q.; Nie, Y.; Li, M.; Zhi, F.; Liu, S.; et al. Gut Microbiota Offers Universal Biomarkers across Ethnicity in Inflammatory Bowel Disease Diagnosis and Infliximab Response Prediction. mSystems 2018, 3, e00188-17. [Google Scholar] [CrossRef] [PubMed]
  73. Xue, A.J.; Miao, S.J.; Sun, H.; Qiu, X.X.; Wang, S.N.; Wang, L.; Ye, Z.Q.; Zheng, C.F.; Huang, Z.H.; Wang, Y.H.; et al. Intestinal dysbiosis in pediatric Crohn’s disease patients with. World J. Gastroenterol. 2020, 26, 3098–3109. [Google Scholar] [CrossRef] [PubMed]
  74. Papa, E.; Docktor, M.; Smillie, C.; Weber, S.; Preheim, S.P.; Gevers, D.; Giannoukos, G.; Ciulla, D.; Tabbaa, D.; Ingram, J.; et al. Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PLoS ONE 2012, 7, e39242. [Google Scholar] [CrossRef]
  75. Tedjo, D.I.; Smolinska, A.; Savelkoul, P.H.; Masclee, A.A.; van Schooten, F.J.; Pierik, M.J.; Penders, J.; Jonkers, D.M. The fecal microbiota as a biomarker for disease activity in Crohn’s disease. Sci. Rep. 2016, 6, 35216. [Google Scholar] [CrossRef] [PubMed]
  76. Sha, S.; Xu, B.; Wang, X.; Zhang, Y.; Wang, H.; Kong, X.; Zhu, H.; Wu, K. The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn. Microbiol. Infect. Dis. 2013, 75, 245–251. [Google Scholar] [CrossRef]
  77. Arumugam, M.; Raes, J.; Pelletier, E.; Le Paslier, D.; Yamada, T.; Mende, D.R.; Fernandes, G.R.; Tap, J.; Bruls, T.; Batto, J.M.; et al. Enterotypes of the human gut microbiome. Nature 2011, 473, 174–180. [Google Scholar] [CrossRef]
  78. Barron, M.R.; Sovacool, K.L.; Abernathy-Close, L.; Vendrov, K.C.; Standke, A.K.; Bergin, I.L.; Schloss, P.D.; Young, V.B. Intestinal Inflammation Reversibly Alters the Microbiota to Drive Susceptibility to Clostridioides difficile Colonization in a Mouse Model of Colitis. mBio 2022, 13, e0190422. [Google Scholar] [CrossRef]
  79. Dunn, K.A.; Moore-Connors, J.; MacIntyre, B.; Stadnyk, A.W.; Thomas, N.A.; Noble, A.; Mahdi, G.; Rashid, M.; Otley, A.R.; Bielawski, J.P.; et al. Early Changes in Microbial Community Structure Are Associated with Sustained Remission After Nutritional Treatment of Pediatric Crohn’s Disease. Inflamm. Bowel. Dis. 2016, 22, 2853–2862. [Google Scholar] [CrossRef]
  80. Blekhman, R.; Goodrich, J.K.; Huang, K.; Sun, Q.; Bukowski, R.; Bell, J.T.; Spector, T.D.; Keinan, A.; Ley, R.E.; Gevers, D.; et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015, 16, 191. [Google Scholar] [CrossRef]
  81. Bonder, M.J.; Kurilshikov, A.; Tigchelaar, E.F.; Mujagic, Z.; Imhann, F.; Vila, A.V.; Deelen, P.; Vatanen, T.; Schirmer, M.; Smeekens, S.P.; et al. The effect of host genetics on the gut microbiome. Nat. Genet. 2016, 48, 1407–1412. [Google Scholar] [CrossRef]
  82. Goodrich, J.K.; Davenport, E.R.; Beaumont, M.; Jackson, M.A.; Knight, R.; Ober, C.; Spector, T.D.; Bell, J.T.; Clark, A.G.; Ley, R.E. Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 2016, 19, 731–743. [Google Scholar] [CrossRef]
  83. Turpin, W.; Espin-Garcia, O.; Xu, W.; Silverberg, M.S.; Kevans, D.; Smith, M.I.; Guttman, D.S.; Griffiths, A.; Panaccione, R.; Otley, A.; et al. Association of host genome with intestinal microbial composition in a large healthy cohort. Nat. Genet. 2016, 48, 1413–1417. [Google Scholar] [CrossRef] [PubMed]
  84. Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 107. [Google Scholar] [CrossRef] [PubMed]
  85. Tong, M.; McHardy, I.; Ruegger, P.; Goudarzi, M.; Kashyap, P.C.; Haritunians, T.; Li, X.; Graeber, T.G.; Schwager, E.; Huttenhower, C.; et al. Reprograming of gut microbiome energy metabolism by the FUT2 Crohn’s disease risk polymorphism. ISME J. 2014, 8, 2193–2206. [Google Scholar] [CrossRef] [PubMed]
  86. The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000164061-BSN (accessed on 20 December 2022).
  87. Gribble, F.M.; Reimann, F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat. Rev. Endocrinol. 2019, 15, 226–237. [Google Scholar] [CrossRef]
  88. Bellono, N.W.; Bayrer, J.R.; Leitch, D.B.; Castro, J.; Zhang, C.; O’Donnell, T.A.; Brierley, S.M.; Ingraham, H.A.; Julius, D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell 2017, 170, 185–198.e116. [Google Scholar] [CrossRef]
  89. Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef]
  90. Lebrun, L.J.; Lenaerts, K.; Kiers, D.; Pais de Barros, J.P.; Le Guern, N.; Plesnik, J.; Thomas, C.; Bourgeois, T.; Dejong, C.H.C.; Kox, M.; et al. Enteroendocrine L Cells Sense LPS after Gut Barrier Injury to Enhance GLP-1 Secretion. Cell Rep. 2017, 21, 1160–1168. [Google Scholar] [CrossRef]
  91. Chimerel, C.; Emery, E.; Summers, D.K.; Keyser, U.; Gribble, F.M.; Reimann, F. Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep. 2014, 9, 1202–1208. [Google Scholar] [CrossRef]
  92. Brighton, C.A.; Rievaj, J.; Kuhre, R.E.; Glass, L.L.; Schoonjans, K.; Holst, J.J.; Gribble, F.M.; Reimann, F. Bile Acids Trigger GLP-1 Release Predominantly by Accessing Basolaterally Located G Protein-Coupled Bile Acid Receptors. Endocrinology 2015, 156, 3961–3970. [Google Scholar] [CrossRef]
  93. Kara, S.; Pirela-Morillo, G.A.; Gilliam, C.T.; Wilson, G.D. Identification of novel susceptibility genes associated with seven autoimmune disorders using whole genome molecular interaction networks. J. Autoimmun. 2019, 97, 48–58. [Google Scholar] [CrossRef] [PubMed]
  94. Grivennikov, S.; Karin, E.; Terzic, J.; Mucida, D.; Yu, G.Y.; Vallabhapurapu, S.; Scheller, J.; Rose-John, S.; Cheroutre, H.; Eckmann, L.; et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells and development of colitis-associated cancer. Cancer Cell 2009, 15, 103–113. [Google Scholar] [CrossRef] [PubMed]
  95. Takeda, K.; Kaisho, T.; Yoshida, N.; Takeda, J.; Kishimoto, T.; Akira, S. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: Generation and characterization of T cell-specific Stat3-deficient mice. J. Immunol. 1998, 161, 4652–4660. [Google Scholar] [CrossRef] [PubMed]
  96. Zhao, Y.; Chen, F.; Wu, W.; Sun, M.; Bilotta, A.J.; Yao, S.; Xiao, Y.; Huang, X.; Eaves-Pyles, T.D.; Golovko, G.; et al. GPR43 mediates microbiota metabolite SCFA regulation of antimicrobial peptide expression in intestinal epithelial cells via activation of mTOR and STAT3. Mucosal. Immunol. 2018, 11, 752–762. [Google Scholar] [CrossRef]
  97. Franke, A.; Hampe, J.; Rosenstiel, P.; Becker, C.; Wagner, F.; Häsler, R.; Little, R.D.; Huse, K.; Ruether, A.; Balschun, T.; et al. Systematic association mapping identifies NELL1 as a novel IBD disease gene. PLoS ONE 2007, 2, e691. [Google Scholar] [CrossRef] [PubMed]
  98. Dolpady, J.; Sorini, C.; Di Pietro, C.; Cosorich, I.; Ferrarese, R.; Saita, D.; Clementi, M.; Canducci, F.; Falcone, M. Oral Probiotic VSL#3 Prevents Autoimmune Diabetes by Modulating Microbiota and Promoting Indoleamine 2,3-Dioxygenase-Enriched Tolerogenic Intestinal Environment. J. Diabetes Res. 2016, 2016, 7569431. [Google Scholar] [CrossRef]
  99. Kouchaki, E.; Tamtaji, O.R.; Salami, M.; Bahmani, F.; Daneshvar Kakhaki, R.; Akbari, E.; Tajabadi-Ebrahimi, M.; Jafari, P.; Asemi, Z. Clinical and metabolic response to probiotic supplementation in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled trial. Clin. Nutr. 2017, 36, 1245–1249. [Google Scholar] [CrossRef]
  100. Granito, A.; Muratori, P.; Muratori, L. Editorial: Gut microbiota profile in patients with autoimmune hepatitis-a clue for adjunctive probiotic therapy? Aliment. Pharmacol. Ther. 2020, 52, 392–394. [Google Scholar] [CrossRef]
  101. Abhari, K.; Shekarforoush, S.S.; Hosseinzadeh, S.; Nazifi, S.; Sajedianfard, J.; Eskandari, M.H. The effects of orally administered Bacillus coagulans and inulin on prevention and progression of rheumatoid arthritis in rats. Food Nutr. Res. 2016, 60, 30876. [Google Scholar] [CrossRef]
  102. Martín, R.; Miquel, S.; Benevides, L.; Bridonneau, C.; Robert, V.; Hudault, S.; Chain, F.; Berteau, O.; Azevedo, V.; Chatel, J.M.; et al. Functional characterization of novel Faecalibacterium prausnitzii strains isolated from healthy volunteers: A step forward in the use of F. prausnitzii as a next-generation probiotic. Front. Microbiol. 2017, 8, 1226. [Google Scholar] [CrossRef]
  103. Yoon, H.S.; Cho, C.H.; Yun, M.S.; Jang, S.J.; You, H.J.; Kim, J.H.; Han, D.; Cha, K.H.; Moon, S.H.; Lee, K.; et al. Akkermansia muciniphila secretes a glucagon-like peptide-1-inducing protein that improves glucose homeostasis and ameliorates metabolic disease in mice. Nat. Microbiol. 2021, 6, 563–573. [Google Scholar] [CrossRef] [PubMed]
  104. Fehily, S.R.; Basnayake, C.; Wright, E.K.; Kamm, M.A. Fecal microbiota transplantation therapy in Crohn’s disease: Systematic review. J. Gastroenterol. Hepatol. 2021, 36, 2672–2686. [Google Scholar] [CrossRef] [PubMed]
  105. Caporaso, J.G.; Kuczynski, J.; Stombaugh, J.; Bittinger, K.; Bushman, F.D.; Costello, E.K.; Fierer, N.; Peña, A.G.; Goodrich, J.K.; Gordon, J.I.; et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 2010, 7, 335–336. [Google Scholar] [CrossRef]
  106. Quast, C.; Pruesse, E.; Yilmaz, P.; Gerken, J.; Schweer, T.; Yarza, P.; Peplies, J.; Glöckner, F.O. The SILVA ribosomal RNA gene database project: Improved data processing and web-based tools. Nucleic. Acids Res. 2013, 41, D590–D596. [Google Scholar] [CrossRef]
  107. Storm, N.; Darnhofer-Patel, B.; van den Boom, D.; Rodi, C.P. MALDI-TOF mass spectrometry-based SNP genotyping. Methods Mol. Biol. 2003, 212, 241–262. [Google Scholar] [CrossRef] [PubMed]
  108. Revelle, W. psych: Procedures for Personality and Psychological Research; Northwestern University: Evanston, IL, USA, 2018. [Google Scholar]
  109. Holmes, I.; Harris, K.; Quince, C. Dirichlet multinomial mixtures: Generative models for microbial metagenomics. PLoS ONE 2012, 7, e30126. [Google Scholar] [CrossRef] [PubMed]
  110. Glas, J.; Konrad, A.; Schmechel, S.; Dambacher, J.; Seiderer, J.; Schroff, F.; Wetzke, M.; Roeske, D.; Török, H.P.; Tonenchi, L.; et al. The ATG16L1 gene variants rs2241879 and rs2241880 (T300A) are strongly associated with susceptibility to Crohn’s disease in the German population. Am. J. Gastroenterol. 2008, 103, 682–691. [Google Scholar] [CrossRef]
  111. Roberts, R.L.; Gearry, R.B.; Hollis-Moffatt, J.E.; Miller, A.L.; Reid, J.; Abkevich, V.; Timms, K.M.; Gutin, A.; Lanchbury, J.S.; Merriman, T.R.; et al. IL23R R381Q and ATG16L1 T300A are strongly associated with Crohn’s disease in a study of New Zealand Caucasians with inflammatory bowel disease. Am. J. Gastroenterol. 2007, 102, 2754–2761. [Google Scholar] [CrossRef]
  112. Kee, B.P.; Ng, J.G.; Ng, C.C.; Hilmi, I.; Goh, K.L.; Chua, K.H. Genetic polymorphisms of ATG16L1 and IRGM genes in Malaysian patients with Crohn’s disease. J. Dig. Dis. 2020, 21, 29–37. [Google Scholar] [CrossRef]
  113. Quaranta, M.; Burden, A.D.; Griffiths, C.E.; Worthington, J.; Barker, J.N.; Trembath, R.C.; Capon, F. Differential contribution of CDKAL1 variants to psoriasis, Crohn’s disease and type II diabetes. Genes Immun. 2009, 10, 654–658. [Google Scholar] [CrossRef]
  114. Anderson, C.A.; Massey, D.C.; Barrett, J.C.; Prescott, N.J.; Tremelling, M.; Fisher, S.A.; Gwilliam, R.; Jacob, J.; Nimmo, E.R.; Drummond, H.; et al. Investigation of Crohn’s disease risk loci in ulcerative colitis further defines their molecular relationship. Gastroenterology 2009, 136, 523–529.e523. [Google Scholar] [CrossRef] [PubMed]
  115. Hradsky, O.; Dusatkova, P.; Lenicek, M.; Bronsky, J.; Duricova, D.; Nevoral, J.; Vitek, L.; Lukas, M.; Cinek, O. Two independent genetic factors responsible for the associations of the IBD5 locus with Crohn’s disease in the Czech population. Inflamm. Bowel. Dis. 2011, 17, 1523–1529. [Google Scholar] [CrossRef]
  116. Tsianos, V.E. Study of the Genetic Polymorphisms of IBD in NW Greece. Ph.D. Thesis, University of Ioannina, Ioannina, Greece, 2019. [Google Scholar]
  117. Zhao, X.D.; Shen, F.C.; Zhang, H.J.; Shen, X.Y.; Wang, Y.M.; Yang, X.Z.; Tu, H.M.; Tai, Y.H.; Shi, R.H. Association of interleukin-23 receptor gene polymorphisms with susceptibility and phenotypes of inflammatory bowel diseases in Jiangsu Han population. Zhonghua Nei Ke Za Zhi 2011, 50, 935–941. [Google Scholar] [PubMed]
  118. Chen, Z.Y.; Zhi, F.C.; Zhi, J. Preliminary study on relationship between gene polymorphisms of interleukin-23 receptor and inflammatory bowel disease. Chin. J. Dig. 2008, 28, 369–372. [Google Scholar]
  119. Glas, J.; Seiderer, J.; Wetzke, M.; Konrad, A.; Török, H.P.; Schmechel, S.; Tonenchi, L.; Grassl, C.; Dambacher, J.; Pfennig, S.; et al. rs1004819 is the main disease-associated IL23R variant in German Crohn’s disease patients: Combined analysis of IL23R, CARD15, and OCTN1/2 variants. PLoS ONE 2007, 2, e819. [Google Scholar] [CrossRef]
  120. Csöngei, V.; Járomi, L.; Sáfrány, E.; Sipeky, C.; Magyari, L.; Faragó, B.; Bene, J.; Polgár, N.; Lakner, L.; Sarlós, P.; et al. Interaction of the major inflammatory bowel disease susceptibility alleles in Crohn’s disease patients. World J. Gastroenterol. 2010, 16, 176–183. [Google Scholar] [CrossRef] [PubMed]
  121. Hong, J.; Yang, H.R.; Moon, J.S.; Chang, J.Y.; Ko, J.S. Association of IL23R Variants With Crohn’s Disease in Korean Children. Front. Pediatr. 2019, 7, 472. [Google Scholar] [CrossRef]
  122. Oliver, J.; Rueda, B.; López-Nevot, M.A.; Gómez-García, M.; Martín, J. Replication of an association between IL23R gene polymorphism with inflammatory bowel disease. Clin. Gastroenterol. Hepatol. 2007, 5, 977–981.e2. [Google Scholar] [CrossRef]
  123. Laserna-Mendieta, E.J.; Salvador-Martín, S.; Arias, A.; López-Cauce, B.; Marín-Jiménez, I.; Menchén, L.A.; Marín-Rubio, L.; Ontañón Rodríguez, J.; López-Fernández, L.A.; Lucendo, A.J. Single nucleotide polymorphisms in ADAM17, IL23R and SLCO1C1 genes protect against infliximab failure in adults with Crohn’s disease. BioMed. Pharmacother. 2023, 159, 114225. [Google Scholar] [CrossRef]
  124. Zhu, Y.; Jiang, H.; Chen, Z.; Lu, B.; Li, J.; Shen, X. Genetic association between IL23R rs11209026 and rs10889677 polymorphisms and risk of Crohn’s disease and ulcerative colitis: Evidence from 41 studies. Inflamm. Res. 2020, 69, 87–103. [Google Scholar] [CrossRef]
  125. Netz, U.; Carter, J.V.; Eichenberger, M.R.; Dryden, G.W.; Pan, J.; Rai, S.N.; Galandiuk, S. Genetic polymorphisms predict response to anti-tumor necrosis factor treatment in Crohn’s disease. World J. Gastroenterol. 2017, 23, 4958–4967. [Google Scholar] [CrossRef] [PubMed]
  126. Lu, Y.; Li, C.Y.; Lin, S.S.; Yuan, P. rs13361189 polymorphism may contribute to susceptibility to Crohn’s disease: A meta-analysis. Exp. Ther. Med. 2014, 8, 607–613. [Google Scholar] [CrossRef] [PubMed]
  127. Kline, B.P.; Weaver, T.; Brinton, D.L.; Deiling, S.; Yochum, G.S.; Berg, A.S.; Koltun, W.A. Clinical and Genetic Factors Associated With Complications After Crohn’s Ileocolectomy. Dis. Colon. Rectum. 2020, 63, 357–364. [Google Scholar] [CrossRef] [PubMed]
  128. Latiano, A.; Palmieri, O.; Cucchiara, S.; Castro, M.; D’Incà, R.; Guariso, G.; Dallapiccola, B.; Valvano, M.R.; Latiano, T.; Andriulli, A.; et al. Polymorphism of the IRGM gene might predispose to fistulizing behavior in Crohn’s disease. Am. J. Gastroenterol. 2009, 104, 110–116. [Google Scholar] [CrossRef] [PubMed]
  129. Lu, X.C.; Tao, Y.; Wu, C.; Zhao, P.L.; Li, K.; Zheng, J.Y.; Li, L.X. Association between variants of the autophagy related gene—IRGM and susceptibility to Crohn’s disease and ulcerative colitis: A meta-analysis. PLoS ONE 2013, 8, e80602. [Google Scholar] [CrossRef]
  130. Umeno, J.; Asano, K.; Matsushita, T.; Matsumoto, T.; Kiyohara, Y.; Iida, M.; Nakamura, Y.; Kamatani, N.; Kubo, M. Meta-analysis of published studies identified eight additional common susceptibility loci for Crohn’s disease and ulcerative colitis. Inflamm. Bowel. Dis. 2011, 17, 2407–2415. [Google Scholar] [CrossRef]
  131. Kugathasan, S.; Baldassano, R.N.; Bradfield, J.P.; Sleiman, P.M.; Imielinski, M.; Guthery, S.L.; Cucchiara, S.; Kim, C.E.; Frackelton, E.C.; Annaiah, K.; et al. Loci on 20q13 and 21q22 are associated with pediatric-onset inflammatory bowel disease. Nat. Genet. 2008, 40, 1211–1215. [Google Scholar] [CrossRef]
  132. Latiano, A.; Palmieri, O.; Latiano, T.; Corritore, G.; Bossa, F.; Martino, G.; Biscaglia, G.; Scimeca, D.; Valvano, M.R.; Pastore, M.; et al. Investigation of multiple susceptibility loci for inflammatory bowel disease in an Italian cohort of patients. PLoS ONE 2011, 6, e22688. [Google Scholar] [CrossRef]
  133. Ditrich, F.; Blümel, S.; Biedermann, L.; Fournier, N.; Rossel, J.B.; Ellinghaus, D.; Franke, A.; Stange, E.F.; Rogler, G.; Scharl, M.; et al. Genetic risk factors predict disease progression in Crohn’s disease patients of the Swiss inflammatory bowel disease cohort. Ther. Adv. Gastroenterol. 2020, 13, 1756284820959252. [Google Scholar] [CrossRef]
  134. Cruz-Romero, C.; Guo, A.; Bradley, W.F.; Vicentini, J.R.T.; Yajnik, V.; Gee, M.S. Novel Associations Between Genome-Wide Single Nucleotide Polymorphisms and MR Enterography Features in Crohn’s Disease Patients. J. Magn. Reson. Imaging 2021, 53, 132–138. [Google Scholar] [CrossRef]
  135. Ferguson, L.R.; Philpott, M.; Dryland, P. Nutrigenomics in the whole-genome scanning era: Crohn’s disease as example. Cell Mol. Life Sci. 2007, 64, 3105–3118. [Google Scholar] [CrossRef] [PubMed]
  136. Ho, P.; Bruce, I.N.; Silman, A.; Symmons, D.; Newman, B.; Young, H.; Griffiths, C.E.; John, S.; Worthington, J.; Barton, A. Evidence for common genetic control in pathways of inflammation for Crohn’s disease and psoriatic arthritis. Arthritis Rheum. 2005, 52, 3596–3602. [Google Scholar] [CrossRef] [PubMed]
  137. Weersma, R.K.; Stokkers, P.C.; van Bodegraven, A.A.; van Hogezand, R.A.; Verspaget, H.W.; de Jong, D.J.; van der Woude, C.J.; Oldenburg, B.; Linskens, R.K.; Festen, E.A.; et al. Molecular prediction of disease risk and severity in a large Dutch Crohn’s disease cohort. Gut 2009, 58, 388–395. [Google Scholar] [CrossRef]
  138. Sarlos, P.; Varszegi, D.; Csongei, V.; Magyari, L.; Jaromi, L.; Nagy, L.; Melegh, B. Susceptibility to ulcerative colitis in Hungarian patients determined by gene-gene interactions. World J. Gastroenterol. 2014, 20, 219–227. [Google Scholar] [CrossRef]
  139. Garcia-Carbonell, R.; Yao, S.J.; Das, S.; Guma, M. Dysregulation of Intestinal Epithelial Cell RIPK Pathways Promotes Chronic Inflammation in the IBD Gut. Front. Immunol. 2019, 10, 1094. [Google Scholar] [CrossRef] [PubMed]
  140. Neuman, M.G.; Nanau, R.M. Single-nucleotide polymorphisms in inflammatory bowel disease. Transl. Res. 2012, 160, 45–64. [Google Scholar] [CrossRef]
  141. Waterman, M.; Xu, W.; Stempak, J.M.; Milgrom, R.; Bernstein, C.N.; Griffiths, A.M.; Greenberg, G.R.; Steinhart, A.H.; Silverberg, M.S. Distinct and overlapping genetic loci in Crohn’s disease and ulcerative colitis: Correlations with pathogenesis. Inflamm. Bowel. Dis. 2011, 17, 1936–1942. [Google Scholar] [CrossRef]
  142. Kakuta, Y.; Kawai, Y.; Naito, T.; Hirano, A.; Umeno, J.; Fuyuno, Y.; Liu, Z.; Li, D.; Nakano, T.; Izumiyama, Y.; et al. A Genome-wide Association Study Identifying RAP1A as a Novel Susceptibility Gene for Crohn’s Disease in Japanese Individuals. J. Crohn’s Colitis 2019, 13, 648–658. [Google Scholar] [CrossRef]
  143. Glas, J.; Seiderer, J.; Pasciuto, G.; Tillack, C.; Diegelmann, J.; Pfennig, S.; Konrad, A.; Schmechel, S.; Wetzke, M.; Török, H.P.; et al. rs224136 on chromosome 10q21.1 and variants in PHOX2B, NCF4, and FAM92B are not major genetic risk factors for susceptibility to Crohn’s disease in the German population. Am. J. Gastroenterol. 2009, 104, 665–672. [Google Scholar] [CrossRef]
  144. Cagliani, R.; Pozzoli, U.; Forni, D.; Cassinotti, A.; Fumagalli, M.; Giani, M.; Fichera, M.; Lombardini, M.; Ardizzone, S.; Asselta, R.; et al. Crohn’s disease loci are common targets of protozoa-driven selection. Mol. Biol. Evol. 2013, 30, 1077–1087. [Google Scholar] [CrossRef]
  145. Laing, B.; Han, D.Y.; Ferguson, L.R. Candidate genes involved in beneficial or adverse responses to commonly eaten brassica vegetables in a New Zealand Crohn’s disease cohort. Nutrients 2013, 5, 5046–5064. [Google Scholar] [CrossRef]
  146. Fischer, A.; Nothnagel, M.; Franke, A.; Jacobs, G.; Saadati, H.R.; Gaede, K.I.; Rosenstiel, P.; Schürmann, M.; Müller-Quernheim, J.; Schreiber, S.; et al. Association of inflammatory bowel disease risk loci with sarcoidosis, and its acute and chronic subphenotypes. Eur. Respir. J. 2011, 37, 610–616. [Google Scholar] [CrossRef] [PubMed]
  147. Mieth, B.; Rozier, A.; Rodriguez, J.A.; Höhne, M.M.C.; Görnitz, N.; Müller, K.R. DeepCOMBI: Explainable artificial intelligence for the analysis and discovery in genome-wide association studies. NAR Genom. Bioinform. 2021, 3, lqab065. [Google Scholar] [CrossRef] [PubMed]
  148. Connelly, T.M.; Berg, A.S.; Harris, L.R.; Brinton, D.L.; Hegarty, J.P.; Deiling, S.M.; Stewart, D.B.; Koltun, W.A. Ulcerative colitis neoplasia is not associated with common inflammatory bowel disease single-nucleotide polymorphisms. Surgery 2014, 156, 253–262. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Analysis of gut microbiota taxonomic composition of CD patients and healthy volunteers. (A)—Number of OTUs and Shannon’s diversity index per group. (B)—Most abundant bacterial families significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test).
Figure 1. Analysis of gut microbiota taxonomic composition of CD patients and healthy volunteers. (A)—Number of OTUs and Shannon’s diversity index per group. (B)—Most abundant bacterial families significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test).
Ijms 24 07998 g001
Figure 2. Analysis of gut microbiota taxonomic composition of CD patients with different disease activity. (A)—Statistically significant Spearman’s correlations between CDAI and gut microbiota taxonomic composition (p < 0.05). a—Bacterial phyla, b—Bacterial families. (B)—Most abundant bacterial families significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test with Benjamini-Hochberg correction for multiple comparisons).
Figure 2. Analysis of gut microbiota taxonomic composition of CD patients with different disease activity. (A)—Statistically significant Spearman’s correlations between CDAI and gut microbiota taxonomic composition (p < 0.05). a—Bacterial phyla, b—Bacterial families. (B)—Most abundant bacterial families significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test with Benjamini-Hochberg correction for multiple comparisons).
Ijms 24 07998 g002
Figure 3. Principal component analysis based on bacterial composition of gut microbiota on family level.
Figure 3. Principal component analysis based on bacterial composition of gut microbiota on family level.
Ijms 24 07998 g003
Figure 4. Top four driver families in different community types of gut microbiota. (A)—I community type; (B)—II community type.
Figure 4. Top four driver families in different community types of gut microbiota. (A)—I community type; (B)—II community type.
Ijms 24 07998 g004
Figure 5. Analysis of gut microbiota taxonomic composition of CD patients with different types of gut microbiota community. (A)—Number of OTU and Shannon’s diversity index per group. (B)—Most abundant bacterial families statistically significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test).
Figure 5. Analysis of gut microbiota taxonomic composition of CD patients with different types of gut microbiota community. (A)—Number of OTU and Shannon’s diversity index per group. (B)—Most abundant bacterial families statistically significantly differentiated between comparison groups. *—p < 0.05 (Kruskall-Wallis test).
Ijms 24 07998 g005
Figure 6. Spearman’s correlations between genetic polymorphisms and gut microbiota taxonomic composition of CD patients. (a)—Number of OTU, (b)—Bacterial phyla, (c)—Bacterial families. *—p < 0.05.
Figure 6. Spearman’s correlations between genetic polymorphisms and gut microbiota taxonomic composition of CD patients. (a)—Number of OTU, (b)—Bacterial phyla, (c)—Bacterial families. *—p < 0.05.
Ijms 24 07998 g006
Table 1. Clinical characteristics of CD patients.
Table 1. Clinical characteristics of CD patients.
Clinical Characteristics % of Samples (Total n = 96)
Location of inflammation Ileitis—13.5%
Colitis—47.9%
Ileocolitis—38.6%
Phenotypic subtype Inflammatory—33.3%
Stricturing—55.2%
Fistulizing—11.5%
Crohn’s disease activity index (CDAI) Mildly active (150–220 points)—68.75%
Moderately active (221–450 points)—25%
Severely active (>451 points)—6.25%
Therapy:
no treatments (0)
5-aminosalicylic acid (1)
steroids (2)
immunosuppressor (3)
biologics (4)
(0)—7.29%
(1)—28.13%
(2)—5.21%
(3)—9.38%
(1) + (2)—5.21%
(1) + (3)—2.08%
(1) + (4)—4.17% (1) + (2) + (3)—1.04%
(1) + (2) + (4)—2.08%
(1) + (3) + (4)—2.08%
(2) + (3)—6.25%
(2) + (4)—8.33%
(2) + (3) + (4)—4.17%
(3) + (4)—14.58%
Table 2. Clinical characteristics of CD patients with different types of communities.
Table 2. Clinical characteristics of CD patients with different types of communities.
Clinical Characteristics Mean ± SD p Value (*-Kruskal-Wallis Test, §-Exact Fisher Test)
Community Type I Community Type II
CD duration, years 9.0 ± 4.4 9.7 ± 4.7 0.75 *
Number of stools per day, n 1.5 ± 1.2 1.8 ± 1.3 0.45 *
Crohn’s disease activity index (CDAI) 245.0 ± 77.2 274.8 ± 204.2 0.86 *
Body Mass Index 21.2 ± 4.1 24.2 ± 7.0 0.22 *
Location of inflammation (ileitis/colitis/ileocolitis), % 9.7/58.1/32.3 17.4/34.8/47.8 0.27 §
Phenotypic subtype (inflammatory/stricturing/fistulizing), % 32.3/54.8/12.9 34.8/56.5/8.7 1.00 §
Table 3. Allelic distribution of 24 SNPs in CD patients and healthy volunteers.
Table 3. Allelic distribution of 24 SNPs in CD patients and healthy volunteers.
SNP (Gene) AllelesCD Patients
(% of Alleles)
Healthy Volunteers
(% of Alleles)
OR (Lower 95% CI; Upper 95% CI)p Value, Exact Fisher Test
rs2241880 (ATG16L1) A/G67.1/32.947.8/52.20.45 (0.23; 0.88) 0.024
rs9858542 (BSN) A/G33.9/66.129.2/70.80.81 (0.39; 1.59)0.609
rs6908425 (CDKAL1) C/T75.5/24.568.8/31.30.71 (0.36; 1.46)0.359
rs6596075 (IBD5) C/G89.6/10.483.3/16.70.58 (0.24; 1.50)0.219
rs11805303 (IL23R) C/T69.3/30.758.3/41.70.62 (0.32; 1.21)0.171
rs1004819 (IL23R) A/G47.9/52.129.2/70.80.45 (0.22; 0.88) 0.023
rs10489629 (IL23R) C/T36.5/63.547.9/52.11.60 (0.84; 3.05)0.185
rs11209026 (IL23R) A/G9.4/90.627.1/72.93.57 (1.57; 7.99) 0.003
rs2522057 (IRF1-AS1) C/G81.8/18.270.8/29.20.54 (0.26; 1.14)0.109
rs13361189 (IRGM) C/T10.9/89.16.3/93.80.57 (0.12; 1.76)0.428
rs4958847 (IRGM) A/G32.3/67.714.6/85.40.37 (0.14; 0.82) 0.020
rs2274910 (ITLN1) C/T66.7/33.337.5/62.50.30 (0.15; 0.58) <0.001
rs1793004 (NELL1) C/G18.8/81.329.2/70.81.79 (0.85; 3.64)0.116
rs2836878 (PSMG1) A/G6.8/93.22.1/97.90.32 (0.01; 1.67)0.313
rs1992662 (PTGER4) A/G33.3/66.756.3/43.82.56 (1.34; 4.93) 0.005
rs8111071 (RSPH6A) A/G91.1/8.995.8/4.22.09 (0.57; 14.73)0.380
rs2631367 (SLC22A5) C/G55.7/44.339.6/60.40.52 (0.27; 0.99)0.053
rs3816769 (STAT3) C/T22.9/77.122.9/77.11.01 (0.45; 2.10)1.000
rs7753394 (TNFAIP3) C/T53.1/46.939.6/60.40.58 (0.30; 1.10)0.108
rs1456893 (intergenic) A/G65.6/34.472.9/27.11.40 (0.70; 2.93)0.393
rs224136 (intergenic) C/T86.0/14.087.5/12.51.07 (0.27; 7.90)1.000
rs6601764 (intergenic) C/T41.1/58.964.6/35.42.59 (1.35; 5.11) 0.006
rs7807268 (intergenic) C/G37.0/63.06.3/93.80.12 (0.03; 0.34) <0.001
rs12037606 (intergenic) A/G42.2/57.831.3/68.80.63 (0.31; 1.22)0.190
Table 4. Allelic distribution of 3 SNPs with significantly differentiated occurrence in CD patients with different types of gut microbiota communities.
Table 4. Allelic distribution of 3 SNPs with significantly differentiated occurrence in CD patients with different types of gut microbiota communities.
SNP (Gene) AllelesCommunity Type II (% of Alleles)Community Type I (% of Alleles)OR (Lower 95% CI; Upper 95% CI)p Value, Exact Fisher Test
rs9858542 (BSN) A/G45.7/54.327.0/73.00.44 (0.24;0.82)0.011
rs3816769 (STAT3) C/T14.3/85.727.9/72.12.29 (1.08; 5.24)0.033
rs1793004 (NELL1) C/G27.1/72.913.9/86.10.44 (0.21;0.92)0.034
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Markelova, M.; Senina, A.; Khusnutdinova, D.; Siniagina, M.; Kupriyanova, E.; Shakirova, G.; Odintsova, A.; Abdulkhakov, R.; Kolesnikova, I.; Shagaleeva, O.; et al. Association between Taxonomic Composition of Gut Microbiota and Host Single Nucleotide Polymorphisms in Crohn’s Disease Patients from Russia. Int. J. Mol. Sci. 2023, 24, 7998. https://doi.org/10.3390/ijms24097998

AMA Style

Markelova M, Senina A, Khusnutdinova D, Siniagina M, Kupriyanova E, Shakirova G, Odintsova A, Abdulkhakov R, Kolesnikova I, Shagaleeva O, et al. Association between Taxonomic Composition of Gut Microbiota and Host Single Nucleotide Polymorphisms in Crohn’s Disease Patients from Russia. International Journal of Molecular Sciences. 2023; 24(9):7998. https://doi.org/10.3390/ijms24097998

Chicago/Turabian Style

Markelova, Maria, Anastasia Senina, Dilyara Khusnutdinova, Maria Siniagina, Elena Kupriyanova, Gulnaz Shakirova, Alfiya Odintsova, Rustam Abdulkhakov, Irina Kolesnikova, Olga Shagaleeva, and et al. 2023. "Association between Taxonomic Composition of Gut Microbiota and Host Single Nucleotide Polymorphisms in Crohn’s Disease Patients from Russia" International Journal of Molecular Sciences 24, no. 9: 7998. https://doi.org/10.3390/ijms24097998

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

Article Metrics

Back to TopTop