Open access peer-reviewed chapter

Gut Microbiota and Inflammatory Bowel Disease

Written By

Bahareh Vakili, Parisa Shoaei, Zahra Esfandiari and Seyed Davar Siadat

Submitted: 01 May 2022 Reviewed: 13 June 2022 Published: 17 September 2022

DOI: 10.5772/intechopen.105842

From the Edited Volume

Effect of Microbiota on Health and Disease

Edited by Hoda El-Sayed

Chapter metrics overview

96 Chapter Downloads

View Full Metrics

Abstract

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder that includes Crohn’s disease and ulcerative colitis. Ulcerative colitis involves the distal colon, proximal colon, and cecum and can lead to ulcerations and bleeding. Crohn’s disease appears as patched lesions in the gastrointestinal tract and inflammation, stenosis, or fistulas. IBD affects millions of people worldwide and has been associated with high morbidity and mortality. Our intestine is colonized by trillions of microorganisms (including bacteria, viruses, fungi, and protozoa), which constitutes the microbiota. Reduction of bacteria with anti-inflammatory capacities and increase of bacteria with inflammatory capacities are observed in patients with IBD when compared with healthy individuals. Microbial balance is needed for the development of a healthy gut and a symbiotic microbiota without problems. Any disturbance in that balance leads to dysbiosis and the host may become more susceptible to disease. Some alteration in the microbiome is protective or causative; thus, we selectively will review IBD disease, pathogenesis, and potential roles of some members of microbiota in IBD. In this chapter, we also explain the therapeutic approaches targeting microbiota (probiotics, prebiotics, postbiotics) and the relationship between gut microbiota imbalance, and how defects in this dysbiosis can lead to disease.

Keywords

  • inflammatory bowel disease
  • gut microbiome
  • Crohn’s disease
  • ulcerative colitis
  • microbiome
  • dysbiosis
  • therapy

1. Introduction

Inflammatory bowel disease (IBD), including ulcerative colitis (UC), Crohn’s disease (CD), and indeterminate colitis, is a chronic and relapsing inflammatory disorder of the gastrointestinal tract [1, 2]. More than 1 million residents in the United States and 2.5 million individuals living in Europe are estimated to be suffering from IBD [2]. The incidence of IBD has been rapidly increasing in newly industrialized countries in Asia, the Middle East, Africa, and South America over the last two decades [3, 4]. IBD has been associated with high morbidity and mortality, low quality of life, and financially demanding medical care [5]. The causes of this disease are multifactorial, the two main types: UC and CD, have similar clinical and pathological presentations and can cause irreversible impairment of the structure and function of the gastrointestinal tract [6]. These diseases are characterized by a relapsing behavior, manifested by alternating phases of inactive states in which there is no intestinal inflammation and active states that present inflammation or any other disease symptoms [7]. Although the main biological processes involved in the development of both conditions are different [6]. CD can affect any part of the GI tract, especially the terminal ileum, associated with inflammation, stenosis, and/or fistulas [7, 8].

CD occurs in patients between the ages of 15 and 35 years, affects the mucosal layer of the colon, and causes abdominal pain, diarrhea, and fever, fistula, lesions in the rectum or intestine, and other symptoms. CD damages the small intestine; therefore, malnutrition is very common in CD [9]. Despite the UC, rectal bleeding is less common in CD patients and more than 50% of patients with CD suffer from folate and vitamin D deficiency, while more than 50% of people with UC suffer from iron deficiency [10].

UC disease is a mucosal inflammation that can only affect the large intestine, i.e., the colon, and the inflammation generally starts in the distal colon, going forward through the proximal colon until the cecum and can lead to ulcerations and bleeding [11]. About 25% of UC patients are diagnosed before the age of 18 years, because this disease affects adolescence [9]. There are different diagnostic tests for UC including clinical, endoscopic, histologic, and radiological tests although approximately 8.5% of IBDs are unclear [12, 13]. UC is the initial subtype of IBD, and the term IBD includes the characteristics of both CD and UC. It has long been difficult to distinguish between these two diseases, but now there is a clinical definition for both. Both diseases can affect specific parts of the lives of patients, such as school, job, social life, and family life Figure 1 [9].

Figure 1.

The effective agents in development of inflammatory bowel disease.

The concept of IBD pathogenesis is based on the theory of a disrupted intestinal barrier and a dysregulated immune response in a genetically susceptible host. IBD presents defects in the detection and control of the gut microbiota, associated with unbalanced immune reactions, genetic mutations that confer susceptibility to the disease, and complex environmental conditions such as a Westernized lifestyle [7, 14]. There is a strong clustering in families and with certain ethnicities. Other studies showed 15–50 times increased relative risk for siblings of a CD patient to also develop CD. The ethiopathology of IBD is multifactorial and is characterized by the interaction between genetic, microbial, environmental, and life style factors, which influences the immune responses and leads to the gut inflammation. Gut microbiota is important for the development and maturation of the immune system and reduced microbial diversity and its dysbiosis observed in IBD patients (Figure 1).

More than 200 IBD-associated susceptible genes have been identified, some of which are known to be involved or implicated in mediating host responses to gut microbiota [14]. This has evoked the possibility that gut microbiota is implicated in the pathogenesis of IBD [3]. Microbial factors have been historically proven to be indispensable for the onset of IBD, and advances in high-throughput sequencing have enabled us to elucidate the gut microbiome in IBD. IBD can be caused by determined infection of an enteric pathogen such as Mycobacterium avium subspecies paratuberculosis, Clostridioides difficile, Helicobacter pylori, Campylobacter concisus, Fusobacterium nucleatum, and adhesion-invasive Escherichia coli. An excess of translocation of intestinal bacteria across the intestinal barrier, the imbalance between beneficial and detrimental commensal bacteria can cause IBD [15, 16]. Some bacteria including AIEC can be considered as both a persistent pathogen and detrimental commensal bacteria [17].

Therefore, this chapter covers the origins, causes, diagnosis, and treatment strategies of this complex disease.

Advertisement

2. IBD and immunity system

Epithelial layer integration permits the gastrointestinal bacteria to communicate with the immune system [18]. The mucosal layer is the first physical barrier on the mucosal surface and is produced by the polymerization of gel-forming mucins secreted by Goblet cells. The second defense barrier against bacterial attack is the intestinal epithelium, which makes up of enterocytes and particular epithelial cells called Goblet and Paneth cells [19]. Intestinal epithelial cells prevent the influx of antigens and the attack of pathogens and commensal microbes [18]. Intestinal epithelial cells (IECs) also express toll-like receptors (TLRs) and nucleotide oligomerization domain receptors (NODs), which are pathogen-sensitive innate immune receptors. IECs then make chemokines and cytokines to engage immune cells [18]. TLR signaling pathways helps the epithelial barrier to remain intact and produce 12 and interleukin 6 [18, 20]. The epithelial barrier impairment causes intestinal permeability to increase, which has been shown in CD and also in UC, and this might be a main pathogenetic mechanism in IBD [19]. TLR acts as pro/anti-inflammatory gene activation inducer and controls the adaptive immune responses [21, 22].

Intestinal immune cells including innate immune cells and adaptive immune cells significantly involve in immune responses in IBD [23]. Macrophages, TLRs, and NOD-like receptors (NLRs) are essential for developing tolerance to certain pathogens and promoting wound treatment. Binding to pathogene receptors leads to the activation of different signaling pathways and the production of proinflammatory cytokines, chemokines, and antimicrobial peptides. The antigen-presenting cells (APCs) link innate immunity and adaptive immunity by secreting cytokines and presenting antigens to the T cells [24]. Fine gut-resident macrophages, described by a lack of CD14 expression, manifest decreased response, proliferation, and chemotactic activity. The gut-resident macrophages have increased phagocytic activity and secretion of cytokines in IBD patients, causing dramatic inflammation [25]. After microorganisms’ invasion, innate immunity activates after a few hours [26]. Macrophage cells kill specific pathogens, such as peptides and lipopolysaccharides. In IBD acute phase, the number of macrophages in the intestinal mucosa increases dramatically, and a large number of T cells and costimulatory molecules such as CD40, CD80, and CD86 are involved in the inflammatory process and intolerance of commensal microbes and immune activity [27].

Malfunction in TLR signaling can induce an intestinal inflammatory response with various clinical phenotypes, including the IBD. A considerable target of the TLR signaling is the activation of the transcription factor NF-kB, which regulates the expression of a variety of genes responsible for controlling the innate response, such as IL-1, IL-2, IL-6, IL-12, and TNF-𝛼 [28, 29]. Table 1 shows the cytokines and cellular sources involved in immune response in IBD. Both IL-1 and TNF-𝛼 share numerous pro-inflammatory properties responsible for the development of IBD [30]. Dendritic cells are professional antigen-presenting cells that activate T cells and induce adaptive immune responses, describing key players in the cross talk between innate and adaptive immunity [38].

Pro-inflammatory
CytokinesCellular SourcesPrinciple functionRole in immune system
IL-1, IL-1βMϕ, IECS, MonocytesInfluence on the T cell and secretory cytokinesInnate immune response [30, 31, 32]
IL-2Th-cellsT & B cells proliferation IFN-γ productionAdaptive immune response [31]
IL-6DCs, MϕDifferention of Th17, Treg cells and activating STAT-3 signaling pathwayInnate immune response [31, 33, 34, 35]
IL-12DCs, MϕPromoting the differentiation of Th1 and Th17 cellsInnate immune response [36, 37]
IL-13Th2 cellsIntestinal permeability inducing and activating of B cellInnate immune response [38]
IL-17Th17 cellsInducing and promoting of secretory cytokinesAdaptive immune response [39, 40]
IL-18Provoking the secretion of pro-inflammatory cytokinesInnate & adaptive immune response [32]
IL-22Th17 cellsInhibiting pathogens of intestinal and repairing of intestinal tissueInnate immune response [41]
IL-23Provoking the production cytokinesInnate immune response
[36]
TNF-αMϕ, DCs, Th-cellsPromoting the production cytokines and Th-cells proliferationInnate immune response [31, 42]
INF-γTh-cellsActivating NF-κB signaling pathway and activating of MϕAdaptive immune response [43]
Anti-inflammatory
IL-4Th2-cellsTh2-cells differentiation and inhibiting the production of cytokines of Th1cellsAdaptive immune response [31]
IL-10DCs, Mϕ, Treg cellsInhibiting the production of cytokines Th1 cellsAdaptive immune response [31, 44]
TGF-βDCs, Treg cells, T cellsTreg and Th17 differentiation and restraining of Th-cellsAdaptive immune response [31, 34, 35]

Table 1.

The pro-inflammatory agents’ contribution in immune response in IBD.

Abbreviations: Mϕ: Macrophage, IECS: Intestinal epithelial cells, DCs: dendritic cells, STAT-3: signal transducer and activator of transcription, NF-κB: nuclear factor kappa B.

The other IBD risk variants in other genes are involved in IL-12 and CCR6, chemokine receptors preferentially expressed on IL-17 producing cells [19]. IL-23/IL- 17 axis has a key role in this cross talk and the IL23R gene encodes a specific subunit of the IL23 receptor that has been identified and largely replicated in independent cohorts of patients with both CD and UC [45]. Other clinical studies have found that the intestinal mucosa and lamina propria of IBD patients contain much higher levels of Th17 cells, IL-17, and IL-23 compared with the healthy controls [24].

Appositive of the innate immune response, the adaptive immune system is very specific, it presents long-lasting immunity. Key players of the adaptive immune response are T cells. Th0 cells can become activated and either differentiate into Th1 or Th2 or Th17 cells [19, 38]. However, a dysregulated T cell response with abnormal development of activated T cell subsets causes inflammation because of an excess release of cytokines and chemokines, which have multiple pathogenic impacts on components of the immune system. Figure 2 shows the immune response in IBD. The levels of T-cell-derived cytokines detected in IBD mucosa, different studies have associated CD and UC with different subtypes of pro-inflammatory immune responses. Therefore, the innate immune response is as important as the adaptive immune system in inducing gut inflammation in these patients [19, 24, 38].

Figure 2.

The role of immune response on progress IBD.

Genome-wide association studies and immunological studies have mentioned that IBD pathogenesis is related to mucosal innate immune responses, including classical Th1 response in CD patients and Th2 type-like response in UC patients [45, 46].

In mouse model studies, induction of CD caused increase of IFN-𝛾 expression in their spleen and local intestinal mucosa [43]. CD evolution is generally mediated by CD4+ Th1 and Th17 cells, and IFN-𝛾 is a major cytokine declared in this disease [47]. Deficiencies of IL17-A and IL17-B in experimental models showed both pro-inflammatory and tissue-protective effects against colitis depending on the model used [19, 48]. However in mucosa of IBD patients, IL-17A cells regulate and induce a number of pro-inflammatory molecules [38].

Regulatory T cells (Treg) produce the anti-inflammatory cytokines (IL-10, TGF) and exert an effective anti-inflammatory action in experimental colitis. Treg are reduced in peripheral blood of patients with active IBD in comparison with quiescent IBD patients and control subjects [49, 50]. In contrast, Treg are increased in the intestinal mucosa of IBD patients, and their function is normal. An intact TGF signaling, which is impaired in inflamed IBD mucosa because of upregulation of the inhibitory molecule Smad7, is needed for Treg function [19]. Treg cells, expressing the transcription factor forkhead box P3 (FOXP3), have a negative immunomodulatory character in immune tolerance and a crucial role in the pathogenesis of IBD [24, 51, 52].

Advertisement

3. IBD and gut microbiota

IBD is obviously related to gut dysbiosis that impairs host-microbe and immune homeostasis [53]. The human gut includes trillions of commensal bacteria per gram of gut lumen content. These bacteria can be nutritious and provide the intestinal epithelium [38, 54]. The gut microbiota leads to intestinal homeostasis due to our physiological procedure and metabolites [55]. There are different phyla, including Bacteroidetes, Firmicutes, Proteobacteria (Escherichia and Helicobacter), and Actinobacteria that include fungi, protists, and viruses (Table 2) [56, 57].

IncreasedDecreased
BacteriaFusobacterium speciesBififidobacterium species
Pasturellaceae Bacteroides species
Proteobacteria (adherent invasive Escherichia coli)Clostridium XIVa, IV
Ruminococcus gnavus Faecalibacterium prausnitzii
Veillonellaceae Roseburia species
Suterella species
FungiCandida albicansSaccharomyces cerevisiae
Candida tropicalis
Clavispora lusitaniae
Cyberlindnera jadinii
Kluyveromyces marxianus
VirusesCaudivirales

Table 2.

Microbiota changes associated with inflammatory bowel disease.

Ecological factors, such as as host diet, hygiene, antibiotic consumption, and lifestyle, induce immune responses that change the intestinal microbiota and damage the mucosal barrier [38, 58]. Gut microbiota plays an important role in the pathogenesis of IBD and impacts energy metabolism host, immune homeostasis, development and maintenance of mucosal integrity [24]. Table 3 shows the effect of gut microbiota in inflammatory bowel disease and its interdependence with the immune response.

DepletedImmune Association
SCFA producing bacteria (F. prausnitzii, Roseburia, Eubacterium)Produce SCFA plays a major role in modulation of inflammation, regulation of immune responses, maintenance of barrier integrity in the gut, enhanced expansion of the Treg population, and skew of human dendritic cells to prime IL-10-secreting T cells [59, 60, 61].
Bacteroides fragilisProduces lipid antigens controlling homeostatic iNKT cell proliferation and activation [62].
BifidobacteriumInhibits intestinal inflammation by acting on Treg cells [63].
Mbb. smithiiWeak association with pro-inflammatory mechanisms [64].
EnrichedImmune association
E. coli (adherent invasive)Invades intestinal epithelial cells replicate in macrophages and induce granulomas [65].
Clostridiaceae
(class)Clostridiales
Clostridium difficile could induce the expansion of regulatory Tcells (Treg) and to mitigate intestinal inflammation [66].
Proteobacteria (Salmonella, Yersinia, Desulfovibrio, Vibrio Helicobacte)Associated with a pro-inflammatory state as revealed by quantification of common pro-inflammatory interleukins. The inflamed gut appears to provide a favorable environment for the expansion of this phylum [67].
R. gnavusSecretes a complex glucomannan polysaccharide inducing TNFα secretion by dendritic cells [68].
FusobacteriumEspecially F. nucleatum, which is a well-recognized proinflammatory bacterium and it may secrete Outer Membrane Vesicles (OMVs) activate epithelial TLR4 to drive inflammation [69, 70].
C. albicansInteracts with mucosal innate immune cells through the pathways associated with Dectin-1 in macrophages [71].
Bacteriophages (Caudovirales and Microviridae)role in physiology of intestinal or change the bacterial in gut microbiota via predator-prey relationships [72]. Enterobacteria are the hosts of Microviridae [73].
Eukaryotic virusesInfect of intestinal and develop host susceptibility to IBD by immune response via inflammatory mediators, and inducing alterations in the composition of the commensal bacteria [74].
Eukaryotic virusesInfect host cells may increase host susceptibility to IBD by supporting a long-standing immune response through inflammatory mediators, as well as by inducing alterations in the composition of the commensal microbiota [74]
M. stastmanaeThis leads to the substantial release of proinflammatory cytokines in monocyte-derived dendritic cells [64].

Table 3.

Gut microbiome in inflammatory bowel disease and its associations with the immune system [7].

For example, Clostridium cluster IV and XIVa were less abundant in IBD patients than in healthy controls [55]. Bacteroides genus is obligate anaerobe bacteria and consists a large amount of the normal gut microbiota. B. fragilis decreases in IBD patients and promotes the quantities of anti-inflammatory cytokines against colitis [24, 75]. The overgrowth of Enterobacteriaceae, Pseudomonas-like bacteria, and Escherichia coli promotes the intestinal inflammation and alters the composition of the microbiota in most colitis models and IBD patients [58, 76, 77]. Faecalibacterium prausnitzii secret anti-inflammatory cytokines, which reduce in the intestine of IBD patients [24, 55]. Fusobacterium and Ruminococcus gnavus have also been increased in CDI patients [78]. In a recent study performed on IBD patients, a functional gut microbiome dysbiosis and impaired microbial transcript were seen. Facultative anaerobes were raised at the expense of obligate anaerobes [79]. Other different studies showed that the diversity of gut microbiota was either decreased or equal in IBD patients versus controls. F. prausnitzii, Eubacterium rectale, and Akkermansia were decreased, and Actinomyces, Veillonella, and E. coli were increased in patients with UC (Table 3) [80].

Other possible pathogens in the exacerbation of the IBD disease are Mycobacterium avium subspecies paratuberculosis, Clostridium difficile, Listeria monocytogenes, and Campylobacter concisus, as well as viruses, including cytomegalovirus, Epstein-Barr virus, and measles virus [17, 81]. In addition, a number of pathogenic parasites may involve in the progression of this disease. Overexposure of immune system in the presence of too many bacterial materials could also cause the loss of immunological tolerance to the bacteria, which are generally considered the normal flora in the gut [81]. Some of the individual bacterial species that associate with human IBD are reviewed here.

3.1 Clostridioides difficile

C. difficile is an obligate anaerobic Gram-positive spore-forming bacterium, which is prevalent in nature and also colonizes the human intestinal tract [81, 82]. C. difficile leads to diarrhea and colitis, frequently in persons who have been treated with antibiotics for other medical complications [81, 83, 84].

C. difficile can produce toxins type A and B, and IBD patients with C. difficile infections (CDI) appear severe clinical symptoms, such as abdominal pain, diarrhea, bloody stools, and leukocytosis [85, 86].

CDI causes relapsed IBD, and IBD patients in remission had a significantly higher presence of toxigenic C. difficile in their intestinal tract as compared with healthy controls [87]. UC patients have a high risk of CDI in comparison with healthy population or CD patients.

The two toxins encoded by tcdA and tcdB genes lead to the disruption of epithelial cytoskeleton and tight junctions, which contribute to the CDI [81, 88, 89]. A reduction in butyrate-producing bacteria and increase in lactic-acid-producing bacteria were seen in CDI status. Overrepresentation of Akkermansia may be a predictive marker for the development of nosocomial diarrhea, which can result in a worse CDI prognosis [82]. Activation of the production of multiple inflammatory cytokines such as IL-8, TNF-a, IL-1, and tumor necrosis factor (TNF-a) could damage the intestinal epithelial cells and trigger IBD in CDI patients [90]. Reduced bile salts happen in the colon of patients with IBD leading to spore germination of C. difficile [86, 91]. Patients with IBD present common infections such as gastrointestinal infections of C. difficile, Salmonella, Shigella, and Campylobacter jejuni [81, 92].

3.2 M. avium subspecies paratuberculosis

M. avium species is commonly present in the environment and comprises four subspecies, including M. avium subspecies avium, M. avium, M. avium subspecies hominissuis, and M. avium subspecies silvaticum [93]. M.avium causes production of some inflammatory cytokines in IBD patients [86]. In IBD patients, the increase in metalloprotease leads to dysregulation in immune system and large level of inflammatory cytokines [94, 95]. Combination of multiple antibiotics including rifabutin, clofazimine, and clarithromycin, adds up to ciprofloxacin, metronidazole, or ethambutol, which are used for treatment of patients with positive different species of M.avium [96]. Also antibiotics such as nitroimidazoles and clofazimine are effective in the treatment of CD [97].

3.3 Helicobacter pylori

Helicobacter species are Gram-negative bacteria. H. pylori is an important pathogen that isolates from gastrointestinal tract of humans and animals. H. pylori infection has been reported in IBD patients and shows a protective effect in IBD [98, 99]. H. pylori increases the expression of forkhead box P3 (FOXP3) with stimulating of the regulatory T cells production, reduces the production of inflammatory cytokines, and finally, decreases inflammation [100, 101]. H. pylori with cytotoxin-associated gene A (CagA+) genotype, in IBD patients, diverts TH1 response to TH2 response that has anti-inflammatory task [102].

Helicobacter species are more detected in intestinal biopsies of patients with CD and UC than controls, although this difference was not significant [103]. Molecular studies detected non-pylori Helicobactor by Helicobacteriaceae family-specific PCR in 3% of IBD patients and 8% controls [104].

3.4 C. concisus and Fusobacterium nucleatum

Most strains of campylobacter colonize in the intestinal tract, but the colonization of C. concisus is in the oral cavity [105]. C.concisus is associated with IBD in the adult patients [106]. The virulence factors of C. concisus infect the lower parts of the intestinal tract [86]. Zonula occludens toxin (Zot) is expressed through a CON-Phi2 prophage and leads to the permeability of the epithelial cells and formation of IBD. This mechanism is similar to the Vibrio cholerae toxin [107]. C. concisus breaks the intestinal epithelial barrier and leads to apoptosis in human intestinal epithelial and intestinal inflammation [108]. The invasive strains of C. concisus enable them to survive in harsh conditions such as in anaerobic conditions [86].

F. nucleatum is an anaerobe bacterium that colonizes the oral cavity and intestinal tract [81]. It is abundant in intestinal tract of UC and IBD patients, and the quantity was linked with disease severity [109]. F. nucleatum leads to the damage of intestinal epithelium and promotes intestinal inflammation by inducing autophagic epithelial cell death [86].

3.5 Adherent-invasive E. coli

Adherent-invasive E. coli (AIEC) is a commensal human gut bacterium and is associated with ileal CD in the adult population. AIEC strains can adhere to and invade intestinal epithelial barrier assessed [110]. AIEC strains have various mechanisms and virulence factors, which are involved in the pathogenesis of IBD patients [86]. Several factors such as type 1 pili adhesion FimH and carcinoembryonic antigen cell adhesion molecule 6 are associated in promoting inflammation [111]. AIEC strains induced production of cytokines such as IL-8, TNF-a, and IL-6 in both epithelial cells and macrophages. Replication of AIEC in macrophages did not cause macrophage death, but increased production of TNF-a and IL-6 [81, 112].

Advertisement

4. Therapeutic approaches targeting microbiota (probiotics, prebiotics, postbiotics, and antibiotics)

Probiotics are live microorganisms, which allocate great health advantages for the host organism when used in an appropriate quantity [113]. Probiotics induce anti-inflammatory effects, enhance or renew barrier work, promote the growth of beneficial bacteria, and inhibit the growth of pathogens [114]. Probiotics rebalance the gut microflora shifting from pro- to anti-inflammatory state [115]. Prebiotics are substrates that are selectively utilized by probiotics allocating health benefits [17]. Inulin is a prebiotic that retains microbial population, helps the epithelium barrier function, and inhibits from pathogens translocation [116]. This process leads to the treatment of functional symptoms in IBD. Postbiotics are bioactive molecules produced by probiotics [117]. There are many reports that showed some probiotics and prebiotics can be beneficial in treatment and prevention of IBD in both human and mice models [118].

In CD, evidence for prebiotics and probiotics is commonly dissatisfactory and antibiotics have moderate effects [66]. The most common strains that are used as beneficial probiotics are Bifidobacterium species, Enterococcus faecium, Lactobacillus strains, and Saccharomyces boulardii, Bacillus species, and Pediococcus [115]. The theoretical risks of probiotics on animal models of IBD in different studies are described that include systemic infections, harmful metabolic activities, extreme immune stimulation in susceptible patients, gene transfer, and gastrointestinal adverse effects [119]. Some traditional probiotics, such as the probiotic cocktail VSL# 3 (containing a mix of four lactobacilli, three Bifidobacteria, and one Streptococcus strain), have shown limited effect in treating CD and UC, by reducing active inflammation and recurrence [17, 66].

Some clinical trials showed that Lactobacillus rhamnosus administration in gastroenteritis children did not have better outcomes than those who received placebo [120]. Although a multi-strain probiotic (including L. rhamnosus, Lactobacillus plantarum, Lactobacillus acidophilus, and E. faecium) is related with lower intestinal inflammation in UC patients, but not in CD patients [121]. Besides the mentioned traditional probiotics, Akkermansia muciniphila and their supernatants that contain postbiotics significantly reduced the severity of colitis [17]. F. prausnitzii produce barrier improving immunosuppressive SCFAs, stimulate Tregs to produce IL-10, which have protective effects on the intestine [66]. In mice models, administration of A. muciniphila or its postbiotic reduced the infiltrating macrophages and CD8+ in clolon and inhibited colitis [122, 123].

These helpful microbes and their metabolites should be investigated as therapeutic determinants in treatment of IBD. Dietary substrates such as oligosaccharides and fiber are prebiotics that selectively increase the quantities of SCFA-producing commensals, blocking the AIEC epithelial adherence, and the virulence products of intestinal pathogens in IBD [66].

Probiotic engineering with emerging technologies such as as CRISPR-Cas system can be used to produce to treat untreatable chronic inflammatory conditions [115]. With increasing our knowledge about viable bacterial strains and synthetic biology tools, we can identify and characterize extra probiotic bacterial strains as potential candidates for probiotic engineering [124].

Antimicrobial agents and IBD have a complex relationship. They have hazardous influences on the homeostasis of the host microbiota, leading to a population shift described by increased Enterobacteriaceae and decreased Clostridia abundant, which is regarded a possible pre-IBD condition [125]. Also, IBD patients treated with antibiotics are at high risk of forming an overgrowth of pathogenic microbes including C. difficile, candida, and bacteriophages [126].

In addition, antibiotics are an integral part of the treatment repertoire in IBD, whereas before the period of immunomodulation and biologic therapy. The mechanisms of antibiotics in treatment of IBD are a direct effect on the gut microbiota, preferring flora that are linked with anti-inflammatory properties, e.g., Bacteroides and Firmicutes, and decreasing pathogenic microbes that are associated with inflammation, such as as Enterobacteriaceae, e.g., E. coli and Fusobacterium [75]. Furthermore, we can choose target-specific pathobiants or to manage individual microbiome in IBD patients by determining patient stool samples prior to treatment [124].

The immunological mechanisms of IBD have made great upgrades, provided novel tactics for IBD treatment. Biological agents induce and maintain clinical remission of IBD and promote mucosal curing. A number of biological agents that have been approved for the treatment of IBD are some of the TNF-α inhibitors such as Infliximab, Adalimumab, Certolizumab pegol, Glimumab, Etanercept, and Tocilizumab [24]. However 10–40% of IBD patients do not respond or lose their response to treatment over time [127].

4.1 Fecal microbiota transplantation (FMT)

FMT appears effective therapy for treatment of recurrent CDI and in UC or CD remission induction but remains strong and safe in the long term is not clear [128, 129]. A significant proportion of recurrent CDI patients have IBD, and FMT is moderately less successful in treatment of CDI from patients with IBD in comparison with patients without IBD [130]. Some issues could affect the FMT outcome in IBD treatment including donor choice; preparation of fecal material; clinical management, the high abundances of fungi or virus communities in donor stool or other essential necessities for implementing an FMT center [131, 132].

Recently, the field of IBD genetics has made enormous progress, and different relative molecular and cellular pathways exist. Fluctuations in specific gene loci promise therapeutics for IBD in the future. Besides, FMT, novel natural medicines, new antimicrobial agents, and combined treatment programs are also anticipated to break the IBD and therapeutically delay. The combined treatment strategies that use anti-inflammatory agents and anti-fibrotic drugs will provide great insights into the existing IBD therapeutics [17].

Advertisement

5. Conclusion

Correct interplay between gut microbiota and the host is essential for human health. Microbial balance is pivotal for host metabolic and immune functions as well as to prevent disease development. Disturbance in that balance generates dysbiosis making the host susceptible to certain diseases. Gut microbiota stimulates the immune system, and altered composition of this microbiota in early life can lead to an inadequately trained immune system that can overreact to commensal microbes and lead to inflammatory diseases. Recent research has provided striking findings supporting that the gut microbiome plays an important function in the etiopathogenesis of IBD.

The clinical and epidemiological evidences showed that the infectious pathogens have possible role in IBD progression, especially, Mycobacterium avium paratuberculosis, C. difficile, E. coli, and C. concisus. Also, some viruses such as cytomegalovirus, Epstein-Barr virus, and measles by different pathogenesis have been be associated with the higher IBD risk; however, H. pylori may reduce intestinal inflammation and protect against IBD.

References

  1. 1. Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annual review of immunology. 2009;28:573-621
  2. 2. Kaplan GG. The global burden of IBD: From 2015 to 2025. Nature reviews Gastroenterology & Hepatology. 2015;12(12):720-727
  3. 3. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clinical journal of gastroenterology. 2018;11(1):1-10
  4. 4. Alshehri D, Saadah O, Mosli M, Edris S, Alhindi R, Bahieldin A. Dysbiosis of gut microbiota in inflammatory bowel disease: Current therapies and potential for microbiota-modulating therapeutic approaches. Bosnian Journal of Basic Medical Sciences. 2021;21(3):270
  5. 5. Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: A systematic review of population-based studies. The Lancet. 2017;390(10114):2769-2778
  6. 6. Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nature Reviews Immunology. 2003;3(7):521-533
  7. 7. Aldars-García L, Marin AC, Chaparro M, Gisbert JP. The interplay between immune system and microbiota in inflammatory bowel disease: A narrative review. International Journal of Molecular Sciences. 2021;22(6):3076
  8. 8. Thoreson R, Cullen JJ. Pathophysiology of inflammatory bowel disease: An overview. Surgical Clinics of North America. 2007;87(3):575-585
  9. 9. Seyedian SS, Nokhostin F, Malamir MD. A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease. Journal of Medicine and Life. 2019;12(2):113
  10. 10. Burgmann T, Clara I, Graff L, Walker J, Lix L, Rawsthorne P, et al. The Manitoba inflammatory bowel disease cohort study: Prolonged symptoms before diagnosis—How much is irritable bowel syndrome? Clinical Gastroenterology and Hepatology. 2006;4(5):614-620
  11. 11. Yang C, Singh P, Singh H, Le ML, El-Matary W. Systematic review: Thalidomide and thalidomide analogues for treatment of inflammatory bowel disease. Alimentary Pharmacology & Therapeutics. 2015;41(11):1079-1093
  12. 12. Cummings JF, Keshav S, Travis SP. Medical management of Crohn’s disease. BMJ. 2008;336(7652):1062-1066
  13. 13. Tremaine WJ. Diagnosis and treatment of indeterminate colitis. Gastroenterology & Hepatology. 2011;7(12):826
  14. 14. Liu JZ, Van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nature Genetics. 2015;47(9):979-986
  15. 15. De Hertogh G, Aerssens J, Geboes KP, Geboes K. Evidence for the involvement of infectious agents in the pathogenesis of Crohn’s disease. World Journal of Gastroenterology: WJG. 2008;14(6):845
  16. 16. Kalischuk LD, Buret AG. A role for campylobacter jejuni-induced enteritis in inflammatory bowel disease? American journal of physiology-gastrointestinal and liver. Physiology. 2010;298(1):G1-G9
  17. 17. Liu S, Zhao W, Lan P, Mou X. The microbiome in inflammatory bowel diseases: From pathogenesis to therapy. Protein & Cell. 2021;12(5):331-345
  18. 18. Hisamatsu T, Kanai T, Mikami Y, Yoneno K, Matsuoka K, Hibi T. Immune aspects of the pathogenesis of inflammatory bowel disease. Pharmacology & Therapeutics. 2013;137(3):283-297
  19. 19. Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmunity Reviews. 2014;13(1):3-10
  20. 20. Bamias G, Cominelli F. Immunopathogenesis of inflammatory bowel disease: Current concepts. Current opinion in gastroenterology. 2007;23(4):365-369
  21. 21. Medzhitov R, Preston-Hurlburt P, Janeway CA. A human homologue of the drosophila toll protein signals activation of adaptive immunity. Nature. 1997;388(6640):394-397
  22. 22. Poltorak A, Smirnova I, He X, Liu M-Y, Van Huffel C, Birdwell D, et al. Genetic and physical mapping of the LpsLocus: Identification of the Toll-4 receptor as a candidate gene in the critical region. Blood Cells, Molecules, and Diseases. 1998;24(3):340-355
  23. 23. Knutson CG, Mangerich A, Zeng Y, Raczynski AR, Liberman RG, Kang P, et al. Chemical and cytokine features of innate immunity characterize serum and tissue profiles in inflammatory bowel disease. Proceedings of the National Academy of Sciences. 2013;110(26):E2332-E2E41
  24. 24. Lu Q , Yang M-f, Liang Y-j, Xu J, Xu H-M, Nie Y-Q , et al. Immunology of inflammatory bowel disease: Molecular mechanisms and therapeutics. Journal of Inflammation Research. 2022;15:1825
  25. 25. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474(7351):298-306
  26. 26. Medzhitov R, Janeway C Jr. Innate immunity. New England Journal of Medicine. 2000;343(5):338-344
  27. 27. Zhang SZ, Zhao XH, Zhang D. Cellular and molecular immunopathogenesis of ulcerative colitis. Cellular & Molecular Immunology. 2006;3(1):35-40
  28. 28. Ghosh S, May MJ, Kopp EB. NF-κB and Rel proteins: Evolutionarily conserved mediators of immune responses. Annual Review of Immunology. 1998;16(1):225-260
  29. 29. Caamano J, Hunter CA. NF-κB family of transcription factors: Central regulators of innate and adaptive immune functions. Clinical Microbiology Reviews. 2002;15(3):414-429
  30. 30. Blam ME, Stein RB, Lichtenstein GR. Integrating anti–tumor necrosis factor therapy in inflammatory bowel disease: Current and future perspectives. The American Journal of Gastroenterology. 2001;96(7):1977-1997
  31. 31. Huang Y, Chen Z. Inflammatory bowel disease related innate immunity and adaptive immunity. American Journal of Translational Research. 2016;8(6):2490
  32. 32. Ligumsky M, Simon P, Karmeli F, Rachmilewitz D. Role of interleukin 1 in inflammatory bowel disease--enhanced production during active disease. Gut. 1990;31(6):686-689
  33. 33. Atreya R, Neurath M. New therapeutic strategies for treatment of inflammatory bowel disease. Mucosal Immunology. 2008;1(3):175-182
  34. 34. Stockinger B, Veldhoen M. Differentiation and function of Th17 T cells. Current Opinion in Immunology. 2007;19(3):281-286
  35. 35. Weaver CT, Harrington LE, Mangan PR, Gavrieli M, Murphy KM. Th17: An effector CD4 T cell lineage with regulatory T cell ties. Immunity. 2006;24(6):677-688
  36. 36. Torres M, Rios A. Current view of the immunopathogenesis in inflammatory bowel disease and its implications for therapy. World journal of gastroenterology: WJG. 2008;14(13):1972
  37. 37. Leppkes M, Neurath M. Cytokines in inflammatory bowel diseases–update 2020. Pharmacological Research. 2020;158:104835
  38. 38. Silva FA, Rodrigues BL, Ayrizono MLS, Leal RF. The immunological basis of inflammatory bowel disease. Gastroenterology Research and Practice. 2016;2016:1-11
  39. 39. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity. 2004;21(4):467-476
  40. 40. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nature Immunology. 2005;6(11):1123-1132
  41. 41. Ramani K, Biswas PS. Interleukin-17: Friend or foe in organ fibrosis. Cytokine. 2019;120:282-288
  42. 42. Aardoom MA, Veereman G, de Ridder L. A review on the use of anti-TNF in children and adolescents with inflammatory bowel disease. International Journal of Molecular Sciences. 2019;20(10):2529
  43. 43. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, et al. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell. 2006;126(6):1121-1133
  44. 44. Saraiva M, O'garra A. The regulation of IL-10 production by immune cells. Nature Reviews Immunology. 2010;10(3):170-181
  45. 45. Duerr RH, Taylor KD, Brant SR, Rioux JD, Silverberg MS, Daly MJ, et al. A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science. 2006;314(5804):1461-1463
  46. 46. Fuss IJ, Neurath M, Boirivant M, Klein JS, De La Motte C, Strong SA, et al. Disparate CD4+ lamina propria (LP) lymphokine secretion profiles in inflammatory bowel disease. Crohn's disease LP cells manifest increased secretion of IFN-gamma, whereas ulcerative colitis LP cells manifest increased secretion of IL-5. The Journal of Immunology. 1996;157(3):1261-1270
  47. 47. Strober W, Fuss IJ. Proinflammatory cytokines in the pathogenesis of inflammatory bowel diseases. Gastroenterology. 2011;140(6):1756-67. e1
  48. 48. MacDonald TT, Biancheri P, Sarra M, Monteleone G. What's the next best cytokine target in IBD? Inflammatory bowel diseases. 2012;18(11):2180-2189
  49. 49. Fantini MC, Becker C, Tubbe I, Nikolaev A, Lehr HA, Galle P, et al. Transforming growth factor β induced FoxP3+ regulatory T cells suppress Th1 mediated experimental colitis. Gut. 2006;55(5):671-680
  50. 50. Chamouard P, Monneaux F, Richert Z, Voegeli A-C, Lavaux T, Gaub MP, et al. Diminution of circulating CD4+ CD25high T cells in naïve Crohn’s disease. Digestive Diseases and Sciences. 2009;54(10):2084-2093
  51. 51. Monteleone G, Kumberova A, Croft NM, McKenzie C, Steer HW, MacDonald TT. Blocking Smad7 restores TGF-β1 signaling in chronic inflammatory bowel disease. The Journal of Clinical Investigation. 2001;108(4):601-609
  52. 52. Fahlén L, Read S, Gorelik L, Hurst SD, Coffman RL, Flavell RA, et al. T cells that cannot respond to TGF-β escape control by CD4+ CD25+ regulatory T cells. The Journal of Experimental Medicine. 2005;201(5):737-746
  53. 53. Miyoshi J, Chang EB. The gut microbiota and inflammatory bowel diseases. Translational Research. 2017;179:38-48
  54. 54. Sartor RB. Mechanisms of disease: Pathogenesis of Crohn's disease and ulcerative colitis. Nature Clinical Practice Gastroenterology & Hepatology. 2006;3(7):390-407
  55. 55. Ihara S, Hirata Y, Koike K. TGF-β in inflammatory bowel disease: A key regulator of immune cells, epithelium, and the intestinal microbiota. Journal of Gastroenterology. 2017;52(7):777-787
  56. 56. Cho JH. The genetics and immunopathogenesis of inflammatory bowel disease. Nature Reviews Immunology. 2008;8(6):458-466
  57. 57. Zheng L, Wen X-L. Gut microbiota and inflammatory bowel disease: The current status and perspectives. World Journal of Clinical Cases. 2021;9(2):321
  58. 58. Mentella MC, Scaldaferri F, Pizzoferrato M, Gasbarrini A, Miggiano GAD. Nutrition, IBD and gut microbiota: A review. Nutrients. 2020;12(4):944
  59. 59. Alameddine J, Godefroy E, Papargyris L, Sarrabayrouse G, Tabiasco J, Bridonneau C, et al. Faecalibacterium prausnitzii skews human DC to prime IL10-producing T cells through TLR2/6/JNK signaling and IL-10, IL-27, CD39, and IDO-1 induction. Frontiers in Immunology. 2019;10:143
  60. 60. Patterson AM, Mulder IE, Travis AJ, Lan A, Cerf-Bensussan N, Gaboriau-Routhiau V, et al. Human gut symbiont Roseburia hominis promotes and regulates innate immunity. Frontiers in Immunology. 2017;8:1166
  61. 61. Mukherjee A, Lordan C, Ross RP, Cotter PD. Gut microbes from the phylogenetically diverse genus Eubacterium and their various contributions to gut health. Gut Microbes. 2020;12(1):1802866
  62. 62. An D, Oh SF, Olszak T, Neves JF, Avci FY, Erturk-Hasdemir D, et al. Sphingolipids from a symbiotic microbe regulate homeostasis of host intestinal natural killer T cells. Cell. 2014;156(1-2):123-133
  63. 63. Imaoka A, Shima T, Kato K, Mizuno S, Uehara T, Matsumoto S, et al. Anti-inflammatory activity of probiotic Bifidobacterium: Enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells. World Journal of Gastroenterology: WJG. 2008;14(16):2511
  64. 64. Bang C, Weidenbach K, Gutsmann T, Heine H, Schmitz RA. The intestinal archaea Methanosphaera stadtmanae and Methanobrevibacter smithii activate human dendritic cells. PLoS One. 2014;9(6):e99411
  65. 65. Martinez-Medina M, Garcia-Gil LJ. Escherichia coli in chronic inflammatory bowel diseases: An update on adherent invasive Escherichia coli pathogenicity. World Journal of Gastrointestinal Pathophysiology. 2014;5(3):213
  66. 66. Zuo T, Ng SC. The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease. Frontiers in Microbiology. 2018;2247: 1-13
  67. 67. Shin N-R, Whon TW, Bae J-W. Proteobacteria: Microbial signature of dysbiosis in gut microbiota. Trends in Biotechnology. 2015;33(9):496-503
  68. 68. Henke MT, Kenny DJ, Cassilly CD, Vlamakis H, Xavier RJ, Clardy J. Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide. Proceedings of the National Academy of Sciences. 2019;116(26):12672-12677
  69. 69. Bashir A, Miskeen AY, Hazari YM, Asrafuzzaman S, Fazili KM. Fusobacterium nucleatum, inflammation, and immunity: The fire within human gut. Tumor Biology. 2016;37(3):2805-2810
  70. 70. Engevik M, Danhof H, Britton R, Versalovic J. 20 elucidating the role of FUSOBACTERIUM NUCLEATUM IN intestinal inflammation. Inflammatory Bowel Diseases. 2020;26(Supplement_1):S29-S2S
  71. 71. Gringhuis SI, Wevers BA, Kaptein TM, Van Capel TM, Theelen B, Boekhout T, et al. Selective C-Rel activation via Malt1 controls anti-fungal TH-17 immunity by dectin-1 and dectin-2. PLoS Pathogens. 2011;7(1):e1001259
  72. 72. Norman JM, Handley SA, Baldridge MT, Droit L, Liu CY, Keller BC, et al. Disease-specific alterations in the enteric virome in inflammatory bowel disease. Cell. 2015;160(3):447-460
  73. 73. Lopetuso LR, Ianiro G, Scaldaferri F, Cammarota G, Gasbarrini A. Gut virome and inflammatory bowel disease. Inflammatory Bowel Diseases. 2016;22(7):1708-1712
  74. 74. Virgin HW. The virome in mammalian physiology and disease. Cell. 2014;157(1):142-150
  75. 75. Frank DN, Amand ALS, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proceedings of the National Academy of Sciences. 2007;104(34):13780-13785
  76. 76. Kamada N, Seo S-U, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nature Reviews Immunology. 2013;13(5):321-335
  77. 77. Carvalho FA, Koren O, Goodrich JK, Johansson ME, Nalbantoglu I, Aitken JD, et al. Transient inability to manage proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell host & Microbe. 2012;12(2):139-152
  78. 78. Joossens M, Huys G, Cnockaert M, De Preter V, Verbeke K, Rutgeerts P, et al. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut. 2011;60(5):631-637
  79. 79. Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655-662
  80. 80. Pittayanon R, Lau JT, Leontiadis GI, Tse F, Yuan Y, Surette M, et al. Differences in gut microbiota in patients with vs without inflammatory bowel diseases: A systematic review. Gastroenterology. 2020;158(4):930-46. e1
  81. 81. Zhang L, Liu F, Xue J, Lee SA, Liu L, Riordan SM. Bacterial species associated with human inflammatory bowel disease and their pathogenic mechanisms. Frontiers in Microbiology. 2022;13
  82. 82. Vakili B, Fateh A, Asadzadeh Aghdaei H, Sotoodehnejadnematalahi F, Siadat SD. Characterization of gut microbiota in hospitalized patients with Clostridioides difficile infection. Current Microbiology. 2020;77(8):1673-1680
  83. 83. Shoaei P, Shojaei H, Siadat SD, Moshiri A, Vakili B, Yadegari S, et al. Gut microbiota in burned patients with Clostridioides difficile infection. Burns. 2021
  84. 84. Shoaei P, Shojaei H, Jalali M, Khorvash F, Hosseini SM, Ataei B, et al. Clostridium difficile isolated from faecal samples in patients with ulcerative colitis. BMC Infectious Diseases. 2019;19(1):1-7
  85. 85. Shoaei P, Shojaei H, Khorvash F, Hosseini SM, Ataei B, Tavakoli H, et al. Molecular epidemiology of Clostridium difficile infection in Iranian hospitals. Antimicrobial Resistance & Infection Control. 2019;8(1):1-7
  86. 86. Azimi T, Nasiri MJ, Chirani AS, Pouriran R, Dabiri H. The role of bacteria in the inflammatory bowel disease development: A narrative review. APMIS. 2018;126(4):275-283
  87. 87. Clayton EM, Rea MC, Shanahan F, Quigley EM, Kiely B, Hill C, et al. The vexed relationship between Clostridium difficile and inflammatory bowel disease: An assessment of carriage in an outpatient setting among patients in remission. Official journal of the American College of Gastroenterology| ACG. 2009;104(5):1162-1169
  88. 88. Shoaei P, Shojaei H, Khorvash F, Ataei B, Esfandiari Z, Vakili B, et al. Clostridium difficile infection in cancer patients with hospital acquired diarrhea at the teaching hospitals in Iran: Multilocus sequence typing analysis (MLST) and antimicrobial resistance pattern. Annali di Igiene. 2019;31(4):365-373
  89. 89. Vakili B, Fateh A, Aghdaei HA, Sotoodehnejadnematalahi F, Siadat SD. Intestinal microbiota in elderly inpatients with Clostridioides difficile infection. Infection and Drug Resistance. 2020;13:2723
  90. 90. Monaghan TM, Cockayne A, Mahida YR. Pathogenesis of Clostridium difficile infection and its potential role in inflammatory bowel disease. Inflammatory Bowel Diseases. 2015;21(8):1957-1966
  91. 91. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature. 2015;517(7533):205-208
  92. 92. Irving PM, de Lusignan S, Tang D, Nijher M, Barrett K. Risk of common infections in people with inflammatory bowel disease in primary care: A population-based cohort study. BMJ open Gastroenterology. 2021;8(1):e000573
  93. 93. Uchiya K-i, Tomida S, Nakagawa T, Asahi S, Nikai T, Ogawa K. Comparative genome analyses of Mycobacterium avium reveal genomic features of its subspecies and strains that cause progression of pulmonary disease. Scientific Reports. 2017;7(1):1-14
  94. 94. Rath T, Roderfeld M, Blöcher S, Rhode A, Basler T, Akineden Ö, et al. Presence of intestinal Mycobacterium avium subspecies paratuberculosis (MAP) DNA is not associated with altered MMP expression in ulcerative colitis. BMC Gastroenterology. 2011;11(1):1-19
  95. 95. Quiding-Järbrink M, Smith DA, Bancroft GJ. Production of matrix metalloproteinases in response to mycobacterial infection. Infection and Immunity. 2001;69(9):5661-5670
  96. 96. Agrawal G, Hamblin H, Clancy A, Borody T. Anti-mycobacterial antibiotic therapy induces remission in active pædiatric Crohn’s disease. Microorganisms. 2020;8(8):1112
  97. 97. Feller M, Huwiler K, Schoepfer A, Shang A, Furrer H, Egger M. Long-term antibiotic treatment for Crohn's disease: Systematic review and meta-analysis of placebo-controlled trials. Clinical Infectious Diseases. 2010;50(4):473-480
  98. 98. Chen Y, Blaser MJ. Helicobacter pylori colonization is inversely associated with childhood asthma. The Journal of Infectious Diseases. 2008;198(4):553-560
  99. 99. Reibman J, Marmor M, Filner J, Fernandez-Beros M-E, Rogers L, Perez-Perez GI, et al. Asthma is inversely associated with helicobacter pylori status in an urban population. PLoS One. 2008;3(12):e4060
  100. 100. Kao JY, Rathinavelu S, Eaton KA, Bai L, Zavros Y, Takami M, et al. Helicobacter pylori-secreted factors inhibit dendritic cell IL-12 secretion: A mechanism of ineffective host defense. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2006;291(1):G73-G81
  101. 101. Rad R, Brenner L, Bauer S, Schwendy S, Layland L, da Costa CP, et al. CD25+/Foxp3+ T cells regulate gastric inflammation and helicobacter pylori colonization in vivo. Gastroenterology. 2006;131(2):525-537
  102. 102. Jafari F, Shokrzadeh L, Dabiri H, Baghaei K, Yamaoka Y, Zojaji H, et al. vacA genotypes of helicobacter pylori in relation to cagA status and clinical outcomes in Iranian populations. Japanese Journal of Infectious Diseases. 2008;61(4):290
  103. 103. Bohr UR, Glasbrenner B, Primus A, Zagoura A, Wex T, Malfertheiner P. Identification of enterohepatic helicobacter species in patients suffering from inflammatory bowel disease. Journal of Clinical Microbiology. 2004;42(6):2766-2768
  104. 104. Basset C, Holton J, Bazeos A, Vaira D, Bloom S. Are helicobacter species and enterotoxigenic Bacteroides fragilis involved in inflammatory bowel disease? Digestive Diseases and Sciences. 2004;49(9):1425-1432
  105. 105. Zhang L. Oral campylobacter species: Initiators of a subgroup of inflammatory bowel disease? World Journal of Gastroenterology: WJG. 2015;21(31):9239
  106. 106. Man SM, Zhang L, Day AS, Leach ST, Lemberg DA, Mitchell H. Campylobacter concisus and other campylobacter species in children with newly diagnosed Crohn's disease. Inflammatory Bowel Diseases. 2010;16(6):1008-1016
  107. 107. Zhang L, Lee H, Grimm MC, Riordan SM, Day AS, Lemberg DA. Campylobacter concisus and inflammatory bowel disease. World Journal of Gastroenterology: WJG. 2014;20(5):1259
  108. 108. Nielsen HL, Nielsen H, Ejlertsen T, Engberg J, Günzel D, Zeitz M, et al. Oral and fecal campylobacter concisus strains perturb barrier function by apoptosis induction in HT-29/B6 intestinal epithelial cells. PLoS One. 2011;6(8):e23858
  109. 109. Liu H, Hong XL, Sun TT, Huang XW, Wang JL, Xiong H. Fusobacterium nucleatum exacerbates colitis by damaging epithelial barriers and inducing aberrant inflammation. Journal of Digestive Diseases. 2020;21(7):385-398
  110. 110. Darfeuille-Michaud A, Boudeau J, Bulois P, Neut C, Glasser A-L, Barnich N, et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology. 2004;127(2):412-421
  111. 111. Dreux N, Denizot J, Martinez-Medina M, Mellmann A, Billig M, Kisiela D, et al. Point mutations in FimH adhesin of Crohn's disease-associated adherent-invasive Escherichia coli enhance intestinal inflammatory response. PLoS Pathogens. 2013;9(1):e1003141
  112. 112. Lapaquette P, Bringer MA, Darfeuille-Michaud A. Defects in autophagy favour adherent-invasive Escherichia coli persistence within macrophages leading to increased pro-inflammatory response. Cellular Microbiology. 2012;14(6):791-807
  113. 113. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document: The international scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nature reviews Gastroenterology & Hepatology. 2014;11:506-514
  114. 114. Abraham BP, Quigley EM. Probiotics in inflammatory bowel disease. Gastroenterology Clinics. 2017;46(4):769-782
  115. 115. Mishra J, Stubbs M, Kuang L, Vara N, Kumar P, Kumar N. Inflammatory bowel disease therapeutics: A focus on probiotic engineering. Mediators of Inflammation. 2022:2022
  116. 116. Akram W, Garud N, Joshi R. Role of inulin as prebiotics on inflammatory bowel disease. Drug discoveries & therapeutics. 2019;13(1):1-8
  117. 117. Colombel J-F, Shin A, Gibson PR. AGA clinical practice update on functional gastrointestinal symptoms in patients with inflammatory bowel disease: Expert review. Clinical Gastroenterology and Hepatology. 2019;17(3):380-90. e1
  118. 118. Jakubczyk D, Leszczyńska K, Górska S. The effectiveness of probiotics in the treatment of inflammatory bowel disease (IBD)—A critical review. Nutrients. 2020;12(7):1973
  119. 119. Doron S, Snydman DR. Risk and safety of probiotics. Clinical Infectious Diseases. 2015;60(suppl_2):S129-SS34
  120. 120. Freedman SB, Williamson-Urquhart S, Farion KJ, Gouin S, Willan AR, Poonai N, et al. Multicenter trial of a combination probiotic for children with gastroenteritis. New England Journal of Medicine. 2018;379(21):2015-2026
  121. 121. Bjarnason I, Sission G, Hayee BH. A randomised, double-blind, placebo-controlled trial of a multi-strain probiotic in patients with asymptomatic ulcerative colitis and Crohn’s disease. Inflammopharmacology. 2019;27(3):465-473
  122. 122. Bian X, Wu W, Yang L, Lv L, Wang Q , Li Y, et al. Administration of Akkermansia muciniphila ameliorates dextran sulfate sodium-induced ulcerative colitis in mice. Frontiers in Microbiology. 2019;2259:1-18
  123. 123. Wang L, Tang L, Feng Y, Zhao S, Han M, Zhang C, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8+ T cells in mice. Gut. 2020;69(11):1988-1997
  124. 124. Eindor-Abarbanel A, Healey GR, Jacobson K. Therapeutic advances in gut microbiome modulation in patients with inflammatory bowel disease from pediatrics to adulthood. International Journal of Molecular Sciences. 2021;22(22):12506
  125. 125. Lee J-Y, Cevallos SA, Byndloss MX, Tiffany CR, Olsan EE, Butler BP, et al. High-fat diet and antibiotics cooperatively impair mitochondrial bioenergetics to trigger dysbiosis that exacerbates pre-inflammatory bowel disease. Cell host & Microbe. 2020;28(2):273-84. e6
  126. 126. Oka A, Sartor RB. Microbial-based and microbial-targeted therapies for inflammatory bowel diseases. Digestive Diseases and Sciences. 2020;65(3):757-788
  127. 127. Roda G, Jharap B, Neeraj N, Colombel J-F. Loss of response to anti-TNFs: Definition, epidemiology, and management. Clinical and Translational Gastroenterology. 2016;7(1):e135
  128. 128. Paramsothy S, Paramsothy R, Rubin DT, Kamm MA, Kaakoush NO, Mitchell HM, et al. Faecal microbiota transplantation for inflammatory bowel disease: A systematic review and meta-analysis. Journal of Crohn's and Colitis. 2017;11(10):1180-1199
  129. 129. Hvas CL, Jørgensen SMD, Jørgensen SP, Storgaard M, Lemming L, Hansen MM, et al. Fecal microbiota transplantation is superior to fidaxomicin for treatment of recurrent Clostridium difficile infection. Gastroenterology. 2019;156(5):1324-32. e3
  130. 130. Khoruts A, Sadowsky MJ. Understanding the mechanisms of faecal microbiota transplantation. Nature reviews Gastroenterology & Hepatology. 2016;13(9):508-516
  131. 131. Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JR, Kassam Z, et al. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut. 2019;68(12):2111-2121
  132. 132. Draper LA, Ryan FJ, Smith MK, Jalanka J, Mattila E, Arkkila P, et al. Long-term colonisation with donor bacteriophages following successful faecal microbial transplantation. Microbiome. 2018;6(1):1-9

Written By

Bahareh Vakili, Parisa Shoaei, Zahra Esfandiari and Seyed Davar Siadat

Submitted: 01 May 2022 Reviewed: 13 June 2022 Published: 17 September 2022