Letter to the Editor Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jan 14, 2025; 31(2): 100827
Published online Jan 14, 2025. doi: 10.3748/wjg.v31.i2.100827
Exploring gut microbiota as a novel therapeutic target in Crohn’s disease: Insights and emerging strategies
Tong Qiao, Department of Clinical Medicine, Jinan University, Guangzhou 510632, Guangdong Province, China
Xian-Hui Wen, College of Life Science and Technology, Jinan University, Guangzhou 510632, Guangdong Province, China
ORCID number: Xian-Hui Wen (0009-0002-0905-8738).
Author contributions: Qiao T wrote the first draft of the manuscript; Wen XH reviewed and revised the final manuscript. All authors listed on the manuscript have read and approved the final version of the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Xian-Hui Wen, PhD, College of Life Science and Technology, Jinan University, No. 601 Huangpu Avenue West, Tianhe District, Guangzhou 510632, Guangdong Province, China. 17818521036@163.com
Received: August 28, 2024
Revised: September 30, 2024
Accepted: November 15, 2024
Published online: January 14, 2025
Processing time: 112 Days and 22 Hours

Abstract

Extensive research has investigated the etiology of Crohn’s disease (CD), encompassing genetic predisposition, lifestyle factors, and environmental triggers. Recently, the gut microbiome, recognized as the human body’s second-largest gene pool, has garnered significant attention for its crucial role in the pathogenesis of CD. This paper investigates the mechanisms underlying CD, focusing on the role of ‘creeping fat’ in disease progression and exploring emerging therapeutic strategies, including fecal microbiota transplantation, enteral nutrition, and therapeutic diets. Creeping fat has been identified as a unique pathological feature of CD and has recently been found to be associated with dysbiosis of the gut microbiome. We characterize this dysbiotic state by identifying key microbiome-bacteria, fungi, viruses, and archaea, and their contributions to CD pathogenesis. Additionally, this paper reviews contemporary therapies, emphasizing the potential of biological therapies like fecal microbiota transplantation and dietary interventions. By elucidating the complex interactions between host-microbiome dynamics and CD pathology, this article aims to advance our understanding of the disease and guide the development of more effective therapeutic strategies for managing CD.

Key Words: Crohn’s disease; Gut microbiome; Dysbiosis; Creeping fat; Fecal microbiota transplantation

Core Tip: Crohn’s disease (CD) represents a complex and challenging medical condition, characterized by the intricate interplay between genetic predisposition, gut microbiota dysbiosis, and immune dysregulation. This paper discusses the key effects of the gut microbiota on CD, as well as how they affect creeping fat. Furthermore, the paper highlights the urgent need to address gut microbiota imbalances, particularly focusing on pathogenic species and their mechanisms, as a critical therapeutic target. The findings underscore the importance of a personalized approach to CD management, emphasizing the need for continued research and innovation to address this complex disease.



TO THE EDITOR

In this article, we review a recently published article by Wu et al[1] in the World Journal of Gastroenterology, titled “Role of gut microbiota in Crohn’s disease pathogenesis: Insights from fecal microbiota transplantation in mouse model”. The study delves into the disrupted characteristics of mucosal-associated lymphoid tissue (MAT) and intestinal tissues in Crohn’s disease (CD) that were directly observed through hematoxylin and eosin (HE) staining, Masson’s trichrome staining, and immunohistochemical staining. Furthermore, by establishing mouse models, they detected excessive expression of proinflammatory factors in creeping fat (CrF), which robustly validates the previous understanding of the role of CrF in the pathogenesis of CD. This paper outlines the critical contributions of gut microbiome studies to the pathogenesis of CD, with a particular focus on emerging therapies targeting the intestinal flora, notably fecal microbiota transplantation (FMT) and dietary interventions. Special emphasis is placed on the significance of gut microbiome research in elucidating disease mechanisms and providing novel perspectives and potential solutions for the treatment of CD.

CD AND ITS PATHOLOGICAL CHARACTERISTICS
CD

CD, one of the major entities within the spectrum of inflammatory bowel diseases (IBDs), commonly affecting the terminal ileum and proximal colon[2]. Most patients present with an inflammatory phenotype at diagnosis, but over time, complications such as fibrosis, strictures, fistulas, or abscesses may develop, often leading to the need for surgery[3].

Etiological analysis

Despite significant advancements in experimental techniques and models, the etiology of IBD remains unclear. Higher incidence rates of IBD observed in monozygotic and dizygotic twins, as well as among certain families, strengthen the case for a genetic influence on the disease. Additionally, researchers have identified hundreds of IBD-associated genetic polymorphisms and mutations through genome-wide association studies, further supporting the genetic underpinnings of IBD[4,5]. However, genetics alone is insufficient to account for the onset of the disease. With industrialization, changes in lifestyle, and urbanization in modern society, the incidence and prevalence of IBD have been on the rise[6]. Nonetheless, the timeframe of these developments is too short to be explained by genetic drift or natural selection. In this context, non-genetic factors such as environmental influences, higher ultra-processed food and lower unprocessed/minimally processed food intakes[7], smoking[8], and antibiotic use[9] all play a significant role in the pathogenesis of these diseases. In recent years, with the increasing depth of investigations into the gastrointestinal microbiota, more studies have indicated that dysbiosis of the gut microbiome is a significant factor in triggering and promoting the development of IBD.

CrF in CD

CrF is a hallmark of CD[10,11], characterized by the abnormal expansion of perienteric adipose tissue. This phenomenon involves the gradual encasement and infiltration of mesenteric fat around affected intestinal segments, resulting in a characteristic “creeping” appearance. A large number of cohort studies and clinical trials demonstrate that CrF exacerbates CD inflammation through various mechanisms[11-13] such as the release of pro-inflammatory molecules (leptin[14] and resistin[15]) and activation of immune cells (macrophages[16] and CD4+ T cells[17,18]). This is further supported by a few clinical trials that CrF promotes inflammation via SP/NKR1 activation and upregulating interleukin-17A (IL-17A) mRNA in preadipocytes and IL-17A receptor mRNA in colonic tissue[19,20].

It is worth noting that the gut microbiome, especially bacteria, may be a potential trigger for CrF. Emerging evidence suggests that CrF, the migration of MAT to sites of intestinal barrier dysfunction, may represent a protective response. This process aims to prevent systemic dissemination of potentially harmful bacteria that have translocated across the compromised intestinal epithelium. A study demonstrated that while bacterial translocation occurs in both normal MAT and CD-associated CrF, the characteristics and metabolic functions of the gut microbiota differ significantly[21]. For instance, a specific population of translocated enteric bacteria, i.e., Clostridium innocuum, is found in both the ileal mucosa and CrF. This bacterium promotes M2 macrophage polarization, stimulating adipose tissue remodeling and CrF formation[21]. Furthermore, adipocytes and preadipocytes within CrF express functional pattern recognition receptors, such as Toll-like receptors and nucleotide-binding oligomerization domain-containing protein 1[22]. These pattern recognition receptors recognize microbe-associated molecular patterns derived from translocated bacteria, triggering downstream signaling cascades that activate transcription factors like nuclear factor-κB (NF-κB) and induce the production of pro-inflammatory cytokines and chemokines[23]. Another study investigated whether microbial translocation in CD is a key factor in the progression of CrF[24]. The researchers identified a subset of mucosa-associated intestinal bacteria that consistently translocate during ileal resections in CD and remain viable in CrF; they pinpointed Clostridium innocuum as a marker of this population, noting strain variation between mucosal and adipose tissue isolates, which suggests a preference for lipid-rich environments[24]. Single-cell RNA sequencing revealed that CrF exhibits both pro-fibrotic and pro-adipogenic effects, along with an activated immune cell environment responsive to microbial stimuli. This was further confirmed in germ-free mice colonized with Clostridium innocuum[24].

Role and alterations of the gut microbiome in CD

The gut microbiota, including bacteria, fungi, viruses, and archaea, is closely related to the immune system and is of great importance to the normal physiological functions of the body[25,26]. Multiple studies have shown that gut microbiota dysbiosis is an important cause of the occurrence and aggravation of CD, and is closely related to the clinical manifestations of CD, such as inflammation and fibrosis. Wu et al[1] suggested a potential role of gut microbiota dysbiosis in the pathogenesis of CD, but their evidence was limited. Given the increasing focus on gut microbiota dysbiosis in CD research, we describe the impact of bacterial, fungal, viral, and archaeal dysbiosis, and their interactions, on CD development.

Intestinal bacterial microbiota in CD

The gut microbiome, especially bacteria, is the most extensively researched microbial community in patients with CD[27]. Studies using culture-independent methods (quantitative polymerase chain reaction, FISH analysis, 16S rRNA gene sequencing, shotgun-sequencing, and Illumina sequencing) on fecal samples consistently show that the observed decrease of some beneficial bacteria, such as Bacteroides and butyrate-producing Firmicutes, including Faecalibacterium prausnitzii and Roseburia spp., alongside an increase in pathogenic bacteria, may significantly contribute to pro-inflammatory effects[28,29]. Such dysbiosis can lead to the disruption of certain metabolites produced by the microbiota, subsequently increasing the inflammatory response. For instance, short-chain fatty acids, particularly butyrate, play a crucial role as a histone deacetylase inhibitor, effectively suppressing the activation of NF-κB and downregulating pro-inflammatory cytokines such as IL-1β, IL-6, and tumor necrosis factor (TNF)-α[29]. Secondary bile acids, which result from microbial metabolism, are critical in immune regulation, promoting regulatory T cell differentiation, and disruptions in bile acid metabolism worsen inflammation in IBD patients[30,31]. The reduced availability of tryptophan-derived metabolites, which modulate immune responses via aryl hydrocarbon receptors, correlates with higher disease susceptibility[32]. Sphingolipids, produced by both host and microbes, also regulate inflammation, with microbial sphingolipids like those from Bacteroides shown to protect against colitis by inhibiting natural killer T cell proliferation[33]. Furthermore, some clinical trials and relevant pre-clinical studies found that Faecalibacterium prausnitzii actively suppresses NF-κB activation and attenuates IL-8 production in intestinal epithelial cells via its secretory metabolites[28,34]. In addition, CD is characterized by an increased abundance of Ruminococcus gnavus and mucosa-associated adherent-invasive Escherichia coli (AIEC), which bind to and invade intestinal epithelial cells through mechanisms involving microtubule polymerization and actin recruitment, inducing the secretion of inflammatory cytokines, particularly the transcriptional levels of interferon-γ and IL-8[35]. These cytokines are closely related to the immune response during CD, indicating the significant role of AIEC in perpetuating intestinal inflammation. When surviving and replicating in macrophages, AIEC can also induce TNF-α secretion, further contributing to the inflammatory milieu in CD[36]. A multi-center clinical study encompassing Spanish and Belgian CD cohorts investigated fecal microbiota composition using sample collection, genomic DNA extraction, high-throughput 16S rRNA gene sequencing, and bioinformatic analysis[37]. The results indicated that the depletion of beneficial bacteria, rather than an overabundance of pathogenic bacteria, is more strongly associated with CD[37]. These beneficial bacteria include butyrate-producing species such as Faecalibacterium prausnitzii, members of the Lachnospiraceae family (e.g., Roseburia and Ruminococcus), and Oscillospira[37]. These findings support and extend previous reports of dysbiosis in CD[37].

Intestinal fungal microbiota in CD

Although the fungal community is far less abundant than bacteria, constituting only 0.1% of the gut microbiota[38], many studies have shown that fungi also play an important role in the pathogenesis of CD[39,40]. The abundance of Candida albicans, Malassezia restricta, and Debaryomyces hansenii, which are the most commonly detected fungal species in stool samples of CD subjects[41-43], shows significant changes in patients with CD. Recent studies have revealed that Malassezia restricta worsens colitis in mice through a caspase-recruitment domain 9-dependent mechanism. Upon Malassezia restricta recognition by C-type lectin receptors on immune cells, caspase-recruitment domain 9 is recruited to form a complex with B-cell lymphoma/leukemia 10 and mucosa-associated lymphoid tissue lymphoma translocation protein 1, activating the NF-κB pathway. This triggers production of TNF-α, IL-8, and other pro-inflammatory mediators, amplifying immune cell recruitment and inflammation[42]. Debaryomyces, specifically Debaryomyces hansenii, has been identified as being enriched in inflamed intestinal tissues of patients with CD and impairs colonic healing by modulating the myeloid cell-interferon γ-C-C chemokine ligand 5 signaling axis[43]. Compared to healthy individuals, patients with CD exhibit an elevated proportion of Candida albicans and a reduced proportion of Saccharomyces cerevisiae in their gut. Candida albicans, originally a symbiotic bacterium in the gut, can delay intestinal mucosal healing and increase the secretion of IL-17 and IL-23 in diseases such as CD, thereby exacerbating inflammation[44]. Saccharomyces cerevisiae, a common probiotic in the gut, can strongly inhibit the adhesion of AIEC to the brush border of intestinal epithelial cells in CD. Furthermore, it restores barrier function by blocking AIEC-induced expression of Claudin-2, which forms tight junctions in pores on the plasma membrane of intestinal epithelial cells. These effects are accompanied by a reduction in the release of pro-inflammatory cytokines IL-6, IL-1β, and KC from the intestinal mucosa[45]. Additionally, several studies have indeed found that high serum titers of anti-Saccharomyces cerevisiae antibodies (ASCA) [immunoglobulin G (IgG) and IgA], which are directed against yeast cell wall-associated mannan, serve as a clinical biomarker for CD. The IgG ASCA-positive rate is 60%-70% in patients with CD[46].

Fungal-bacterial interactions

With the deepening of research on the intestinal microbiota in recent years, the interaction between different species has gradually attracted attention. The coexistence of fungi and some specific intestinal bacteria may be crucial for the development or improvement of intestinal diseases. Recently, Seelbinder et al[47] confirmed that antibiotic treatment significantly altered the composition of fungi, demonstrating that bacterial dysbiosis may lead to fungal dysbiosis. This study also showed that the use of Candida albicans aggravated the severity of the disease, while the use of Saccharomyces boulardii alleviated the symptoms of the disease. A cohort study, including patients from Northern France-Belgium, found a positive correlation between the abundance of Candida tropicalis and both Streptococcus mitis and Escherichia coli[48]. Additionally, the study showed an association between the levels of ASCA, a known biomarker for CD, and the abundance of Candida tropicalis[48].

Intestinal viruses and archaea in CD

The role of virome in CD may promote intestinal inflammation. CD is associated with an increase in temperate phages, critical for gut bacterial diversity and fitness, impacting microbial balance and CD progression. The “core phage” comprises double-stranded DNA Caudovirales (Myoviridae, Podoviridae, and Siphoviridae) and single-strand DNA Microviridae. CD patients exhibit altered viromes, with decreased Microviridae and Virgaviridae, and increased Caudovirales, Alteronomonadales, and Clostridiales phages. However, the specific virus-CD relationship remains elusive[49,50]. There is a paucity of relevant research in this area, and in vitro studies have demonstrated that bacteriophages can stimulate macrophages to induce MyD88-dependent pro-inflammatory cytokine production[51]. Besides, bacteriophages may also play a therapeutic role in CD treatment. A cocktail of three bacteriophages was demonstrated to reduce symptoms and significantly reduce AIEC in dextran sodium sulfate-induced colitis in mice[52]. Currently, research on archaea in CD is limited, with the majority of studies focusing on methanogens, which constitute approximately 10% of the intestinal anaerobic microbiota[53]. For instance, the presence of methanogens and the role of methane as a signaling molecule have been shown to prolong the contact time between the mucosa and toxic metabolic byproducts such as hydrogen sulfide. This extended exposure can enhance their absorption by the intestinal epithelium or lead to damage to the intestinal epithelial barrier, thereby increasing the risk of pathogen translocation[54]. Methanobrevibacter smithii is the most abundant species among methanogens, which accounts for approximately 94% of them, and recent studies have revealed that Methanobrevibacter smithii has been associated with increased acetate production[54,55] and acetate may exert a proinflammatory effect[56]. Additionally, the release of single-strand DNA from Methanosphaera stadtmanae, recognized as a microbe-associated molecular pattern, has been shown to stimulate the secretion of significant amounts of proinflammatory cytokines, such as IL-1β, TNF-α, and type-I and type-III interferons, through the activation of Toll-like receptor 8[57].

Virus-bacterial interactions

The trans-cohort study by Cao et al[58] describes the cross-relationship between 50 types of bacteriophages and 30 species of bacteria, revealing a significant dysbiosis in the bacteriophage-bacteria ecological network of the small intestinal mucosa in patients with CD. Specifically, the phage-bacteria interaction network, which is typically centered around Bifidobacterium and Lachnospiraceae in healthy individuals, is significantly weakened, while the phage-bacteria interaction network centered around Prevotella is markedly strengthened in CD. Another study has focused on the virus-bacteria interaction network, identifying a positive correlation between DNA bacteriophages and certain bacteria (such as Enterobacter cloacae and Escherichia coli), while showing a negative correlation with Faecalibacterium prausnitzii[59]. Furthermore, these studies have noted distinct changes in virus-bacteria associations under different disease states, including a close correlation of torquetenovirus, Bacteriophage spp., and Bacteroides phage B124-14 with fecal bacteria during active disease periods, which significantly diminishes during remission.

MICROBIOTA TRANSPLANTATION AND NUTRITIONAL THERAPY IN CD TREATMENT EFFICACY AND MECHANISMS

For the intractable chronic inflammation in CD, as well as severe complications such as intestinal stenosis and fistulas, various targeted surgical options and biological agents have been developed so far. With the updating of the concept of targeted drugs in recent years, some targeted therapies with theoretically better therapeutic effects and fewer side effects, such as FMT and dietary interventions, have gradually been developed and put into clinical use, providing more options for the treatment of CD from a new perspective.

Conventional therapy

Approximately 15%-50% of patients with CD will require surgery within ten years following the diagnosis[60], including fistula repair, colectomy and ileostomy, proctectomy, and strictureplasty[2,61,62]. Notably, there is evidence indicating that biological therapy is more effective if introduced earlier in the disease course, preparing for early intervention[2]. The selection of medical therapy is determined by the patient’s risk profile and the severity of the disease. Mild-to-moderate disease can be treated by oral mesalamine, immunomodulators like thiopurines (6-mercaptopurine and azathioprine), methotrexate, and steroids[63]. Moderate-to-severe disease (including fistulizing disease) is best treated with combined immunomodulators and biologics or biologics alone, including budesonide (corticosteroids), azathioprine, and mercaptopurine (immunomodulators)[64]. Anti-TNF agents (vedolizumab, ustekinumab, and infliximab) block the downstream effects of the TNF inflammatory cascade. These agents are efficacious in steroid-resistant or immunomodulator-refractory CD[65-67]. Notably, combination therapy with an immunomodulator and anti-TNF is more effective than monotherapy with either medication. Anti-IL-12/23 agents (ustekinumab and risankizumab) are efficacious in patients who failed prior corticosteroid, immunomodulator, or anti-TNF treatment.

Emerging biological therapies

Despite demonstrating efficacy in managing CD, conventional therapeutic interventions are correlated with a myriad of adverse reactions, encompassing metabolic disorders, serious infections, and dermatitis, etc.[68,69]. For instance, corticosteroids have the risk of causing cystoid appearance and hepatic steatosis, while anti-tumor necrosis factor therapy may cause severe bacterial infections in the lungs and a psoriatic eczema-like skin reaction. Initial analysis of early treatment regimens showed that some of these treatments disrupted the host gut microbiome, an occurrence linked to increased pathogenesis of CD[70-72]. Thus, introducing an adjuvant or supporting therapy that can modulate the gut microbiota and prevent gut microbiome dysbiosis may be a novel approach to supporting the management of CD. Not only does this approach have the potential of restoring the microbiome balance and it may also help achieve better response to the currently used medications[73,74].

Dietary therapy for maintenance of remission in CD

As the pathogenesis of CD becomes clearer, dietary factors are increasingly recognized for their role in disease management. Exclusive enteral nutrition (EEN) is well-established as an effective therapy for inducing remission in pediatric CD, with success rates of 60% to 80%[75]. EEN not only avoids the adverse effects of steroids but also improves nutritional status, promotes mucosal healing, and manages complications like strictures and fistulae[76-79], potentially reducing the need for surgery. In adult CD patients, corticosteroids and biologics are typically first-line treatments. However, a high-quality study has proposed EEN as an alternative therapy for adults, aiming to mitigate or avoid the side effects of conventional treatments and offering a standardized protocol for future research[80]. Despite its benefits, EEN has limitations, including its limited long-term effects on the gut microbiota and poor palatability, which hinder widespread adoption[81]. To address these limitations, alternative dietary strategies, such as partial enteral nutrition (PEN), low FODMAP diets, the specific carbohydrate diet, and Mediterranean diets, have been explored[82-84]. The efficacy of these interventions remains debated, and they are rarely used as standalone treatments[85].

The CD exclusion diet (CDED), characterized by low fat and animal protein and high carbohydrates and dietary fiber, has emerged as a promising alternative to EEN[86-88]. Clinical trials in pediatric CD patients have shown that CDED, combined with PEN, achieves remission rates comparable to EEN[89-91]. Recent studies indicate that CDED, alone or with PEN, effectively induces remission, reduces inflammation, and produces lasting changes in the gut microbiota[88,92]. Specifically, the combination of PEN and CDED reduces Actinobacteria and Proteobacteria while increasing beneficial Clostridia species after six weeks. Unlike EEN, which often sees microbiota reverting to baseline after remission, CDED-induced changes persist for 12 weeks post-therapy, suggesting a more sustainable approach to gut microbiota modulation and CD management[86]. It is noteworthy that the 2024 European Crohn’s and Colitis Organization Guidelines on pharmacological treatment of CD have emphasized the importance of dietary therapy, offering a highly valuable option for personalized treatment of CD.

FMT

FMT involves the processing of stool from a healthy donor into a therapeutic formulation that is then administered to individuals with gut microbiota dysbiosis, with the aim of ameliorating the imbalance in the patient’s intestinal microbiota. FMT has shown remarkable efficacy in treating refractory Clostridioides difficile infection[93-96], with a 90% success rate in the treatment of antibiotic recurrence cases. Given the critical impact of gut microbiota dysbiosis on the pathogenesis of CD, FMT is currently being explored as a therapeutic option for IBD.

A retrospective study integrated 11 high-quality CD reports involving a total of 106 CD patients and conducted a meta-analysis. The pooled clinical remission rate with a random effects model across these 11 reports was 0.62 (95% confidence interval: 0.48-0.81). Among them, seven studies reported a combined proportion of 0.79 for CD patients achieving clinical response after FMT treatment, with low heterogeneity ( = 43%). Additionally, metagenomic analysis revealed that pre-FMT patients displayed reduced species diversity and significant disparities in microbiome composition when compared to their respective donors. Notably, following FMT, clinical responders, in contrast to non-responders, developed significantly elevated species diversity, more closely aligning with the donor’s microbiome configuration[97]. Several additional meta-analyses also support this viewpoint. Gutin et al[98] reported that in FMT for CD patients, responders had a more abundant presence of Enterobacteriaceae and Bifidobacteriaceae members, while non-responders had a higher relative abundance of Lachnospiraceae and Ruminococcaceae members[98]. Kao et al[99] reported that several groups of bacteria disappeared after FMT (Enterococcus, Lactobacillus, Streptococcus, Burkholderia group, and Erysipelothrix group), while the abundance of strains beneficial for CD (Faecalibacterium and Roseburia) increased. This indicates that the overall efficacy of FMT in CD patients is relatively good based on these studies. Additionally, the postoperative complications including abdominal pain, bloating, diarrhea and constipation typically following FMT are mild. And Gutin et al[98] and Sokol et al[100] further verified that the adverse reactions observed in patients were not associated with FMT. This validates the favorable safety characteristics of FMT as a treatment modality. Wu et al[26] have validated in mice models that FMT from healthy donors can improve CD, whereas FMT from CD patients exacerbates the condition. These findings strongly support the aforementioned viewpoint.

FUTURE PERSPECTIVES

Future research should focus on the extensive coverage of diverse populations across different regions, including germ-free animal models and genetically edited animal samples, while integrating cross-sectional and longitudinal cohort studies to ensure sample diversity and representativeness. The identification of key microbial strains and the development of related research methodologies should be expanded, emphasizing the application of metagenomic single nucleotide polymorphism/single-nucleotide variation analysis workflows, such as GT-Pro, and the utilization of AI algorithms to construct multidimensional diagnostic models linking species, genes, and single-nucleotide variations. The research should incorporate culture-based omics, bacterial gene editing technologies, and germ-free animal models to minimize and control the impact of confounding variables in experiments. By systematically summarizing research findings and validating causal relationships in animal models, further clinical trials can be conducted. Based on these results, treatment protocols for celiac disease should be optimized to achieve more precise early diagnosis, complemented by advanced imaging techniques to expedite disease assessment and promote the development of personalized therapeutic strategies. Microbiome-based therapeutic approaches, such as FMT and dietary interventions, have already been implemented in clinical settings and have shown promising therapeutic outcomes. These strategies aim to more effectively alleviate disease symptoms, reduce adverse events during treatment, and enhance the quality of life for patients.

CONCLUSION

CD, a complex chronic inflammatory bowel disorder, is characterized by its unique symptom of CrF. Dysbiosis of the gut microbiome exerts profound influence on the progression of the disease. Traditional therapies often exacerbate microbial imbalance and are accompanied by numerous adverse effects. Microbiome-based targeted therapies, with their unique efficacy and safety profiles, have paved a new avenue for personalized treatment of CD.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report’s classification

Scientific Quality: Grade C, Grade C

Novelty: Grade B, Grade C

Creativity or Innovation: Grade C, Grade C

Scientific Significance: Grade B, Grade B

P-Reviewer: Chen K; Feng R S-Editor: Wang JJ L-Editor: Wang TQ P-Editor: Zheng XM

References
1.  Wu Q, Yuan LW, Yang LC, Zhang YW, Yao HC, Peng LX, Yao BJ, Jiang ZX. Role of gut microbiota in Crohn's disease pathogenesis: Insights from fecal microbiota transplantation in mouse model. World J Gastroenterol. 2024;30:3689-3704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
2.  Dolinger M, Torres J, Vermeire S. Crohn's disease. Lancet. 2024;403:1177-1191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
3.  Tsai L, Ma C, Dulai PS, Prokop LJ, Eisenstein S, Ramamoorthy SL, Feagan BG, Jairath V, Sandborn WJ, Singh S. Contemporary Risk of Surgery in Patients With Ulcerative Colitis and Crohn's Disease: A Meta-Analysis of Population-Based Cohorts. Clin Gastroenterol Hepatol. 2021;19:2031-2045.e11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 147]  [Article Influence: 36.8]  [Reference Citation Analysis (0)]
4.  Santos MPC, Gomes C, Torres J. Familial and ethnic risk in inflammatory bowel disease. Ann Gastroenterol. 2018;31:14-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 22]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
5.  Brant SR. Update on the heritability of inflammatory bowel disease: the importance of twin studies. Inflamm Bowel Dis. 2011;17:1-5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 76]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
6.  Wang R, Li Z, Liu S, Zhang D. Global, regional and national burden of inflammatory bowel disease in 204 countries and territories from 1990 to 2019: a systematic analysis based on the Global Burden of Disease Study 2019. BMJ Open. 2023;13:e065186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 141]  [Reference Citation Analysis (0)]
7.  Narula N, Chang NH, Mohammad D, Wong ECL, Ananthakrishnan AN, Chan SSM, Carbonnel F, Meyer A. Food Processing and Risk of Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol. 2023;21:2483-2495.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 30]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
8.  Jones DP, Richardson TG, Davey Smith G, Gunnell D, Munafò MR, Wootton RE. Exploring the Effects of Cigarette Smoking on Inflammatory Bowel Disease Using Mendelian Randomization. Crohns Colitis 360. 2020;2:otaa018.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
9.  Faye AS, Allin KH, Iversen AT, Agrawal M, Faith J, Colombel JF, Jess T. Antibiotic use as a risk factor for inflammatory bowel disease across the ages: a population-based cohort study. Gut. 2023;72:663-670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 35]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
10.  Dickson I. Creeping fat in Crohn's disease explained. Nat Rev Gastroenterol Hepatol. 2020;17:713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
11.  Aggeletopoulou I, Tsounis EP, Mouzaki A, Triantos C. Creeping Fat in Crohn's Disease-Surgical, Histological, and Radiological Approaches. J Pers Med. 2023;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
12.  Yin Y, Xie Y, Ge W, Li Y. Creeping fat formation and interaction with intestinal disease in Crohn's disease. United European Gastroenterol J. 2022;10:1077-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 7]  [Reference Citation Analysis (0)]
13.  Mao R, Kurada S, Gordon IO, Baker ME, Gandhi N, McDonald C, Coffey JC, Rieder F. The Mesenteric Fat and Intestinal Muscle Interface: Creeping Fat Influencing Stricture Formation in Crohn's Disease. Inflamm Bowel Dis. 2019;25:421-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 143]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
14.  Karmiris K, Koutroubakis IE, Xidakis C, Polychronaki M, Voudouri T, Kouroumalis EA. Circulating levels of leptin, adiponectin, resistin, and ghrelin in inflammatory bowel disease. Inflamm Bowel Dis. 2006;12:100-105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 229]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
15.  Bokarewa M, Nagaev I, Dahlberg L, Smith U, Tarkowski A. Resistin, an adipokine with potent proinflammatory properties. J Immunol. 2005;174:5789-5795.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 664]  [Cited by in F6Publishing: 713]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
16.  Lendeckel U, Venz S, Wolke C. Macrophages: shapes and functions. ChemTexts. 2022;8:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
17.  Luckheeram RV, Zhou R, Verma AD, Xia B. CD4⁺T cells: differentiation and functions. Clin Dev Immunol. 2012;2012:925135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 580]  [Cited by in F6Publishing: 847]  [Article Influence: 65.2]  [Reference Citation Analysis (0)]
18.  Suau R, Pardina E, Domènech E, Lorén V, Manyé J. The Complex Relationship Between Microbiota, Immune Response and Creeping Fat in Crohn's Disease. J Crohns Colitis. 2022;16:472-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
19.  Karagiannides I, Kokkotou E, Tansky M, Tchkonia T, Giorgadze N, O'Brien M, Leeman SE, Kirkland JL, Pothoulakis C. Induction of colitis causes inflammatory responses in fat depots: evidence for substance P pathways in human mesenteric preadipocytes. Proc Natl Acad Sci U S A. 2006;103:5207-5212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 66]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
20.  Sideri A, Bakirtzi K, Shih DQ, Koon HW, Fleshner P, Arsenescu R, Arsenescu V, Turner JR, Karagiannides I, Pothoulakis C. Substance P mediates pro-inflammatory cytokine release form mesenteric adipocytes in Inflammatory Bowel Disease patients. Cell Mol Gastroenterol Hepatol. 2015;1:420-432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Ha CWY, Martin A, Sepich-Poore GD, Shi B, Wang Y, Gouin K, Humphrey G, Sanders K, Ratnayake Y, Chan KSL, Hendrick G, Caldera JR, Arias C, Moskowitz JE, Ho Sui SJ, Yang S, Underhill D, Brady MJ, Knott S, Kaihara K, Steinbaugh MJ, Li H, McGovern DPB, Knight R, Fleshner P, Devkota S. Translocation of Viable Gut Microbiota to Mesenteric Adipose Drives Formation of Creeping Fat in Humans. Cell. 2020;183:666-683.e17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 178]  [Cited by in F6Publishing: 222]  [Article Influence: 44.4]  [Reference Citation Analysis (0)]
22.  Yin Y, Zhu ZX, Li Z, Chen YS, Zhu WM. Role of mesenteric component in Crohn's disease: A friend or foe? World J Gastrointest Surg. 2021;13:1536-1549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 1]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
23.  Kredel L, Batra A, Siegmund B. Role of fat and adipokines in intestinal inflammation. Curr Opin Gastroenterol. 2014;30:559-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
24.  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. Proc Natl Acad Sci U S A. 2019;116:12672-12677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 472]  [Article Influence: 78.7]  [Reference Citation Analysis (0)]
25.  Lopez-Siles M, Duncan SH, Garcia-Gil LJ, Martinez-Medina M. Faecalibacterium prausnitzii: from microbiology to diagnostics and prognostics. ISME J. 2017;11:841-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 348]  [Cited by in F6Publishing: 473]  [Article Influence: 59.1]  [Reference Citation Analysis (0)]
26.  Quévrain E, Maubert MA, Michon C, Chain F, Marquant R, Tailhades J, Miquel S, Carlier L, Bermúdez-Humarán LG, Pigneur B, Lequin O, Kharrat P, Thomas G, Rainteau D, Aubry C, Breyner N, Afonso C, Lavielle S, Grill JP, Chassaing G, Chatel JM, Trugnan G, Xavier R, Langella P, Sokol H, Seksik P. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease. Gut. 2016;65:415-425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 450]  [Cited by in F6Publishing: 528]  [Article Influence: 58.7]  [Reference Citation Analysis (0)]
27.  Levine A, Rhodes JM, Lindsay JO, Abreu MT, Kamm MA, Gibson PR, Gasche C, Silverberg MS, Mahadevan U, Boneh RS, Wine E, Damas OM, Syme G, Trakman GL, Yao CK, Stockhamer S, Hammami MB, Garces LC, Rogler G, Koutroubakis IE, Ananthakrishnan AN, McKeever L, Lewis JD. Dietary Guidance From the International Organization for the Study of Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol. 2020;18:1381-1392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 163]  [Article Influence: 32.6]  [Reference Citation Analysis (0)]
28.  Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux JJ, Blugeon S, Bridonneau C, Furet JP, Corthier G, Grangette C, Vasquez N, Pochart P, Trugnan G, Thomas G, Blottière HM, Doré J, Marteau P, Seksik P, Langella P. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731-16736.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2747]  [Cited by in F6Publishing: 3054]  [Article Influence: 179.6]  [Reference Citation Analysis (0)]
29.  Gevers D, Kugathasan S, Denson LA, Vázquez-Baeza Y, Van Treuren W, Ren B, Schwager E, Knights D, Song SJ, Yassour M, Morgan XC, Kostic AD, Luo C, González A, McDonald D, Haberman Y, Walters T, Baker S, Rosh J, Stephens M, Heyman M, Markowitz J, Baldassano R, Griffiths A, Sylvester F, Mack D, Kim S, Crandall W, Hyams J, Huttenhower C, Knight R, Xavier RJ. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe. 2014;15:382-392.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1945]  [Cited by in F6Publishing: 2213]  [Article Influence: 201.2]  [Reference Citation Analysis (0)]
30.  Hang S, Paik D, Yao L, Kim E, Trinath J, Lu J, Ha S, Nelson BN, Kelly SP, Wu L, Zheng Y, Longman RS, Rastinejad F, Devlin AS, Krout MR, Fischbach MA, Littman DR, Huh JR. Bile acid metabolites control T(H)17 and T(reg) cell differentiation. Nature. 2019;576:143-148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 456]  [Cited by in F6Publishing: 764]  [Article Influence: 127.3]  [Reference Citation Analysis (0)]
31.  Song X, Sun X, Oh SF, Wu M, Zhang Y, Zheng W, Geva-Zatorsky N, Jupp R, Mathis D, Benoist C, Kasper DL. Microbial bile acid metabolites modulate gut RORγ(+) regulatory T cell homeostasis. Nature. 2020;577:410-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 321]  [Cited by in F6Publishing: 605]  [Article Influence: 100.8]  [Reference Citation Analysis (0)]
32.  Nikolaus S, Schulte B, Al-Massad N, Thieme F, Schulte DM, Bethge J, Rehman A, Tran F, Aden K, Häsler R, Moll N, Schütze G, Schwarz MJ, Waetzig GH, Rosenstiel P, Krawczak M, Szymczak S, Schreiber S. Increased Tryptophan Metabolism Is Associated With Activity of Inflammatory Bowel Diseases. Gastroenterology. 2017;153:1504-1516.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 300]  [Cited by in F6Publishing: 354]  [Article Influence: 44.3]  [Reference Citation Analysis (0)]
33.  An D, Oh SF, Olszak T, Neves JF, Avci FY, Erturk-Hasdemir D, Lu X, Zeissig S, Blumberg RS, Kasper DL. Sphingolipids from a symbiotic microbe regulate homeostasis of host intestinal natural killer T cells. Cell. 2014;156:123-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 470]  [Cited by in F6Publishing: 440]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
34.  Giri R, Hoedt EC, Khushi S, Salim AA, Bergot AS, Schreiber V, Thomas R, McGuckin MA, Florin TH, Morrison M, Capon RJ, Ó Cuív P, Begun J. Secreted NF-κB suppressive microbial metabolites modulate gut inflammation. Cell Rep. 2022;39:110646.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 25]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
35.  Palmela C, Chevarin C, Xu Z, Torres J, Sevrin G, Hirten R, Barnich N, Ng SC, Colombel JF. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut. 2018;67:574-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 250]  [Cited by in F6Publishing: 338]  [Article Influence: 48.3]  [Reference Citation Analysis (0)]
36.  Zhao Z, Xu S, Zhang W, Wu D, Yang G. Probiotic Escherichia coli NISSLE 1917 for inflammatory bowel disease applications. Food Funct. 2022;13:5914-5924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 32]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
37.  Pascal V, Pozuelo M, Borruel N, Casellas F, Campos D, Santiago A, Martinez X, Varela E, Sarrabayrouse G, Machiels K, Vermeire S, Sokol H, Guarner F, Manichanh C. A microbial signature for Crohn's disease. Gut. 2017;66:813-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 454]  [Cited by in F6Publishing: 543]  [Article Influence: 67.9]  [Reference Citation Analysis (0)]
38.  Qin J, Li R, Raes J, Arumugam M, Burgdorf KS, Manichanh C, Nielsen T, Pons N, Levenez F, Yamada T, Mende DR, Li J, Xu J, Li S, Li D, Cao J, Wang B, Liang H, Zheng H, Xie Y, Tap J, Lepage P, Bertalan M, Batto JM, Hansen T, Le Paslier D, Linneberg A, Nielsen HB, Pelletier E, Renault P, Sicheritz-Ponten T, Turner K, Zhu H, Yu C, Li S, Jian M, Zhou Y, Li Y, Zhang X, Li S, Qin N, Yang H, Wang J, Brunak S, Doré J, Guarner F, Kristiansen K, Pedersen O, Parkhill J, Weissenbach J; MetaHIT Consortium, Bork P, Ehrlich SD, Wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7052]  [Cited by in F6Publishing: 7505]  [Article Influence: 500.3]  [Reference Citation Analysis (4)]
39.  Sendid B, Cornu M, Cordier C, Bouckaert J, Colombel JF, Poulain D. From ASCA breakthrough in Crohn's disease and Candida albicans research to thirty years of investigations about their meaning in human health. Autoimmun Rev. 2024;23:103486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
40.  Jain U, Ver Heul AM, Xiong S, Gregory MH, Demers EG, Kern JT, Lai CW, Muegge BD, Barisas DAG, Leal-Ekman JS, Deepak P, Ciorba MA, Liu TC, Hogan DA, Debbas P, Braun J, McGovern DPB, Underhill DM, Stappenbeck TS. Debaryomyces is enriched in Crohn's disease intestinal tissue and impairs healing in mice. Science. 2021;371:1154-1159.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 132]  [Article Influence: 33.0]  [Reference Citation Analysis (0)]
41.  Gerard R, Sendid B, Colombel JF, Poulain D, Jouault T. An immunological link between Candida albicans colonization and Crohn's disease. Crit Rev Microbiol. 2015;41:135-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
42.  Limon JJ, Tang J, Li D, Wolf AJ, Michelsen KS, Funari V, Gargus M, Nguyen C, Sharma P, Maymi VI, Iliev ID, Skalski JH, Brown J, Landers C, Borneman J, Braun J, Targan SR, McGovern DPB, Underhill DM. Malassezia Is Associated with Crohn's Disease and Exacerbates Colitis in Mouse Models. Cell Host Microbe. 2019;25:377-388.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 272]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
43.  Magnaye KM, Lynch SV. Fungus fuels mucosal wounds in Crohn's disease. Immunity. 2021;54:856-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
44.  Standaert-Vitse A, Jouault T, Vandewalle P, Mille C, Seddik M, Sendid B, Mallet JM, Colombel JF, Poulain D. Candida albicans is an immunogen for anti-Saccharomyces cerevisiae antibody markers of Crohn's disease. Gastroenterology. 2006;130:1764-1775.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 164]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
45.  Sivignon A, de Vallée A, Barnich N, Denizot J, Darcha C, Pignède G, Vandekerckove P, Darfeuille-Michaud A. Saccharomyces cerevisiae CNCM I-3856 prevents colitis induced by AIEC bacteria in the transgenic mouse model mimicking Crohn's disease. Inflamm Bowel Dis. 2015;21:276-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 61]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
46.  Mitsuyama K, Niwa M, Takedatsu H, Yamasaki H, Kuwaki K, Yoshioka S, Yamauchi R, Fukunaga S, Torimura T. Antibody markers in the diagnosis of inflammatory bowel disease. World J Gastroenterol. 2016;22:1304-1310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 86]  [Cited by in F6Publishing: 89]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
47.  Seelbinder B, Chen J, Brunke S, Vazquez-Uribe R, Santhaman R, Meyer AC, de Oliveira Lino FS, Chan KF, Loos D, Imamovic L, Tsang CC, Lam RP, Sridhar S, Kang K, Hube B, Woo PC, Sommer MOA, Panagiotou G. Antibiotics create a shift from mutualism to competition in human gut communities with a longer-lasting impact on fungi than bacteria. Microbiome. 2020;8:133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 79]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
48.  Hoarau G, Mukherjee PK, Gower-Rousseau C, Hager C, Chandra J, Retuerto MA, Neut C, Vermeire S, Clemente J, Colombel JF, Fujioka H, Poulain D, Sendid B, Ghannoum MA. Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn's Disease. mBio. 2016;7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 302]  [Article Influence: 33.6]  [Reference Citation Analysis (0)]
49.  Qv L, Mao S, Li Y, Zhang J, Li L. Roles of Gut Bacteriophages in the Pathogenesis and Treatment of Inflammatory Bowel Disease. Front Cell Infect Microbiol. 2021;11:755650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
50.  Lin DM, Lin HC. A theoretical model of temperate phages as mediators of gut microbiome dysbiosis. F1000Res. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
51.  Eriksson F, Tsagozis P, Lundberg K, Parsa R, Mangsbo SM, Persson MA, Harris RA, Pisa P. Tumor-specific bacteriophages induce tumor destruction through activation of tumor-associated macrophages. J Immunol. 2009;182:3105-3111.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 86]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
52.  Galtier M, De Sordi L, Sivignon A, de Vallée A, Maura D, Neut C, Rahmouni O, Wannerberger K, Darfeuille-Michaud A, Desreumaux P, Barnich N, Debarbieux L. Bacteriophages Targeting Adherent Invasive Escherichia coli Strains as a Promising New Treatment for Crohn's Disease. J Crohns Colitis. 2017;11:840-847.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 70]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
53.  Mottawea W, Chiang CK, Mühlbauer M, Starr AE, Butcher J, Abujamel T, Deeke SA, Brandel A, Zhou H, Shokralla S, Hajibabaei M, Singleton R, Benchimol EI, Jobin C, Mack DR, Figeys D, Stintzi A. Altered intestinal microbiota-host mitochondria crosstalk in new onset Crohn's disease. Nat Commun. 2016;7:13419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 274]  [Article Influence: 30.4]  [Reference Citation Analysis (0)]
54.  Ruaud A, Esquivel-Elizondo S, de la Cuesta-Zuluaga J, Waters JL, Angenent LT, Youngblut ND, Ley RE. Syntrophy via Interspecies H(2) Transfer between Christensenella and Methanobrevibacter Underlies Their Global Cooccurrence in the Human Gut. mBio. 2020;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 70]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
55.  Kumpitsch C, Fischmeister FPS, Mahnert A, Lackner S, Wilding M, Sturm C, Springer A, Madl T, Holasek S, Högenauer C, Berg IA, Schoepf V, Moissl-Eichinger C. Reduced B12 uptake and increased gastrointestinal formate are associated with archaeome-mediated breath methane emission in humans. Microbiome. 2021;9:193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 33]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
56.  Tjellström B, Högberg L, Stenhammar L, Fälth-Magnusson K, Magnusson KE, Norin E, Sundqvist T, Midtvedt T. Faecal short-chain fatty acid pattern in childhood coeliac disease is normalised after more than one year's gluten-free diet. Microb Ecol Health Dis. 2013;24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Cisek AA, Szymańska E, Aleksandrzak-Piekarczyk T, Cukrowska B. The Role of Methanogenic Archaea in Inflammatory Bowel Disease-A Review. J Pers Med. 2024;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
58.  Cao Z, Fan D, Sun Y, Huang Z, Li Y, Su R, Zhang F, Li Q, Yang H, Zhang F, Miao Y, Lan P, Wu X, Zuo T. The gut ileal mucosal virome is disturbed in patients with Crohn's disease and exacerbates intestinal inflammation in mice. Nat Commun. 2024;15:1638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
59.  Chevallereau A, Pons BJ, van Houte S, Westra ER. Interactions between bacterial and phage communities in natural environments. Nat Rev Microbiol. 2022;20:49-62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 207]  [Article Influence: 51.8]  [Reference Citation Analysis (0)]
60.  Regueiro M, Velayos F, Greer JB, Bougatsos C, Chou R, Sultan S, Singh S. American Gastroenterological Association Institute Technical Review on the Management of Crohn's Disease After Surgical Resection. Gastroenterology. 2017;152:277-295.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 99]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
61.  Holubar SD, Gunter RL, Click BH, Achkar JP, Lightner AL, Lipman JM, Hull TL, Regueiro M, Rieder F, Steele SR. Mesenteric Excision and Exclusion for Ileocolic Crohn's Disease: Feasibility and Safety of an Innovative, Combined Surgical Approach With Extended Mesenteric Excision and Kono-S Anastomosis. Dis Colon Rectum. 2022;65:e5-e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
62.  Luglio G, Rispo A, Imperatore N, Giglio MC, Amendola A, Tropeano FP, Peltrini R, Castiglione F, De Palma GD, Bucci L. Surgical Prevention of Anastomotic Recurrence by Excluding Mesentery in Crohn's Disease: The SuPREMe-CD Study - A Randomized Clinical Trial. Ann Surg. 2020;272:210-217.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 110]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
63.  Ford AC, Kane SV, Khan KJ, Achkar JP, Talley NJ, Marshall JK, Moayyedi P. Efficacy of 5-aminosalicylates in Crohn's disease: systematic review and meta-analysis. Am J Gastroenterol. 2011;106:617-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 142]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
64.  Gordon H, Minozzi S, Kopylov U, Verstockt B, Chaparro M, Buskens C, Warusavitarne J, Agrawal M, Allocca M, Atreya R, Battat R, Bettenworth D, Bislenghi G, Brown SR, Burisch J, Casanova MJ, Czuber-Dochan W, de Groof J, El-Hussuna A, Ellul P, Fidalgo C, Fiorino G, Gisbert JP, Sabino JG, Hanzel J, Holubar S, Iacucci M, Iqbal N, Kapizioni C, Karmiris K, Kobayashi T, Kotze PG, Luglio G, Maaser C, Moran G, Noor N, Papamichael K, Peros G, Reenaers C, Sica G, Sigall-Boneh R, Vavricka SR, Yanai H, Myrelid P, Adamina M, Raine T. ECCO Guidelines on Therapeutics in Crohn's Disease: Medical Treatment. J Crohns Colitis. 2024;18:1531-1555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Reference Citation Analysis (0)]
65.  Sandborn WJ, Feagan BG, Rutgeerts P, Hanauer S, Colombel JF, Sands BE, Lukas M, Fedorak RN, Lee S, Bressler B, Fox I, Rosario M, Sankoh S, Xu J, Stephens K, Milch C, Parikh A; GEMINI 2 Study Group. Vedolizumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2013;369:711-721.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1416]  [Cited by in F6Publishing: 1502]  [Article Influence: 125.2]  [Reference Citation Analysis (0)]
66.  Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, Blank MA, Johanns J, Gao LL, Miao Y, Adedokun OJ, Sands BE, Hanauer SB, Vermeire S, Targan S, Ghosh S, de Villiers WJ, Colombel JF, Tulassay Z, Seidler U, Salzberg BA, Desreumaux P, Lee SD, Loftus EV Jr, Dieleman LA, Katz S, Rutgeerts P; UNITI–IM-UNITI Study Group. Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med. 2016;375:1946-1960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1053]  [Cited by in F6Publishing: 1246]  [Article Influence: 138.4]  [Reference Citation Analysis (0)]
67.  Sands BE, Anderson FH, Bernstein CN, Chey WY, Feagan BG, Fedorak RN, Kamm MA, Korzenik JR, Lashner BA, Onken JE, Rachmilewitz D, Rutgeerts P, Wild G, Wolf DC, Marsters PA, Travers SB, Blank MA, van Deventer SJ. Infliximab maintenance therapy for fistulizing Crohn's disease. N Engl J Med. 2004;350:876-885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1581]  [Cited by in F6Publishing: 1519]  [Article Influence: 72.3]  [Reference Citation Analysis (0)]
68.  Aluzaite K, Braund R, Seeley L, Amiesimaka OI, Schultz M. Adherence to Inflammatory Bowel Disease Medications in Southern New Zealand. Crohns Colitis 360. 2021;3:otab056.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
69.  Xu F, Tang J, Zhu Z, Chen Y, Hu W, Lu S, Zhou Y, Lin D. Medication Adherence and Its Influencing Factors Among Inflammatory Bowel Disease Patients in China. Int J Gen Med. 2022;15:4141-4149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
70.  Paes AH, Bakker A, Soe-Agnie CJ. Impact of dosage frequency on patient compliance. Diabetes Care. 1997;20:1512-1517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 262]  [Cited by in F6Publishing: 264]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
71.  Bermejo F, López-San Román A, Algaba A, Guerra I, Valer P, García-Garzón S, Piqueras B, Villa C, Bermejo A, Rodríguez-Agulló JL. Factors that modify therapy adherence in patients with inflammatory bowel disease. J Crohns Colitis. 2010;4:422-426.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
72.  Cross RK, Lapshin O, Finkelstein J. Patient subjective assessment of drug side effects in inflammatory bowel disease. J Clin Gastroenterol. 2008;42:244-251.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 37]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
73.  Sanchis-Artero L, Martínez-Blanch JF, Manresa-Vera S, Cortés-Castell E, Valls-Gandia M, Iborra M, Paredes-Arquiola JM, Boscá-Watts M, Huguet JM, Gil-Borrás R, Rodríguez-Morales J, Cortés-Rizo X. Evaluation of changes in intestinal microbiota in Crohn's disease patients after anti-TNF alpha treatment. Sci Rep. 2021;11:10016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
74.  Rashed R, Valcheva R, Dieleman LA. Manipulation of Gut Microbiota as a Key Target for Crohn's Disease. Front Med (Lausanne). 2022;9:887044.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 11]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
75.  Limketkai BN, Iheozor-Ejiofor Z, Gjuladin-Hellon T, Parian A, Matarese LE, Bracewell K, MacDonald JK, Gordon M, Mullin GE. Dietary interventions for induction and maintenance of remission in inflammatory bowel disease. Cochrane Database Syst Rev. 2019;2:CD012839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
76.  Ashton JJ, Gavin J, Beattie RM. Exclusive enteral nutrition in Crohn's disease: Evidence and practicalities. Clin Nutr. 2019;38:80-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 88]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
77.  Heerasing N, Thompson B, Hendy P, Heap GA, Walker G, Bethune R, Mansfield S, Calvert C, Kennedy NA, Ahmad T, Goodhand JR. Exclusive enteral nutrition provides an effective bridge to safer interval elective surgery for adults with Crohn's disease. Aliment Pharmacol Ther. 2017;45:660-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
78.  Zhu W, Guo Z, Zuo L, Gong J, Li Y, Gu L, Cao L, Li N, Li J. CONSORT: Different End-Points of Preoperative Nutrition and Outcome of Bowel Resection of Crohn Disease: A Randomized Clinical Trial. Medicine (Baltimore). 2015;94:e1175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
79.  Swaminath A, Feathers A, Ananthakrishnan AN, Falzon L, Li Ferry S. Systematic review with meta-analysis: enteral nutrition therapy for the induction of remission in paediatric Crohn's disease. Aliment Pharmacol Ther. 2017;46:645-656.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 118]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
80.  Day A, Wood J, Melton S, Bryant RV. Exclusive enteral nutrition: An optimal care pathway for use in adult patients with active Crohn's disease. JGH Open. 2020;4:260-266.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
81.  Matuszczyk M, Kierkus J. Nutritional Therapy in Pediatric Crohn's Disease-Are We Going to Change the Guidelines? J Clin Med. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
82.  Malesza IJ, Malesza M, Walkowiak J, Mussin N, Walkowiak D, Aringazina R, Bartkowiak-Wieczorek J, Mądry E. High-Fat, Western-Style Diet, Systemic Inflammation, and Gut Microbiota: A Narrative Review. Cells. 2021;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 288]  [Article Influence: 72.0]  [Reference Citation Analysis (0)]
83.  Oka A, Sartor RB. Microbial-Based and Microbial-Targeted Therapies for Inflammatory Bowel Diseases. Dig Dis Sci. 2020;65:757-788.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 94]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
84.  Dahl WJ, Rivero Mendoza D, Lambert JM. Diet, nutrients and the microbiome. Prog Mol Biol Transl Sci. 2020;171:237-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
85.  Comeche JM, Gutierrez-Hervás A, Tuells J, Altavilla C, Caballero P. Predefined Diets in Patients with Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. Nutrients. 2020;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
86.  Levine A, Wine E, Assa A, Sigall Boneh R, Shaoul R, Kori M, Cohen S, Peleg S, Shamaly H, On A, Millman P, Abramas L, Ziv-Baran T, Grant S, Abitbol G, Dunn KA, Bielawski JP, Van Limbergen J. Crohn's Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology. 2019;157:440-450.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 372]  [Article Influence: 62.0]  [Reference Citation Analysis (0)]
87.  Sigall-Boneh R, Pfeffer-Gik T, Segal I, Zangen T, Boaz M, Levine A. Partial enteral nutrition with a Crohn's disease exclusion diet is effective for induction of remission in children and young adults with Crohn's disease. Inflamm Bowel Dis. 2014;20:1353-1360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
88.  Sigall Boneh R, Sarbagili Shabat C, Yanai H, Chermesh I, Ben Avraham S, Boaz M, Levine A. Dietary Therapy With the Crohn's Disease Exclusion Diet is a Successful Strategy for Induction of Remission in Children and Adults Failing Biological Therapy. J Crohns Colitis. 2017;11:1205-1212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 116]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
89.  Svolos V, Hansen R, Nichols B, Quince C, Ijaz UZ, Papadopoulou RT, Edwards CA, Watson D, Alghamdi A, Brejnrod A, Ansalone C, Duncan H, Gervais L, Tayler R, Salmond J, Bolognini D, Klopfleisch R, Gaya DR, Milling S, Russell RK, Gerasimidis K. Treatment of Active Crohn's Disease With an Ordinary Food-based Diet That Replicates Exclusive Enteral Nutrition. Gastroenterology. 2019;156:1354-1367.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 163]  [Cited by in F6Publishing: 204]  [Article Influence: 34.0]  [Reference Citation Analysis (0)]
90.  Szczubełek M, Pomorska K, Korólczyk-Kowalczyk M, Lewandowski K, Kaniewska M, Rydzewska G. Effectiveness of Crohn's Disease Exclusion Diet for Induction of Remission in Crohn's Disease Adult Patients. Nutrients. 2021;13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
91.  Scarallo L, Banci E, Pierattini V, Lionetti P. Crohn's disease exclusion diet in children with Crohn's disease: a case series. Curr Med Res Opin. 2021;37:1115-1120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
92.  Yanai H, Levine A, Hirsch A, Boneh RS, Kopylov U, Eran HB, Cohen NA, Ron Y, Goren I, Leibovitzh H, Wardi J, Zittan E, Ziv-Baran T, Abramas L, Fliss-Isakov N, Raykhel B, Gik TP, Dotan I, Maharshak N. The Crohn's disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn's disease (CDED-AD): an open-label, pilot, randomised trial. Lancet Gastroenterol Hepatol. 2022;7:49-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 94]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
93.  van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, Kuijper EJ, Bartelsman JF, Tijssen JG, Speelman P, Dijkgraaf MG, Keller JJ. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368:407-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2582]  [Cited by in F6Publishing: 2580]  [Article Influence: 215.0]  [Reference Citation Analysis (0)]
94.  Brandt LJ, Aroniadis OC. An overview of fecal microbiota transplantation: techniques, indications, and outcomes. Gastrointest Endosc. 2013;78:240-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 179]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
95.  Yoon SS, Brandt LJ. Treatment of refractory/recurrent C. difficile-associated disease by donated stool transplanted via colonoscopy: a case series of 12 patients. J Clin Gastroenterol. 2010;44:562-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 137]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
96.  Kassam Z, Lee CH, Yuan Y, Hunt RH. Fecal microbiota transplantation for Clostridium difficile infection: systematic review and meta-analysis. Am J Gastroenterol. 2013;108:500-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 617]  [Cited by in F6Publishing: 633]  [Article Influence: 52.8]  [Reference Citation Analysis (0)]
97.  Cheng F, Huang Z, Wei W, Li Z. Fecal microbiota transplantation for Crohn's disease: a systematic review and meta-analysis. Tech Coloproctol. 2021;25:495-504.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
98.  Gutin L, Piceno Y, Fadrosh D, Lynch K, Zydek M, Kassam Z, LaMere B, Terdiman J, Ma A, Somsouk M, Lynch S, El-Nachef N. Fecal microbiota transplant for Crohn disease: A study evaluating safety, efficacy, and microbiome profile. United European Gastroenterol J. 2019;7:807-814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 43]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
99.  Kao D, Hotte N, Gillevet P, Madsen K. Fecal microbiota transplantation inducing remission in Crohn's colitis and the associated changes in fecal microbial profile. J Clin Gastroenterol. 2014;48:625-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 59]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
100.  Sokol H, Landman C, Seksik P, Berard L, Montil M, Nion-Larmurier I, Bourrier A, Le Gall G, Lalande V, De Rougemont A, Kirchgesner J, Daguenel A, Cachanado M, Rousseau A, Drouet É, Rosenzwajg M, Hagege H, Dray X, Klatzman D, Marteau P; Saint-Antoine IBD Network, Beaugerie L, Simon T. Fecal microbiota transplantation to maintain remission in Crohn's disease: a pilot randomized controlled study. Microbiome. 2020;8:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 199]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]