Next Article in Journal
Iron Overload, Microbleeding and the Role of Bilirubin in Alzheimer’s Disease Brain: Revisiting the Vascular Hypothesis
Next Article in Special Issue
Circadian Rhythm Dysregulation in Inflammatory Bowel Disease: Mechanisms and Chronotherapeutic Approaches
Previous Article in Journal
The Use of Neurons Derived from Pluripotent Stem Cells to Study Nerve–Cancer Cell Interactions
Previous Article in Special Issue
A Novel Microbial Dysbiosis Index and Intestinal Microbiota-Associated Markers as Tools of Precision Medicine in Inflammatory Bowel Disease Paediatric Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools

by
Marco Murgiano
1,*,
Bianca Bartocci
1,
Pierluigi Puca
1,2,
Federica di Vincenzo
1,
Angelo Del Gaudio
1,
Alfredo Papa
1,2,
Giovanni Cammarota
1,2,
Antonio Gasbarrini
1,2,
Franco Scaldaferri
1,2 and
Loris Riccardo Lopetuso
1,3,*
1
Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
2
Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
3
Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(7), 3059; https://doi.org/10.3390/ijms26073059
Submission received: 12 January 2025 / Revised: 18 March 2025 / Accepted: 26 March 2025 / Published: 27 March 2025
(This article belongs to the Special Issue Molecular Diagnostics and Treatment of Inflammatory Bowel Disease)

Abstract

Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn’s disease. The gut microbiota’s role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies—including prebiotics, probiotics, postbiotics, and symbiotics—have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients’ quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.

1. Introduction

Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders of the gastrointestinal tract characterized by a relapsing-remitting course, primarily including Crohn’s disease (CD) and ulcerative colitis (UC) [1]. Although the exact etiology of IBD remains unknown, the most widely accepted hypothesis suggests that IBD results from an inappropriate immune response triggered by a combination of genetic, microbial, and environmental factors [2]. Despite its multifactorial nature, microbial dysfunction is recognized as one of the key contributors to IBD pathogenesis [3].
The gut microbiome is a complex ecosystem that maintains a mutualistic relationship with the host, playing a crucial role in preserving homeostasis [4]. Dysbiosis refers to any disruption in the gut microbiota composition that disturbs microbial balance, ultimately impairing gut functions and disrupting host–microbiota homeostasis [5,6]. Many studies have identified dysbiosis as a significant factor contributing to both the pathogenesis and progression of IBD [7,8].
Inflammatory bowel diseases (IBDs) arise from a dysregulated immune response to an altered microbiota in genetically predisposed individuals, with environmental factors acting as triggers [9]. Understanding the dynamic interplay between host genetics and the gut microbiota is challenging due to the large number of genomic markers and the diversity of microbial taxa. Genetic polymorphisms can affect immune responses or gut barrier function, leading to impaired bacterial clearance and establishing a link between genetic variants and gut dysbiosis. Many studies have shown similarities in the gut microbiota of twins, regardless of their concordance or discordance for IBD, highlighting the genetic influence on microbiota composition [10,11].
Currently, around 240 risk loci associated with IBD have been identified, with some of the most well-documented being NOD2, ATG16L1, IGRM, and CARD9 [12,13,14]. For instance, associations between NOD2 and ATG16L1 genotypes and dysbiosis have been observed in Crohn’s disease patients [15]. Specific variants of the NOD2 gene have been linked to increased abundance of the Enterobacteriaceae family in IBD patients [16]. Moreover, genetic variations can influence gut-derived metabolites, such as alterations in the MYRF gene, which are associated with changes in short-chain fatty acids (SCFAs)—key molecules for maintaining gut health [17].
In recent years, increasing attention has been given to microbial genome sequencing and host–microbe interactions to advance targeted IBD therapies. Integrative multiomics studies have characterized IBD patients by analyzing host genetics, microbiota, and other risk factors. For instance, a recent case-control study documented gut microbiota alterations (e.g., decreased Roseburia abundance) in healthy individuals with a high genetic risk for IBD [18].
Beyond genetic factors, environmental exposures such as diet, antibiotic use, infections, and smoking can disrupt the microbial balance, further implicating the microbiota in IBD pathogenesis [19,20]. Smoking influences both microbiota composition and immune responses [21] and can impair intestinal barrier function with chronic exposure [22]. The Western diet, rich in saturated fats and refined carbohydrates but low in fiber, is considered a trigger for IBD due to its impact on microbiota diversity, mucosal integrity, and immunity [23]. This diet promotes the proliferation of mucin-degrading bacteria like Bacteroides thetaiotaomicron and Akkermansia muciniphila, as well as potentially pathogenic species like Proteobacteria and adherent-invasive E. coli, ultimately leading to barrier dysfunction [24,25,26]. In contrast, a fiber-rich plant-based diet promotes microbial diversity and increases the production of SCFAs, which help maintain intestinal health [27].
Early-life antibiotic exposure can also shape gut microbiota composition, with different effects depending on the antibiotic type and duration of use. A recent meta-analysis linked exposure to cephalosporins, quinolones, tetracyclines, and metronidazole with an increased risk of developing IBD, particularly with earlier antibiotic exposure [28,29].
The gut–brain axis adds another layer of complexity to IBD. Stress, depression, and anxiety can alter microbiota composition and gut-derived metabolites, exacerbating inflammation, while gut microbiota modulation can influence stress responses [30,31]. Fecal microbiome richness and diversity are lower in IBD patients with psychological disorders than in those without, with a reduction in beneficial bacteria (e.g., Lachnospiraceae, Fusobacteriaceae, Ruminococcaceae, and Veillonellaceae) and an increase in opportunistic pathogens like Enterococcus [32,33,34]. Notably, reduced abundance of Roseburia, a butyrate-producing bacterium, has been linked to depression in Crohn’s disease [33].
Together, genetic and environmental factors influence not only disease pathogenesis but also therapeutic responses. It remains unclear whether gut microbiota alterations are a cause or consequence of intestinal inflammation, but unraveling this relationship could pave the way for innovative therapeutic strategies [35,36].
Despite advancements in understanding IBD, current therapies—including 5-aminosalicylates (5-ASA), corticosteroids, biologics, and small molecules—have limitations, such as adverse effects, drug resistance, and variable responsiveness. This has driven interest in therapies that modulate the gut microbiota to influence disease outcomes. Probiotics, for example, have demonstrated safety and efficacy in preventing disease flares and improving quality of life, with minimal contraindications in IBD patients [37,38].
Fecal microbiota transplantation (FMT) was the first microbiota-targeted therapy, showing promise in treating not only IBD but also conditions like chronic liver disorders and extraintestinal autoimmune diseases [39]. Meanwhile, preclinical studies on microbial biotherapeutics—including next-generation probiotics, bacterial consortia, bacteriophage-based therapies, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics—have yielded encouraging, albeit preliminary, results.

2. Changes in Gut Microbiota Are Associated with IBD

Gut microbiota is a dynamic community composed of a vast array of commensal bacterial species [40]. More than 90% of the human gut microbiota belongs to four main phyla: Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria [41,42]. The relative abundance of these phyla, which reflects microbial diversity, can vary significantly between individuals [43]. Two essential concepts in understanding intestinal health are eubiosis and dysbiosis, representing opposite states of microbial balance with distinct effects on human health. Eubiosis refers to a state of microbial equilibrium, characterized by a predominance of beneficial bacteria that contribute to overall intestinal well-being [44].
In a healthy gut, the microbiota and host maintain a symbiotic relationship that strengthens immune system function, aids digestion and nutrient absorption, and reduces the risk of developing inflammatory bowel disease (IBD) [45]. Conversely, dysbiosis refers to an imbalance in the composition and function of the gut microbiota, disrupting the microbe–host relationship and impairing critical physiological functions [5].
Many studies have identified dysbiosis as a central factor in IBD pathogenesis [7,8]. Several mechanisms explain this connection. A hallmark of dysbiosis in IBD patients is the depletion of butyrate-producing bacteria, alongside an increase in sulfate-reducing bacteria. The loss of short-chain fatty acid (SCFA)-producing species, such as Faecalibacterium prausnitzii, weakens the intestinal epithelial barrier, increasing gut permeability and promoting bacterial translocation across the intestinal epithelium and into the lamina propria [46]. This also impairs the differentiation of regulatory T cells (Tregs), which are essential for immune tolerance [47].
Additionally, an overgrowth of lipopolysaccharide (LPS)-producing bacteria can trigger toll-like receptor 4 (TLR4) signaling, activating NF-κB and perpetuating chronic inflammation [48]. The expansion of sulfate-reducing bacteria exacerbates the condition by converting sulfate to hydrogen sulfide, a compound that inhibits butyrate utilization, impairs immune responses, and facilitates bacterial persistence [49].
Oxidative stress is another driver of dysbiosis and IBD. The increased production of reactive oxygen species (ROS) disrupts the gut’s anaerobic environment, fostering the growth of facultative anaerobes and furthering microbial imbalance [50].
Bile acid metabolism is also intricately linked to dysbiosis and IBD. Alterations in the gut microbiota can disturb bile acid homeostasis, reducing secondary bile acid production and impairing farnesoid X receptor (FXR) signaling, a key regulator of bile acid metabolism [51]. Similarly, disruptions in tryptophan metabolism limit the generation of microbial-derived aryl hydrocarbon receptor (AhR) ligands, compromising intestinal barrier integrity and promoting immune dysregulation [52].
Together, these mechanisms position dysbiosis as an active driver of IBD, emphasizing the potential of microbiome-targeted therapeutic strategies. Interventions such as dietary modifications, metabolite-based therapies, and fecal microbiota transplantation offer promising avenues for restoring microbial balance and mitigating IBD severity.
The composition of the gut microbiota differs significantly between IBD patients and healthy controls [53]. Individuals with IBD exhibit distinct alterations in microbial abundance, alongside reduced α- and β-diversity of intestinal flora [54]. At the phylum level, IBD patients typically show a decrease in Firmicutes—commensal bacteria essential for maintaining the intestinal barrier and mucosal immunity [55,56]—while displaying an increased abundance of Proteobacteria and Bacteroidetes, both associated with pro-inflammatory properties [55,57].
At the family level, a reduction in beneficial, anti-inflammatory bacteria, such as Lachnospiraceae and Ruminococcaceae, which play a crucial role in short-chain fatty acid (SCFA) production, has been observed in IBD patients compared to healthy controls [58,59]. At the genus level, lower levels of Roseburia, Akkermansia, and Faecalibacterium have been documented [60,61,62]. Additionally, IBD patients show decreased levels of beneficial bacteria like Bacteroides, Lactobacillus, and Eubacterium [63,64], alongside an increase in potentially harmful species such as Enterobacteriaceae, Pasteurellaceae, Veillonellaceae, and Fusobacteriaceae [3,65,66,67,68].
These microbial changes are not only associated with the presence of IBD but also correlate with disease activity. A meta-analysis of 16 studies, including 1669 subjects, demonstrated a significantly lower abundance of F. prausnitzii in IBD patients compared to healthy controls. Furthermore, F. prausnitzii levels were lower in patients with active IBD (both UC and CD) compared to those in remission, suggesting a negative correlation between F. prausnitzii abundance and disease activity [69].
Interestingly, shifts in microbiota composition have also been observed following treatment. For example, anti-TNF-α therapy has been associated with a significant increase in F. prausnitzii abundance [70]. Higher fecal levels of F. prausnitzii have also been linked to lower Crohn’s Disease Activity Index (CDAI) scores, C-reactive protein (CRP) levels, and erythrocyte sedimentation rates [71]. Given the extreme sensitivity of F. prausnitzii to changes in the intestinal environment, its detection in feces or mucosa may serve as a valuable biomarker for diagnosing IBD and monitoring disease prognosis [72].
The rise of integrated multiomics approaches now enables a more comprehensive analysis of the relationship between gut dysbiosis and IBD, characterizing bacterial, fungal, and metabolic fingerprints associated with the disease [73]. Considering the well-established link between gut microbiota alterations and IBD, ongoing research is increasingly focused on developing innovative microbiome-targeted therapeutic strategies.

3. The Old Paradigm of Gut Microbiota Modulation in IBD

Despite the wide range of therapeutic options available for UC and CD, including anti-inflammatory and immunosuppressive drugs, treatment efficacy remains limited, recurrence rates are high, and symptom control is often inadequate. As a result, alternative therapeutic strategies have been developed to improve IBD management.

3.1. Antibiotics

Several antibiotics, including metronidazole, ciprofloxacin (alone or in combination), rifaximin, and antitubercular therapy, have been evaluated in multiple trials for the treatment of IBD [74]. A meta-analysis of 10 randomized trials reported improved clinical outcomes in IBD patients following antibiotic therapy (ciprofloxacin, metronidazole, and clarithromycin, either individually or combined) [75]. In CD, antibiotics serve various purposes: metronidazole has been used in colonic Crohn’s disease [76], while ciprofloxacin has been compared to mesalamine in active luminal disease [76]. Complications like abscesses and fistulas often require drainage and antibiotic therapy (ciprofloxacin, metronidazole, or both) [77], and antibiotics also play a role in preventing post-operative recurrence [78]. Conversely, evidence supporting antibiotic use in UC is more limited [79]. Antibiotics, particularly metronidazole and ciprofloxacin, remain the cornerstone of pouchitis treatment [80]. Other indications include small intestinal bacterial overgrowth, septic complications, or perianal disease.
Despite their potential benefits, the use of antibiotics in IBD remains controversial due to adverse effects, the risk of dysbiosis, increased susceptibility to C. difficile infections, and the emergence of antibiotic-resistant strains [57,81]. For example, rifaximin, long considered a safe option, has recently been linked to resistance development. Rifaximin prophylaxis has been associated with daptomycin resistance—a last-resort therapy for multidrug-resistant pathogens like vancomycin-resistant Enterococcus faecium (VREfm)—especially in patients with liver cirrhosis [82]. These findings emphasize the importance of judicious antibiotic use, even with ‘low-risk’ agents, as they may contribute to antimicrobial cross-resistance. Antibiotic resistance is a global crisis, driven by overuse in both the general population and healthcare settings. In IBD patients, especially in scenarios like bowel obstruction, the indication for antibiotics remains unclear due to the elevated risk of C. difficile infection.

3.2. Probiotics

Probiotics are live microorganisms that exhibit antibacterial properties, counteracting pathogenic bacteria, modulating gut microbiota composition, regulating the immune system, enhancing anti-inflammatory responses, and strengthening the intestinal barrier [83,84,85]. Several studies have shown that administering specific probiotic strains, such as E. coli Nissle 1917, Bifidobacteria, and Lactobacillus, can regulate and modulate immune responses. Probiotics influence immune activity by modulating the functions of macrophages, dendritic cells, and T lymphocytes [86]. Probiotics have been studied not only for IBD but also for other conditions, such as irritable bowel syndrome and diverticulitis. For instance, various Lactobacilli strains (e.g., Limosilactobacillus reuteri ATCC PTA 4659) have shown anti-inflammatory effects, reducing serum and fecal inflammatory markers in acute uncomplicated diverticulitis [87]. The administration of Clostridium butyricum has been associated with improved symptoms, including increased stool consistency and reduced urinary urgency in irritable bowel syndrome [88]. The most significant benefits of probiotics are observed in UC patients (both in active disease and remission), while data regarding their efficacy in CD remain inconsistent [89,90]. Recent studies on the effectiveness of probiotics are summarized in Table 1.
Although probiotics are widely used to prevent and treat various gastrointestinal (GI) diseases, several factors must be carefully considered: the mechanisms of action of each strain, the optimal dosage, the timing and duration of treatment, the selection of individual strains or their combinations, as well as safety and stability. Therefore, large-scale randomized controlled trials are essential to confirm the efficacy of probiotics and establish their role in clinical practice.

3.3. Prebiotics

Prebiotics are nondigestible (oligo)saccharides that confer physiological benefits to the host by selectively promoting the growth or activity of a limited number of commensal bacteria [102]. They must undergo selective fermentation by specific beneficial bacteria in the intestine. The most commonly used prebiotics are β-fructans, oligosaccharides, and inulin [103,104].
The effects of prebiotics have been primarily investigated in experimental colitis models. Their role in maintaining intestinal barrier integrity by fostering the growth of bacteria that upregulate defense mechanisms has been documented [105]. Prebiotics can also influence the production of pro- and anti-inflammatory cytokines, mitigating the inflammatory response [106]. Lactulose and inulin have demonstrated promising anti-inflammatory properties [107]. In a dextran sulfate sodium (DSS)-induced mouse model, administering fructo-oligosaccharide (400 mg/kg) for 37 days alleviated DSS-induced colitis symptoms by modulating intestinal dysbiosis and tryptophan metabolism [108]. Fructo-oligosaccharide also exhibits anti-inflammatory effects in T lymphocyte-dependent colitis models by reducing pro-inflammatory cytokine levels [109].
The effects of prebiotics in human studies remain under investigation. In a small open-label trial involving ten patients with ileocolic Crohn’s disease, supplementation with 15 g/day of oligofructose and inulin for 3 weeks reduced clinical disease activity, accompanied by an increase in mucosal Bifidobacteria [110]. This finding was corroborated by subsequent studies showing that administering 12 g/day of oligofructose-enriched inulin reduced quantitative fecal calprotectin levels [111]. Recent research has highlighted the potential of prebiotics in reducing clinical and endoscopic disease activity. In a single-center, randomized, double-blind, placebo-controlled trial, 9.8 g/day of trisaccharide fructooligosaccharides-1 ketose led to higher clinical remission and response rates (with no significant difference in endoscopic outcomes) and a significant reduction in α-diversity in patients with mild to moderate UC [112]. An improvement in endoscopic activity was observed in a dual-arm exploratory study involving the administration of either 15 g/day or 7.5 g/day of β-fructans inulin plus fructooligosaccharides in patients with mild to moderate active UC [113].
Despite these promising results, further human studies are necessary to confirm the true effectiveness of prebiotics in IBD management.

3.4. Postbiotics

Postbiotics are inactivated microbial cells or their components, with or without metabolites, that provide health benefits. They play a crucial role in the development of the intestinal immune system and the maintenance of the intestinal barrier, while also indirectly shaping microbiota composition [114].
Interest in postbiotics for IBD treatment is increasing, with several studies highlighting their potential benefits for patients. For instance, microencapsulated sodium butyrate, known for its anti-inflammatory and regenerative properties, has been shown to induce microbiota changes in IBD patients. In UC patients, it promoted the growth of beneficial short-chain fatty acid-producing bacteria, such as Lachnospiraceae spp., while in CD patients, it increased the abundance of butyrogenic colonic bacteria, including Butyricicoccus [115]. Additionally, the administration of SER-287 (Firmicutes spores) after vancomycin preconditioning demonstrated efficacy in achieving clinical remission in patients with mild to moderately active UC [116].
Regulatory guidelines require a precise limit on the number of viable microorganisms remaining after postbiotic preparation. Further exploration of postbiotics may open new frontiers in IBD treatment, offering a dual benefit: maintaining homeostasis and modulating the intestinal barrier to restore eubiosis.

3.5. Symbiotics

Beneficial effects in IBD can also arise from symbiotics, a combination of probiotics and prebiotics. The most well-known and widely used combinations include Bifidobacteria with fructooligosaccharides, Bifidobacteria and Lactobacilli with fructooligosaccharides or inulin, and Lactobacillus GG with inulin. Several trials have demonstrated the efficacy of symbiotic therapy in IBD [117,118]. For example, supplementation with symbiotics (Bifidobacterium longum combined with inulin-oligofructose or psyllium) in UC patients showed greater clinical improvement compared to probiotics or prebiotics alone [119]. Additionally, the use of B. longum, inulin, and oligofructose in UC patients reduced endoscopic inflammation and serum markers of inflammation, including TNF-α and IL-1β [119]. Similarly, the combination of L. rhamnosus 1.0320 with inulin alleviated DSS-induced colitis by enhancing gut flora diversity and increasing the abundance of beneficial bacteria [120].
More recently, a symbiotic combination of Lactobacillus plantarum SC-5 and olive oil extract tyrosol in a DSS-induced mice model led to colitis remission and reduced colonic inflammation by inhibiting mitogen-activated protein kinase (MAPK) and NF-κB pathways. The combination also preserved gut barrier integrity by regulating the expression of occludin, claudin-3, and zona occludens-1 [121].
Although symbiotics have demonstrated greater effectiveness than probiotics or prebiotics alone, their precise clinical impact remains unclear due to variability in benefits depending on the specific probiotic–prebiotic combinations. Existing studies often use heterogeneous methodologies and suffer from limitations in study design. Moreover, many findings come from preclinical models, limiting their immediate application to clinical practice. The optimal dosage and duration of treatment also remain undefined.

4. Revolutionizing Microbiota Therapeutics in IBD: The New Paradigm

More recently, building on the encouraging results from studies on probiotics, prebiotics, symbiotics, and postbiotics, research has shifted towards exploring even more innovative experimental therapeutic approaches. The modulation of gut microbiota remains the underlying rationale for these strategies. However, the goal is to develop more effective and targeted tools for clinical practice—with the ambitious aims of increasing response rates, minimizing the impact on microbial resistance, and ensuring an excellent safety profile.

4.1. Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) is emerging as a promising therapeutic tool with a wide range of potential applications. Its primary goal is to restore the natural composition of intestinal flora by transferring fecal microbiota from healthy donors [122]. Various administration methods are available, including nasogastric or nasojejunal tubes, colonoscopy, enema, and oral capsules [123,124]. Currently, the most established use of FMT is for treating antibiotic-resistant C. difficile infections [125]. A randomized controlled trial reported clinical improvement, with diarrhea resolution in 81% of patients receiving FMT, compared to only 31% in the antibiotic-treated group [126].
Many studies have evaluated FMT’s efficacy in ulcerative colitis (UC), exploring different delivery routes and infusion schedules, though results have been inconsistent: they are summarized in Table 2 [127,128,129,130,131,132,133,134,135]. A 2023 meta-analysis of 13 randomized controlled trials, including 580 UC patients (293 receiving FMT and 287 controls), showed significantly higher clinical and endoscopic remission rates at 8–12 weeks in the FMT group, suggesting its potential to induce UC remission [136]. Interestingly, clinical remission and response rates were consistent across trials, despite methodological heterogeneity.
Another meta-analysis, including 9 studies with 520 UC patients (261 receiving FMT and 259 controls), found significantly improved clinical remission [RR = 1.53; 95% CI: 1.19–1.94; p < 0.0008] and endoscopic remission [RR = 2.80; 95% CI: 1.93–4.05; p < 0.00001] in the FMT group. Administration routes included colonoscopy, enema, and nasoduodenal tubes, with follow-ups ranging from 7 to 48 weeks, though remission differences beyond 8 weeks were not statistically significant, emphasizing the need for long-term studies [137].
Long-term FMT efficacy in UC remains underexplored. In a randomized, double-blind, placebo-controlled trial, Haifer et al. followed UC patients on oral lyophilized FMT. At week 8, responders were randomized to continue or discontinue FMT for an additional 48 weeks. By week 56, all patients continuing FMT (4/4) maintained remission, while none in the discontinuation group (0/6) remained in remission [128]. Similarly, Costello et al. tracked UC patients for 12 months after an 8-week FMT induction via colonoscopy and enema, finding that 42% (5/12) maintained remission at 12 months [131]. These findings highlight FMT’s potential but underscore the need for larger, long-term trials.
FMT research in Crohn’s disease (CD) is more limited, often lacking control groups and featuring small sample sizes, leading to mixed outcomes [138]. Methodological variability—such as FMT type, delivery routes, antibiotic preconditioning, infusion frequency, and donor selection—further complicates interpretation. A meta-analysis of 12 studies (1 RCT, 7 cohort studies, 4 case reports) showed clinical remission and response rates of 62% and 79%, respectively, with follow-ups from 4 weeks to 15 months. Remission correlated with increased microbiota diversity and stability [139]. A systematic review of 11 cohort studies and 1 RCT supported these findings, showing short-term clinical remission, improved disease activity indices, and enhanced microbial diversity [140]. Notably, multiple FMT infusions yielded higher response rates than single infusions [141].
For chronic pouchitis, evidence is sparse. A meta-analysis of 8 studies (74 receiving FMT, 15 placebo) showed short-term clinical responses in 42% of patients [142]. Another analysis of 7 cohort studies and 2 RCTs (103 patients) reported a clinical response rate of 42.6% and remission in 29.8%, though delivery methods varied widely—ranging from upper GI tract routes (nasogastric tube, jejunal endoscopy) to lower GI tract options (pouchoscopy, enemas) [143].
An intriguing aspect of FMT is the potential transfer of not only bacteria but also viruses. Although bacteria constitute the dominant gut microbiota, viruses can interact with the host, influence immune responses, and affect metabolic functions [144].
Despite its promise, FMT poses challenges. Most studies have short follow-ups, so long-term safety and efficacy data are lacking. Donor variability introduces qualitative and quantitative microbiota differences, affecting outcomes [145]. The transfer of live microorganisms to immunocompromised patients carries inherent risks, though rigorous donor screening can mitigate this [146]. Donor selection is complex and costly, requiring extensive clinical and microbiological screening to minimize pathogen transmission. Additionally, standardizing fecal preparations is difficult, as microbiota composition varies with diet, lifestyle, and antibiotic history, influencing treatment effectiveness and remission rates. The logistical costs of biological material handling and storage further complicate clinical implementation [147].
Given these complexities, patient and donor selection must be meticulous. While FMT holds great potential for managing IBD and other conditions, addressing current limitations through robust, long-term research will be essential to fully unlock its clinical benefits.

4.2. Rising Star Probiotics: Faecalibacterium Prausnitzii

Recently, live biotherapeutic products have emerged as a promising new treatment. These products are defined as commensal strains isolated from healthy donors, subsequently cultured, characterized, and carefully selected based on their anti-inflammatory and biological effects. One notable example is the anti-inflammatory strain of F. prausnitzii, which constitutes 3–5% of the total bacterial population and is one of the most predominant bacterial groups in human feces [148].
It has been proposed that Faecalibacterium plays a key role in stabilizing gut microbiota [149]. This bacterium exerts anti-inflammatory effects by producing molecules that target the IL-1β-induced NF-κB pathway, thereby reducing IL-8 secretion by intestinal epithelial cells [150]. Additionally, F. prausnitzii promotes the production of anti-inflammatory cytokines while inhibiting the secretion of pro-inflammatory cytokines such as IFN-γ, TNF-α, IL-6, and IL-12 [151].
Beyond its immunomodulatory properties, F. prausnitzii generates metabolites that enhance intestinal barrier function and regulate gut permeability by increasing the expression of tight junction proteins like E-cadherin [152]. It is also considered one of the primary intestinal producers of butyrate, a short-chain fatty acid (SCFA) with potent anti-inflammatory effects [153,154]. Through these mechanisms, F. prausnitzii contributes to maintaining host homeostasis, limiting intestinal bacterial translocation, and reducing inflammation—making it a highly relevant candidate for innovative IBD therapies [155].
Notably, a reduction in Faecalibacterium abundance has been observed in IBD patients, with a significant inverse correlation between F. prausnitzii levels and disease activity in UC, further highlighting the bacterium’s protective and anti-inflammatory effects [156]. These findings have been validated in moderate and severe DNBS-induced chronic colitis mouse models. Colitis was induced via intrarectal DNBS administration, and after a 4-day recovery period (for moderate colitis) or 10 days (for severe colitis), intragastric F. prausnitzii A2-165 was administered for 7 or 10 days. When colitis was reactivated with a lower DNBS dose, F. prausnitzii treatment significantly reduced disease severity in both models [157].

4.3. Rising Star Probiotics: Akkermansia Muciniphila

Another promising probiotic is Akkermansia muciniphila (A. muciniphila), one of the most abundant species in the intestinal microbiota [158]. It plays a crucial role in maintaining intestinal health, regulating metabolism, and modulating immune responses [159]. Many studies have demonstrated its ability to reduce macrophage and CD8 lymphocyte infiltration, thereby alleviating colitis and increasing the number of anti-inflammatory T-reg cells [160,161]. Its role in metabolic diseases such as obesity, diabetes, and alcoholic liver disease is well established [162,163,164], although its function in IBD remains less clear.
Different administration methods for A. muciniphila have been explored, including viable, pasteurized forms, and through specific components such as AmEVs, Amuc_1100, and P9 [165]. Reduced levels of A. muciniphila have been observed in patients with active UC or CD [68,166,167], with a significant decrease in active UC patients compared to those with quiescent UC and healthy controls [168]. Furthermore, the lower density of A. muciniphila in active UC patients has been confirmed, suggesting that its levels may serve as a marker for disease exacerbation [169]. In animal models, oral administration of A. muciniphila in DSS-induced colitis reduced CD8+ lymphocytes and macrophages, effectively limiting inflammation [170].
Despite promising findings, the roles of A. muciniphila and F. prausnitzii in IBD are not yet fully understood. While next-generation probiotics (NGPs) offer potential therapeutic benefits, concerns remain regarding their safety, dosage, and efficacy. Safety is a critical factor in NGP development [171], as long-term exposure to probiotics may alter microbial functions or negatively impact the surrounding microbiota [172]. Additionally, disruption of the intestinal barrier could allow probiotics to enter systemic circulation, increasing the risk of invasive infections [173].
Next-generation probiotics are being designed to be more resilient, targeted, and personalized to individual microbiome compositions, enhancing their therapeutic potential. Although preclinical studies and early-phase clinical trials show encouraging results, larger and more rigorous studies are necessary to confirm the efficacy of NGPs for treating chronic diseases [174]. Regulatory approval remains a major challenge, requiring extensive research to assess long-term safety and efficacy. For this reason, ethical approval is essential for larger sample sizes and long-term evaluations [174].
The development of new probiotics requires complete genome sequencing to characterize bacterial strains and their physiological traits, including antibiotic resistance and growth dynamics [175]. However, little is known about the safety profiles of Faecalibacterium and Akkermansia.
F. prausnitzii is an obligate anaeorobe, thriving only in oxygen-free environments.
Its extreme oxygen sensitivity makes cultivation, stabilization, growth, and production of a viable oral probiotic particularly challenging [176]. Even brief exposure to air can rapidly kill the bacteria, complicating manufacturing, packaging, and delivery processes for human use [148]. Its biodiversity and physiological characteristics remain unclear, and genome analysis suggests potential resistance to multiple antibiotics [177]. For example, Machado et al. found that F. prausnitzii DSM17677 exhibited phenotypic resistance to ampicillin, gentamicin, kanamycin, and streptomycin [178], while Hu et al. showed susceptibility to vancomycin but resistance to kanamycin and gentamicin [179]. Further studies are needed to understand the mechanisms underlying antibiotic resistance in F. prausnitzii and its clinical implications.
Although A. muciniphila is also challenging to culture, more oral formulations are available, making it a more promising candidate for clinical use [180]. However, its extreme oxygen sensitivity complicates isolation and purification. Since only a single strain of A. muciniphila is widely available for research, analyzing resistance patterns across different strains is difficult. Experiments have shown that A. muciniphila is susceptible to imipenem and doxycycline but resistant to metronidazole and vancomycin [181]. Strain-specific variations in antibiotic resistance have also been observed. For example, A. muciniphila D22959 exhibited different susceptibilities to gentamicin and kanamycin [181,182]. These findings underscore the need to establish standardized protocols for accurately assessing the antibiotic sensitivity of A. muciniphila strains.
Determining the correct NGP dosage for beneficial effects in humans remains a challenge [183]. Additional research is also needed to explore the possibility of administering multiple NGPs concurrently and to investigate potential synergistic effects [184]. With rigorous clinical trials and strong regulatory support, next-generation probiotics could become a cornerstone of personalized medicine, providing targeted therapies for chronic diseases and improving overall health outcomes.

4.4. Bacterial Consortia

An innovative and promising therapeutic strategy for microbiota modulation involves the use of bacterial consortia to regulate gut microbiota commensals. A microbial consortium consists of two or more symbiotic microorganisms capable of surviving under various conditions by forming synergistic population structures such as stromatolites, microbial mats, and biofilms [185].
The concept of bacterial consortia is gaining traction for treating conditions like C. difficile infection, IBS, and IBD. Different bacterial consortia have shown anti-inflammatory properties—depending on the bacterial species involved—and have demonstrated efficacy in reducing pro-inflammatory cytokines, vascular endothelial growth factor (VEGF), TGF-β, and matrix metalloproteinase (MMP) levels [186]. In a DSS-induced colitis mouse model, a designed bile acid consortium (BAC) composed of Clostridium AP sp000509125, Bacteroides ovatus, and Eubacterium limosum showed protective effects against inflammation through the production of secondary bile acids (ursodeoxycholic acid and lithocholic acid). These secondary bile acids exert protective effects by activating TGR5, which enhances gut barrier integrity and reduces inflammation [187].
The efficacy of bacterial consortia, such as a combination of Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus acidophilus, and Bifidobacterium lactis, has been compared to both placebo and single-strain treatments in DSS-induced colitis mouse models, demonstrating superior outcomes in reducing inflammation and accelerating recovery. Additionally, bacterial consortia can reshape the structure and composition of gut microbiota, reinforcing the link between gut flora and colitis phenotype [188].
Similarly, administering GUT-103, a bacterial consortium composed of 17 strains, in DSS-induced colitis mouse models, reduces pathobionts while increasing beneficial bacteria. It also decreases inflammatory cytokines and activates interleukin-10 cells, leading to reduced inflammatory responses [189].
Recent studies on the role of bacterial consortia in animal colitis models are summarized in Table 3.
The long-term application of bacterial consortia depends on their effects on the host, which are influenced by factors such as patient age, microbial characteristics, and concomitant diseases. Although some studies have reported side effects—including nausea, diarrhea, constipation, frequent bowel movements, and, less frequently, upper respiratory tract infections, as well as musculoskeletal and mediastinal disorders—treatment with bacterial consortia is generally considered safe. Careful donor screening and the selection of specific strains could further reduce the incidence of adverse effects. Additionally, bacterial consortia may influence drug efficacy, either enhancing or diminishing their therapeutic activity [191].
Bacterial consortia have demonstrated immense potential and may represent the most promising microbiota-targeting therapy. They offer a more precise and targeted modulation of the gut microbiota compared to FMT, with the added advantage of a more standardized preparation process—similar to the production of conventional pharmaceuticals. However, several challenges remain, including issues related to efficacy, safety, microorganism compatibility, host adaptation, competition with native microbes, standardization, and donor selection. Most importantly, a precise definition of the agents composing bacterial consortia is essential for their successful application.
Research on FMT may provide valuable insights in this regard. As highlighted in a recent consensus conference, an ideal FMT trial should test ‘supervised’ FMT, where donor stool is pre-screened and selected based on specific microbiome characteristics to ensure optimal composition and safety. Such a study design would offer deeper insights into the effects of specific bacterial strains on the recipient’s microbiota, guiding the selection of bacterial consortia components [192].
Given the current lack of human studies, further preclinical and clinical research is essential to validate the potential application of bacterial consortia in the context of IBD.

4.5. Bacteriophages

Bacteriophages are ubiquitous, self-replicating viruses that infect bacteria by interacting with bacterial receptors [193]. Bacteria have evolved various defense mechanisms against bacteriophages, including clustered regularly interspaced short palindromic repeats (CRISPR), restriction endonucleases, and other mechanisms that remain unknown [194]. The use of bacteriophages in treating multidrug-resistant (MDR) pathogens has shown promising short-term results, although bacterial resistance and host immunity remain challenges [195]. The long-term efficacy of phage therapy for gastrointestinal pathogens is still unclear. Notably, phages are currently being tested against specific bacteria associated with intestinal infections, such as C. difficile and Fusobacterium nucleatum [196,197].
Understanding the functions of gut commensal bacteria and identifying the microbes involved in dysbiosis and inflammatory bowel disease (IBD) could open new avenues for phage-based therapies. According to experimental results, gut phages may influence IBD pathogenesis through three main pathways: modulating inflammation and immune responses, regulating gut microbiota, and altering gut phage diversity [198]. Recent studies have highlighted the effectiveness of orally administered combination phage therapy in suppressing pathogenic bacteria linked to disease exacerbation. For example, the gut microbiota of four geographically distinct IBD cohorts was analyzed, revealing Klebsiella pneumoniae strains as key contributors to disease severity. Transferring these strains from IBD patients to colitis-prone, germ-free mice worsened intestinal inflammation. However, a lytic five-phage combination targeting both sensitive and resistant K. pneumoniae strains through distinct mechanisms successfully suppressed bacterial growth, reduced inflammation, and alleviated disease severity in colitis-prone mice [199].
Future next-generation K. pneumoniae phage combinations could target additional strains or even the entire species, potentially enhancing the efficacy of phage therapy against intestinal auto-inflammation. However, developing reliable phage-based treatments will require not only precise bacterial identification but also a thorough understanding of phage safety profiles, bacterial resistance mechanisms, and effective phage delivery systems.
So far, no life-threatening adverse effects have been linked to phage therapy, which generally appears to be safe [200,201]. Nevertheless, several challenges must be addressed. Bacteriophages exhibit a narrow spectrum of activity, often targeting only a few strains within a single bacterial species, which limits their utility for polymicrobial infections [202,203]. The lysogenic cycle presents another challenge, as lysogenic phages can integrate into bacterial genomes, preventing lysis and interfering with other lytic phages [204].
Standardizing phage therapy preparations remains difficult, complicating the definition of dosage, administration routes, and the evaluation of clinical efficacy [205,206]. Moreover, prolonged use of a single phage can lead to the emergence of bacteriophage-resistant strains, threatening the long-term sustainability of phage therapy. Resistance mechanisms include abortive infection, adsorption inhibition, and restriction-modification systems [202,207].
Another concern is the release of endotoxins when bacteriophages lyse their bacterial hosts, potentially exacerbating infections and increasing the risk of sepsis, as shown in preclinical studies [208]. Additionally, phages themselves can act as immunogenic agents: the release of foreign proteins from phages may trigger immune responses, including allergic reactions [209].
Despite ongoing research, studies on virome composition and its variations during IBD progression remain scarce. The clinical application of phage therapy will require overcoming these challenges by refining phage combinations, establishing regulatory frameworks, improving policies, and determining appropriate treatment doses to minimize endotoxin release.

4.6. Engineered Probiotics

Engineered probiotics have emerged as a novel strategy to deliver active molecules directly into the intestine, reducing drug-related side effects and evolving into promising therapeutic tools for IBD. The most common engineering techniques involve plasmid transfection. Plasmids are small, circular DNA molecules that are physically separate from chromosomal DNA and capable of autonomous replication. Due to their ability to transfer genetic material into bacterial cells, plasmids are considered essential vehicles for inducing the expression of recombinant DNA in target probiotic strains [210].
The advantage of engineered probiotics lies in their ability to selectively express anti-inflammatory molecules with localized release, increasing target site concentrations while minimizing systemic side effects [211]. Research has primarily focused on transfecting plasmids encoding anti-inflammatory cytokines, with E. coli strains being the preferred choice due to their genetic stability and non-transferability of plasmids pMUT1 and pMUT2 [212,213]. Lactococcus lactis has also shown promising results, thanks to its fully sequenced genome and versatility [214], making it easier to manage and a viable candidate for IBD treatment [215].
In a DSS-induced colitis mouse model, the administration of genetically engineered E. coli Nissle 1917 (EcN-Sj16) reduced clinical colitis severity, increased microbiota diversity, and enhanced butyrate production [216]. Similarly, in another DSS-treated colitis model, recombinant E. coli Nissle 1917 secreting interleukin-10 successfully suppressed intestinal inflammation in UC mice and protected the intestinal mucosa from injury [217].
Few studies have explored the efficacy of engineered Lactobacillus paracasei strains in colitis models through the release of palmitoylethanolamide (PEA), a molecule with anti-inflammatory effects [218]. Its effectiveness was further confirmed in mucosal biopsies of UC patients [219]. The administration of L. paracasei strains (KBL382, 384, 385) demonstrated protective effects in DSS-induced colitis models, with L. paracasei KBL382-treated mice showing reduced levels of pro-inflammatory cytokines, including IL-4, IL-6, TNF-α, and IL-17a [220].
Engineered Bifidobacterium longum has been studied for its high acid and bile tolerance, enabling survival in the gastrointestinal tract [221]. For example, administering B. longum expressing a PEP-1-hMnSOD fusion protein in DSS-induced colitis mice reduced inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-8) and mitigated histological damage in colonic tissues [222,223].
Engineered probiotics also include outer membrane vesicles (OMVs), nanostructures (20–300 nm) derived from the outer membrane of Gram-negative bacteria [224]. OMVs are involved in various physiological processes, facilitating intracellular factor transfer and modulating immune responses. Recently, bacterial OMVs have gained attention as promising drug delivery vehicles. In murine IBD models, OMVs derived from E. coli Nissle 1917 and Bacteroides fragilis demonstrated significant reductions in intestinal inflammation [225,226].
Recent studies on engineered probiotics are summarized in Table 4.
Recent advances in synthetic biology suggest that engineered probiotics could be used to selectively detect disease biomarkers already utilized in clinical practice. For instance, in the DSS-mice model, a bacterial promoter in the probiotic strain E. coli Nissle 1917 (ECN) showed increased expression in response to calprotectin. Moreover, the engineered probiotic successfully differentiated human patients with active IBD from those in remission or without IBD, using stool samples [229].
Although numerous in vivo and in vitro studies have demonstrated the potential efficacy of engineered probiotics in various animal models of IBD, significant differences between murine and human microbiota often prevent the successful translation of preclinical findings to human studies. Consequently, the current lack of robust evidence in clinical models limits their immediate application in clinical practice [230]. Some studies indicate that engineered probiotics may not transfer modified DNA to the environment, while still being capable of releasing bioactive molecules directly to target tissues, thereby reducing systemic side effects [210].
This therapeutic approach presents important challenges, including concerns about safety, specificity, bioavailability, and efficacy, alongside ethical and regulatory considerations. As innovative biological products, engineered probiotics straddle the intersection of gene therapy and personalized medicine. Regulatory barriers persist due to the need to establish clear safety and approval standards [211]. Another critical challenge is the high cost of development, driven by advanced technologies and rigorous validation processes, making engineered probiotics more expensive than traditional alternatives [231].
Finally, as with fecal microbiota transplantation (FMT), microbiota manipulation raises ethical questions about long-term safety and the risk of potential off-target effects that may compromise patient safety [232].
Despite these challenges, current findings suggest that engineered probiotics are promising candidates for the treatment of IBD.

4.7. Direct Metabolic Pathways Modulation and Nanotherapeutics

The alteration of metabolic pathways in IBD has emerged as one of the most promising microbial-based therapeutic strategies. Recent literature has highlighted the critical role of tryptophan and its metabolic pathways, as several tryptophan metabolism end-products are essential for maintaining intestinal homeostasis [233]. Certain metabolites modulate the activation of the aryl hydrocarbon receptor (AhR), influencing immune cell functions, promoting IL-17 and IL-22 production, and sustaining ILC3 populations in the gut [234,235,236]. A reduced output of tryptophan metabolites contributes to intestinal inflammation and impaired tissue healing, although the precise mechanisms remain unclear [237]. In patients with IBD, serum tryptophan levels are lower than in healthy controls, with a corresponding increase in the KYN/TRP ratio [238].
Several gene loci associated with the TRP–KYN metabolic pathway, including IL23R, NOD2, and ATG16L1, have been linked to IBD susceptibility and progression [239,240]. The role of metabolites from the kynurenine pathway has been extensively studied. A recent investigation in both mice and humans demonstrated a negative correlation between lower levels of xanthurenic acid (XANA) and kynurenic acid (KYNA) and the severity of intestinal inflammation. Notably, supplementation with XANA or KYNA had protective effects on intestinal epithelial cells and T cells, reducing the severity of inflammation. Additionally, the use of recombinant aminoadipate aminotransferase (AADAT), an enzyme involved in XANA and KYNA production, modulated tryptophan metabolism and conferred protective effects in DSS-induced colitis models [241]. Another study revealed an increase in serum KYNA levels, alongside a significant decrease in serum KYNA levels, regulated by the rate-limiting enzymes IDO-1 and KAT2, in DSS-induced colitis mice [242]. However, the precise mechanisms linking tryptophan metabolism to IBD remain poorly understood, and further investigations, including randomized controlled trials, are necessary to assess effectiveness and safety in human models [242].
Oxidative stress caused by excessive reactive oxygen species (ROS) production is a key factor in the initiation and progression of IBD. Nanotherapeutics represent a novel, targeted, and safer therapeutic strategy aimed at reducing oxidative stress levels. Targeting oxidative stress through molecular pathways, such as MAPK, TLR4/NF-κB, Nrf2, and PI3K/Akt—which regulate inflammation and cellular responses to oxidative damage—offers new opportunities for IBD management [243]. In DSS-induced colitis models, LBL-CO@MPDA nanotherapeutics have shown efficacy in alleviating inflammation and restoring gut barrier integrity through multiple mechanisms, including microbiota modulation, oxidative stress reduction, and immune homeostasis restoration. Specifically, the LBL-CO@MDPA nanocomposite exploits electrostatic interactions between its negatively charged surface and positively charged inflamed colon lesions, enabling targeted accumulation in inflamed tissues. The release of carbon monoxide promotes macrophage M2 polarization via the notch/Hes1/Stat3 signaling pathway while suppressing inflammation by downregulating p38 MAPK and NF-κB (p50/p65) signaling [244].
Similarly, inflamed colon-targeted antioxidant nanotherapeutics (ICANs) modulate oxidative stress in DSS-induced colitis models. ICANs consist of mesoporous silica nanoparticles (MSNs) with surface-attached ROS-scavenging ceria nanoparticles (CeNPs) and poly(acrylic acid) (PAA), which adhere to inflamed tissues via electrostatic interactions. Orally administered ICANs have demonstrated their ability to regulate the gut microenvironment by restoring redox balance, reducing inflammatory cell infiltration, and suppressing colitis-associated immune responses [245].
Despite their specificity and precision in targeting inflamed tissues, nanotherapeutics have not yet been licensed for clinical use in IBD, primarily due to challenges in translating findings from animal models to humans. The safety and toxicity profiles of these nanoparticles in human cells remain incomplete [246]. Moreover, the complex interplay of oxidative stress, immune dysregulation, and microbiota imbalances in the IBD microenvironment is not yet fully understood. Additional concerns include the low reliability and acceptability of nanotherapeutics, as well as the potential toxicity of nanoparticles. As nanotherapeutics represent an emerging research tool, further studies are essential to determine their long-term effectiveness, safety, and pharmacokinetics.
The old and the new therapeutic tools for modulating the gut microbiota in IBDs are summarized in Figure 1.

5. Conclusions

There is strong evidence supporting the role of gut microbiota in IBD, particularly in modulating the host’s immune response and inflammation. Building on this understanding, probiotics, prebiotics, symbiotics, and postbiotics have shown promising potential, particularly in patients with UC, for inducing clinical remission, preventing relapse, and improving quality of life. However, disappointing results from other studies have limited their use, and probiotics are primarily considered an adjunct therapy alongside conventional treatments. Probiotics are generally regarded as safe, affordable, and well-tolerated, although bloating and flatulence are the most reported side effects. The wide variety of probiotic strains and inconsistent study outcomes have raised questions about their practical applicability in clinical settings.
Advances in genetic engineering have enabled the development of innovative approaches to modulate gut microbiota despite methodological variations. Fecal microbiota transplantation (FMT) is the only method that has shown promising results in human models, both in the early and advanced stages of the disease. It has demonstrated short-term efficacy, especially in ulcerative colitis (UC) models. However, major challenges remain in evaluating long-term efficacy in UC and Crohn’s disease (CD). The possibility of FMT becoming a routine clinical practice for treating inflammatory bowel diseases (IBD) is tied to issues such as donor variability and selection.
Other microbiota modulation strategies, such as next-generation probiotics, bacterial consortia, bacteriophages, pathway modulation, and nanotherapeutics, have yielded promising results in both in vitro and animal models, providing stronger scientific validation. These methods differ from older-generation techniques because they act in a more targeted manner: not only correcting dysbiosis but also directly releasing therapeutically active molecules into the gut, thereby avoiding systemic drug administration and its associated side effects. However, their application also comes with significant limitations that may hinder widespread clinical adoption.
For example, the incomplete sequencing of bacterial genomes complicates the determination of optimal bacterial species concentrations and hinders the ability to predict potential side effects. Clinical translation of these approaches has been limited, as the human microbiota differs substantially from that of animal models. Additionally, the long-term impact on host tissues and the microbial ecosystem remains poorly understood, making the long-term adaptation of the host microbiota an ongoing challenge. Potential antibiotic resistance mechanisms and the risk of exacerbating infections are additional factors that must be assessed to determine their long-term clinical applicability. Finally, manufacturing complexity and costs may limit accessibility to these therapies.
Despite these limitations, ongoing research is continuously refining these techniques, addressing safety, scalability, and regulatory concerns. The future of microbiota modulation lies in personalized, data-driven approaches that balance efficacy with minimal risk. Integrating multiomics approaches—including metagenomics, transcriptomics, and metabolomics—will enhance our ability to characterize the gut ecosystem and its interactions with the host. This will lead to the development of highly targeted treatments and move increasingly closer to the practical application of personalized medicine in IBD management.

Author Contributions

Conceptualization, M.M. and L.R.L.; data curation, M.M.; writing—original draft preparation, M.M.; writing—review and editing, P.P., B.B., F.d.V., A.D.G.; visualization, L.R.L.; supervision, L.R.L., A.P., G.C., A.G. and F.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Dorofeyev, A.E.; Vasilenko, I.V.; Rassokhina, O.A.; Kondratiuk, R.B. Mucosal barrier in ulcerative colitis and Crohn’s disease. Gastroenterol. Res. Pract. 2013, 2013, 431231. [Google Scholar] [CrossRef] [PubMed]
  2. Carrière, J.; Darfeuille-Michaud, A.; Nguyen, H.T.T. Infectious etiopathogenesis of Crohn’s disease. World J. Gastroenterol. 2014, 20, 12102–12117. [Google Scholar] [CrossRef]
  3. Knox, N.C.; Forbes, J.D.; Peterson, C.L.; Van Domselaar, G.; Bernstein, C.N. The Gut Microbiome in Inflammatory Bowel Disease: Lessons Learned from Other Immune-Mediated Inflammatory Diseases. Am. J. Gastroenterol. 2019, 114, 1051–1070. [Google Scholar] [CrossRef]
  4. Maloy, K.J.; Powrie, F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 2011, 474, 298–306. [Google Scholar] [CrossRef]
  5. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef] [PubMed]
  6. Toor, D.; Wsson, M.K.; Kumar, P.; Karthikeyan, G.; Kaushik, N.K.; Goel, C.; Singh, S.; Kumar, A.; Prakash, H. Dysbiosis Disrupts Gut Immune Homeostasis and Promotes Gastric Diseases. Int. J. Mol. Sci. 2019, 20, 2432. [Google Scholar] [CrossRef] [PubMed]
  7. Casén, C.; Vebø, H.C.; Sekelja, M.; Hegge, F.T.; Karlsson, M.K.; Ciemniejewska, E.; Dzankovic, S.; Frøyland, C.; Nestestog, R.; Engstrand, L.; et al. Deviations in human gut microbiota: A novel diagnostic test for determining dysbiosis in patients with IBS or IBD. Aliment. Pharmacol. Ther. 2015, 42, 71–83. [Google Scholar] [CrossRef]
  8. 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. Bosn. J. Basic Med. Sci. 2021, 21, 270–283. [Google Scholar] [CrossRef]
  9. Garrett, W.S.; Gordon, J.I.; Glimcher, L.H. Homeostasis and inflammation in the intestine. Cell 2010, 140, 859–870. [Google Scholar] [CrossRef]
  10. Hedin, C.R.; McCarthy, N.E.; Louis, P.; Farquharson, F.M.; McCartney, S.; Taylor, K.; Prescott, N.J.; Murrells, T.; Stagg, A.J.; Whelan, K.; et al. Altered intestinal microbiota and blood T cell phenotype are shared by patients with Crohn’s disease and their unaffected siblings. Gut 2014, 63, 1578–1586. [Google Scholar] [CrossRef]
  11. Willing, B.P.; Dicksved, J.; Halfvarson, J.; Andersson, A.F.; Lucio, M.; Zheng, Z.; Järnerot, G.; Tysk, C.; Jansson, J.K.; Engstrand, L. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 2010, 139, 1844–1854.e1. [Google Scholar] [CrossRef]
  12. Ashton, J.J.; Seaby, E.G.; Beattie, R.M.; Ennis, S. NOD2 in Crohn’s Disease-Unfinished Business. J. Crohn’s Colitis 2023, 17, 450–458. [Google Scholar] [CrossRef] [PubMed]
  13. Luo, P.; Yang, Z.; Chen, B.; Zhong, X. The multifaceted role of CARD9 in inflammatory bowel disease. J. Cell. Mol. Med. 2020, 24, 34–39. [Google Scholar] [CrossRef] [PubMed]
  14. Lavoie, S.; Conway, K.L.; Lassen, K.G.; Jijon, H.B.; Pan, H.; Chun, E.; Michaud, M.; Lang, J.K.; Gallini Comeau, C.A.; Dreyfuss, J.M.; et al. The Crohn’s disease polymorphism, ATG16L1 T300A, alters the gut microbiota and enhances the local Th1/Th17 response. eLfie 2019, 8, e39982. [Google Scholar] [CrossRef]
  15. Frank, D.N.; Robertson, C.E.; Hamm, C.M.; Kpadeh, Z.; Zhang, T.; Chen, H.; Zhu, W.; Sartor, R.B.; Boedeker, E.C.; Harpaz, N.; et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 179–184. [Google Scholar] [CrossRef] [PubMed]
  16. Knights, D.; Silverberg, M.S.; Weersma, R.K.; Gevers, D.; Dijkstra, G.; Huang, H.; Tyler, A.D.; van Sommeren, S.; Imhann, F.; Stempak, J.M.; et al. Complex host genetics influence the microbiome in inflammatory bowel disease. Genome Med. 2014, 6, 107. [Google Scholar] [CrossRef]
  17. Hu, S.; Vich Vila, A.; Gacesa, R.; Collij, V.; Stevens, C.; Fu, J.M.; Wong, I.; Talkowski, M.E.; Rivas, M.A.; Imhann, F.; et al. Whole Exome Sequencing Analyses Reveal Gene–Microbiota Interactions in the Context of IBD. Gut 2021, 70, 285–296. [Google Scholar] [CrossRef]
  18. Imhann, F.; Vich Vila, A.; Bonder, M.J.; Fu, J.; Gevers, D.; Visschedijk, M.C.; Spekhorst, L.M.; Alberts, R.; Franke, L.; van Dullemen, H.M.; et al. Interplay of host genetics and gut microbiota underlying the onset and clinical presentation of inflammatory bowel disease. Gut 2018, 67, 108–119. [Google Scholar] [CrossRef]
  19. Ananthakrishnan, A.N. Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 205–217. [Google Scholar] [CrossRef]
  20. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef]
  21. Biedermann, L.; Brülisauer, K.; Zeitz, J.; Frei, P.; Scharl, M.; Vavricka, S.R.; Fried, M.; Loessner, M.J.; Rogler, G.; Schuppler, M. Smoking cessation alters intestinal microbiota: Insights from quantitative investigations on human fecal samples using FISH. Inflamm. Bowel Dis. 2014, 20, 1496–1501. [Google Scholar] [CrossRef]
  22. Allais, L.; Kerckhof, F.M.; Verschuere, S.; Bracke, K.R.; De Smet, R.; Laukens, D.; Van den Abbeele, P.; De Vos, M.; Boon, N.; Brusselle, G.G.; et al. Chronic cigarette smoke exposure induces microbial and inflammatory shifts and mucin changes in the murine gut. Environ. Microbiol. 2016, 18, 1352–1363. [Google Scholar] [CrossRef] [PubMed]
  23. Adolph, T.E.; Zhang, J. Diet fuelling inflammatory bowel diseases: Preclinical and clinical concepts. Gut 2022, 71, 2574–2586. [Google Scholar] [CrossRef] [PubMed]
  24. Agus, A.; Denizot, J.; Thévenot, J.; Martinez-Medina, M.; Massier, S.; Sauvanet, P.; Bernalier-Donadille, A.; Denis, S.; Hofman, P.; Bonnet, R.; et al. Western diet induces a shift in microbiota composition enhancing susceptibility to adherent-invasive E. coli infection and intestinal inflammation. Sci. Rep. 2016, 6, 19032. [Google Scholar] [CrossRef]
  25. Cabral, D.J.; Wurster, J.I.; Korry, B.J.; Penumutchu, S.; Belenky, P. Consumption of a Western-style diet modulates the response of the murine gut microbiome to ciprofloxacin. mSystems 2020, 5, e00317–20. [Google Scholar] [CrossRef]
  26. Martinez-Medina, M.; Denizot, J.; Dreux, N.; Robin, F.; Billard, E.; Bonnet, R.; Darfeuille-Michaud, A.; Barnich, N. Western diet induces dysbiosis with increased E coli in CEABAC10 mice, alters host barrier function favouring AIEC colonisation. Gut 2014, 63, 116–124. [Google Scholar] [CrossRef]
  27. Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef]
  28. Pan, R.; He, Y.; Yuan, J.; Zhao, S.; Ma, M.; Chai, Z.; Ji, X.; Hu, X.; He, C.; Zhou, D.; et al. The Role of Antibiotic Exposure as Risk Factor for IBD Epidemic: An Updated Meta-Analysis. J. Gastroenterol. Hepatol. 2024, 39, 2561–2571. [Google Scholar] [CrossRef]
  29. Mårild, K.; Lerchova, T.; Östensson, M.; Imberg, H.; Størdal, K.; Ludvigsson, J. Early-Life Infections, Antibiotics and Later Risk of Childhood and Early Adult-Onset Inflammatory Bowel Disease: Pooled Analysis of Two Scandinavian Birth Cohorts. Aliment. Pharmacol. Ther. 2024, 61, 323–334. [Google Scholar] [CrossRef]
  30. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  31. Xiong, R.G.; Li, J.; Cheng, J.; Zhou, D.D.; Wu, S.X.; Huang, S.Y.; Saimaiti, A.; Yang, Z.J.; Gan, R.Y.; Li, H.B. The Role of Gut Microbiota in Anxiety, Depression, and Other Mental Disorders as Well as the Protective Effects of Dietary Components. Nutrients 2023, 15, 3258. [Google Scholar] [CrossRef]
  32. Yuan, X.; Chen, B.; Duan, Z.; Xia, Z.; Ding, Y.; Chen, T.; Liu, H.; Wang, B.; Yang, B.; Wang, X.; et al. Depression and anxiety in patients with active ulcerative colitis: Crosstalk of gut microbiota, metabolomics and proteomics. Gut Microbes 2021, 13, 1987779. [Google Scholar] [CrossRef]
  33. Humbel, F.; Rieder, J.H.; Franc, Y.; Juillerat, P.; Scharl, M.; Misselwitz, B.; Schreiner, P.; Begré, S.; Rogler, G.; von Känel, R.; et al. Association of alterations in intestinal microbiota with impaired psychological function in patients with inflammatory bowel diseases in remission. Clin. Gastroenterol. Hepatol. 2020, 18, 2019–2029.e11. [Google Scholar] [CrossRef]
  34. Lo Presti, A.; Zorzi, F.; Del Chierico, F.; Altomare, A.; Cocca, S.; Avola, A.; De Biasio, F.; Russo, A.; Cella, E.; Reddel, S.; et al. Fecal and mucosal microbiota profiling in irritable bowel syndrome and inflammatory bowel disease. Front. Microbiol. 2019, 10, 1655. [Google Scholar] [CrossRef]
  35. Gkouskou, K.K.; Deligianni, C.; Tsatsanis, C.; Eliopoulos, A.G. The gut microbiota in mouse models of inflammatory bowel disease. Front. Cell. Infect. Microbiol. 2014, 4, 28. [Google Scholar] [CrossRef]
  36. Rosen, C.E.; Palm, N.W. Navigating the Microbiota Seas: Triangulation Finds a Way Forward. Cell Host Microbe 2018, 23, 1–3. [Google Scholar] [CrossRef]
  37. Orel, R.; Trop, T.K. Intestinal microbiota, probiotics and prebiotics in inflammatory bowel disease. World J. Gastroenterol. 2014, 20, 11505–11524. [Google Scholar] [CrossRef]
  38. Mullish, B.H.; Tohumcu, E.; Porcari, S.; Fiorani, M.; Di Tommaso, N.; Gasbarrini, A.; Cammarota, G.; Ponziani, F.R.; Ianiro, G. The role of faecal microbiota transplantation in chronic noncommunicable disorders. J. Autoimmun. 2023, 141, 103034. [Google Scholar] [CrossRef]
  39. Airola, C.; Severino, A.; Porcari, S.; Fusco, W.; Mullish, B.H.; Gasbarrini, A.; Cammarota, G.; Ponziani, F.R.; Ianiro, G. Future Modulation of Gut Microbiota: From Eubiotics to FMT, Engineered Bacteria, and Phage Therapy. Antibiotics 2023, 12, 868. [Google Scholar] [CrossRef]
  40. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
  41. Hoffmann, C.; Dollive, S.; Grunberg, S.; Chen, J.; Li, H.; Wu, G.D.; Lewis, J.D.; Bushman, F.D. Archaea and fungi of the human gut microbiome: Correlations with diet and bacterial residents. PLoS ONE 2013, 8, e66019. [Google Scholar] [CrossRef]
  42. Tlaskalová-Hogenová, H.; Stěpánková, R.; Kozáková, H.; Hudcovic, T.; Vannucci, L.; Tučková, L.; Rossmann, P.; Hrnčíř, T.; Kverka, M.; Zákostelská, Z.; et al. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: Contribution of germ-free and gnotobiotic animal models of human diseases. Cell. Mol. Immunol. 2011, 8, 110–120. [Google Scholar] [CrossRef]
  43. Koboziev, I.; Webb, C.; Furr, K.L.; Grisham, M.B. Role of the enteric microbiota in intestinal homeostasis and inflammation. Free Radic Biol. Med. 2014, 68, 122–133. [Google Scholar] [CrossRef]
  44. Iebba, V.; Totino, V.; Gagliardi, A.; Santangelo, F.; Cacciotti, F.; Trancassini, M.; Mancini, C.; Cicerone, C.; Corazziari, E.; Pantanella, F.; et al. Eubiosis and dysbiosis: The two sides of the microbiota. New Microbiol. 2016, 39, 1–12. [Google Scholar]
  45. Talapko, J.; Včev, A.; Meštrović, T.; Pustijanac, E.; Jukić, M.; Škrlec, I. Homeostasis and Dysbiosis of the Intestinal Microbiota: Comparing Hallmarks of a Healthy State with Changes in Inflammatory Bowel Disease. Microorganisms 2022, 10, 2405. [Google Scholar] [CrossRef]
  46. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  47. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short chain fatty acids (SCFAs)-mediated gut microbiota-host interaction: Role in health and disease. Front. Immunol. 2019, 10, 277. [Google Scholar] [CrossRef]
  48. Ciesielska, A.; Matyjek, M.; Kwiatkowska, K. TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cell. Mol. Life Sci. 2021, 78, 1233–1261. [Google Scholar] [CrossRef]
  49. Fava, F.; Danese, S. Intestinal microbiota in inflammatory bowel disease: Friend of foe? World J. Gastroenterol. 2011, 17, 557–566. [Google Scholar] [CrossRef]
  50. Rigottier-Gois, L. Dysbiosis in inflammatory bowel diseases: The oxygen hypothesis. ISME J. 2013, 7, 1256–1261. [Google Scholar] [CrossRef]
  51. Nenkov, M.; Shi, Y.; Ma, Y.; Gaßler, N.; Chen, Y. Targeting Farnesoid X Receptor in Tumor and the Tumor Microenvironment: Implication for Therapy. Int. J. Mol. Sci. 2023, 25, 6. [Google Scholar] [CrossRef]
  52. Fu, Y.; Lyu, J.; Wang, S. The role of intestinal microbes on intestinal barrier function and host immunity from a metabolite perspective. Front. Immunol. 2023, 14, 1277102. [Google Scholar] [CrossRef]
  53. Glassner, K.L.; Abraham, B.P.; Quigley, E.M.M. The microbiome and inflammatory bowel disease. J. Allergy Clin. Immunol. 2020, 145, 16–27. [Google Scholar] [CrossRef]
  54. Abdel-Rahman, L.I.H.; Morgan, X.C. Searching for a Consensus Among Inflammatory Bowel Disease Studies: A Systematic Meta-Analysis. Inflamm. Bowel Dis. 2023, 29, 125–139. [Google Scholar] [CrossRef]
  55. Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 3464. [Google Scholar] [CrossRef]
  56. Mah, C.; Jayawardana, T.; Leong, G.; Koentgen, S.; Lemberg, D.; Connor, S.J.; Rokkas, T.; Grimm, M.C.; Leach, S.T.; Hold, G.L. Assessing the Relationship between the Gut Microbiota and Inflammatory Bowel Disease Therapeutics: A Systematic Review. Pathogens 2023, 12, 262. [Google Scholar] [CrossRef]
  57. Khan, I.; Ullah, N.; Zha, L.; Bai, Y.; Khan, A.; Zhao, T.; Che, T.; Zhang, C. Alteration of gut microbiota in inflammatory bowel disease (IBD): Cause or consequence? IBD treatment targeting the gut microbiome. Pathogens 2019, 8, 126. [Google Scholar] [CrossRef]
  58. Gevers, D.; Kugathasan, S.; Denson, L.A.; Vázquez-Baeza, Y.; Van Treuren, W.; Ren, B.; Schwager, E.; Knights, D.; Song, S.J.; Yassour, M.; et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe 2014, 15, 382–392. [Google Scholar] [CrossRef]
  59. Maukonen, J.; Kolho, K.L.; Paasela, M.; Honkanen, J.; Klemetti, P.; Vaarala, O.; Saarela, M. Altered Fecal Microbiota in Paediatric Inflammatory Bowel Disease. J. Crohn’s Colitis 2015, 9, 1088–1095. [Google Scholar] [CrossRef]
  60. Zhou, Y.; Xu, H.; Xu, J.; Guo, X.; Zhao, H.; Chen, Y.; Zhou, Y.; Nie, Y.F. prausnitzii and its supernatant increase SCFAs-producing bacteria to restore gut dysbiosis in TNBS-induced colitis. AMB Express 2021, 11, 33. [Google Scholar] [CrossRef] [PubMed]
  61. Zhang, T.; Ji, X.; Lu, G.; Zhang, F. The potential of Akkermansia muciniphila in inflammatory bowel disease. Appl. Microbiol. Biotechnol. 2021, 105, 5785–5794. [Google Scholar] [CrossRef]
  62. Faden, H. The Role of Faecalibacterium, Roseburia, and Butyrate in Inflammatory Bowel Disease. Dig. Dis. 2022, 40, 793–795. [Google Scholar] [CrossRef] [PubMed]
  63. Sha, S.; Xu, B.; Wang, X.; Zhang, Y.; Wang, H.; Kong, X.; Zhu, H.; Wu, K. The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn. Microbiol. Infect. Dis. 2013, 75, 245–251. [Google Scholar] [CrossRef] [PubMed]
  64. Nemoto, H.; Kataoka, K.; Ishikawa, H.; Ikata, K.; Arimochi, H.; Iwasaki, T.; Ohnishi, Y.; Kuwahara, T.; Yasutomo, K. Reduced diversity and imbalance of fecal microbiota in patients with ulcerative colitis. Dig. Dis. Sci. 2012, 57, 2955–2964. [Google Scholar] [CrossRef]
  65. Li, J.; Butcher, J.; Mack, D.; Stintzi, A. Functional impacts of the intestinal microbiome in the pathogenesis of inflammatory bowel disease. Inflamm. Bowel Dis. 2015, 21, 139–153. [Google Scholar] [CrossRef]
  66. Mondot, S.; Kang, S.; Furet, J.P.; Aguirre de Carcer, D.; McSweeney, C.; Morrison, M.; Marteau, P.; Doré, J.; Leclerc, M. Highlighting new phylogenetic specificities of Crohn’s disease microbiota. Inflamm. Bowel Dis. 2011, 17, 185–192. [Google Scholar] [CrossRef]
  67. Sartor, R.B.; Wu, G.D. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017, 152, 327–339.e4. [Google Scholar] [CrossRef]
  68. Lo Sasso, G.; Khachatryan, L.; Kondylis, A.; Battey, J.N.D.; Sierro, N.; Danilova, N.A.; Grigoryeva, T.V.; Markelova, M.I.; Khusnutdinova, D.R.; Laikov, A.V.; et al. Inflammatory Bowel Disease-Associated Changes in the Gut: Focus on Kazan Patients. Inflamm. Bowel Dis. 2021, 27, 418–433. [Google Scholar] [CrossRef]
  69. Zhao, H.; Xu, H.; Chen, S.; He, J.; Zhou, Y.; Nie, Y. Systematic review and meta-analysis of the role of Faecalibacterium prausnitzii alteration in inflammatory bowel disease. J. Gastroenterol. Hepatol. 2021, 36, 320–328. [Google Scholar] [CrossRef] [PubMed]
  70. Schäffler, H.; Kaschitzki, A.; Alberts, C.; Bodammer, P.; Bannert, K.; Köller, T.; Warnke, P.; Kreikemeyer, B.; Lamprecht, G. Alterations in the mucosa-associated bacterial composition in Crohn’s disease: A pilot study. Int. J. Color. Dis. 2016, 31, 961–971. [Google Scholar] [CrossRef]
  71. Sokol, H.; Seksik, P.; Furet, J.P.; Firmesse, O.; Nion-Larmurier, I.; Beaugerie, L.; Cosnes, J.; Corthier, G.; Marteau, P.; Doré, J. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm. Bowel Dis. 2009, 15, 1183–1189. [Google Scholar] [CrossRef] [PubMed]
  72. Lopez-Siles, M.; Duncan, S.H.; Garcia-Gil, L.J.; Martinez-Medina, M. Faecalibacterium prausnitzii: From microbiology to diagnostics and prognostics. ISME J. 2017, 11, 841–852. [Google Scholar] [CrossRef]
  73. Scanu, M.; Toto, F.; Petito, V.; Masi, L.; Fidaleo, M.; Puca, P.; Baldelli, V.; Reddel, S.; Vernocchi, P.; Pani, G.; et al. An integrative multi-omic analysis defines gut microbiota, mycobiota, and metabolic fingerprints in ulcerative colitis patients. Front. Cell Infect. Microbiol. 2024, 14, 1366192. [Google Scholar] [CrossRef] [PubMed]
  74. Khan, K.J.; Ullman, T.A.; Ford, A.C.; Abreu, M.T.; Abadir, A.; Marshall, J.K.; Talley, N.J.; Moayyedi, P. Antibiotic therapy in inflammatory bowel disease: A systematic review and meta-analysis. Am. J. Gastroenterol. 2011, 106, 661–673. [Google Scholar] [CrossRef]
  75. Wang, S.L.; Wang, Z.R.; Yang, C.Q. Meta-analysis of broad-spectrum antibiotic therapy in patients with active inflammatory bowel disease. Exp. Ther. Med. 2012, 4, 1051–1056. [Google Scholar] [CrossRef]
  76. Torres, J.; Bonovas, S.; Doherty, G.; Kucharzik, T.; Gisbert, J.P.; Raine, T.; Adamina, M.; Armuzzi, A.; Bachmann, O.; Bager, P.; et al. ECCO guidelines on therapeutics in Crohn’s disease: Medical treatment. J. Crohn’s Colitis 2020, 14, 4–22. [Google Scholar] [CrossRef]
  77. Su, J.W.; Ma, J.J.; Zhang, H.J. Use of antibiotics in patients with Crohn’s disease: A systematic review and meta-analysis. J. Dig. Dis. 2015, 16, 58–66. [Google Scholar] [CrossRef] [PubMed]
  78. Vaughn, B.P.; Moss, A.C. Prevention of post-operative recurrence of Crohn’s disease. World J. Gastroenterol. 2014, 20, 1147–1154. [Google Scholar] [CrossRef]
  79. Guslandi, M. Rifaximin in the treatment of inflammatory bowel disease. World J. Gastroenterol. 2011, 17, 4643–4646. [Google Scholar] [CrossRef]
  80. Bar, N.; Avraham, Y.; Dubinsky, V.; Cohen, N.A.; Weiss, G.A.; Banon, L.; Tulchinsky, H.; Maharshak, N.; Gophna, U.; Dotan, I. Long-term Antibiotic Treatment in Pouchitis—Patterns of Use and Safety. Inflamm. Bowel Dis. 2022, 28, 1027–1033. [Google Scholar] [CrossRef]
  81. Hildebrand, H.; Malmborg, P.; Askling, J.; Ekbom, A.; Montgomery, S.M. Early-life exposures associated with antibiotic use and risk of subsequent Crohn’s disease. Scand. J. Gastroenterol. 2008, 43, 961–966. [Google Scholar] [CrossRef]
  82. Turner, A.M.; Li, L.; Monk, I.R.; Lee, J.Y.H.; Ingle, D.J.; Portelli, S.; Sherry, N.L.; Isles, N.; Seemann, T.; Sharkey, L.K.; et al. Rifaximin prophylaxis causes resistance to the last-resort antibiotic daptomycin. Nature 2024, 635, 969–977. [Google Scholar] [CrossRef]
  83. Scott, B.M.; Gutiérrez-Vázquez, C.; Sanmarco, L.M.; da Silva Pereira, J.A.; Li, Z.; Plasencia, A.; Hewson, P.; Cox, L.M.; O’Brien, M.; Chen, S.K.; et al. Self-tunable engineered yeast probiotics for the treatment of inflammatory bowel disease. Nat. Med. 2021, 27, 1212–1222. [Google Scholar] [CrossRef] [PubMed]
  84. Müller, L.; Lorentz, A. Probiotics in the Treatment of Inflammatory Bowel Diseases in Adulthood: A Systematic Review. J. Gastrointest. Liver Dis. 2022, 31, 74–84. [Google Scholar] [CrossRef]
  85. Jakubczyk, D.; Leszczyńska, K.; Górska, S. The effectiveness of probiotics in the treatment of inflammatory bowel disease (Ibd)—A critical review. Nutrients 2020, 12, 1973. [Google Scholar] [CrossRef]
  86. Yan, F.; Polk, D.B. Probiotics and immune health. Curr. Opin. Gastroenterol. 2011, 27, 496–501. [Google Scholar] [CrossRef]
  87. Ojetti, V.; Saviano, A.; Brigida, M.; Petruzziello, C.; Caronna, M.; Gayani, G.; Franceschi, F. Randomized control trial on the efficacy of Limosilactobacillus reuteri ATCC PTA 4659 in reducing inflammatory markers in acute uncomplicated diverticulitis. Eur. J. Gastroenterol. Hepatol. 2022, 34, 496–502. [Google Scholar] [CrossRef]
  88. Sun, Y.Y.; Li, M.; Li, Y.Y.; Li, L.X.; Zhai, W.Z.; Wang, P.; Yang, X.X.; Gu, X.; Song, L.J.; Li, Z.; et al. The effect of Clostridium butyricum on symptoms and fecal microbiota in diarrhea-dominant irritable bowel syndrome: A randomized, double-blind, placebo-controlled trial. Sci. Rep. 2018, 8, 2964. [Google Scholar] [CrossRef]
  89. Bourreille, A.; Cadiot, G.; Le Dreau, G.; Laharie, D.; Beaugerie, L.; Dupas, J.L.; Marteau, P.; Rampal, P.; Moyse, D.; Saleh, A.; et al. Saccharomyces boulardii does not prevent relapse of Crohn’s disease. Clin. Gastroenterol. Hepatol. 2013, 11, 982–987. [Google Scholar] [CrossRef]
  90. Bjarnason, I.; Sission, G.; Hayee, B.H. 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, 465–473. [Google Scholar] [CrossRef]
  91. Park, S.K.; Kang, S.B.; Kim, S.; Kim, T.O.; Cha, J.M.; Im, J.P.; Choi, C.H.; Kim, E.S.; Seo, G.S.; Eun, C.S.; et al. Additive effect of probiotics (Mutaflor) on 5-aminosalicylic acid therapy in patients with ulcerative colitis. Korean J. Intern. Med. 2022, 37, 949–957. [Google Scholar] [CrossRef]
  92. Agraib, L.M.; Yamani, M.I.; Tayyem, R.; Abu-Sneineh, A.T.; Rayyan, Y.M. Probiotic supplementation induces remission and changes in the immunoglobulins and inflammatory response in active ulcerative colitis patients: A pilot, randomized, double-blind, placebo-controlled study. Clin. Nutr. ESPEN 2022, 51, 83–91. [Google Scholar] [CrossRef] [PubMed]
  93. Oh, G.M.; Moon, W.; Seo, K.I.; Jung, K.; Kim, J.H.; Kim, S.E.; Park, M.I.; Park, S.J. Therapeutic Potential of Escherichia coli Nissle 1917 in Clinically Remission-attained Ulcerative Colitis Patients: A Hospital-based Cohort Study. Korean J. Gastroenterol. 2021, 77, 12–21. [Google Scholar] [CrossRef]
  94. Yılmaz, İ.; Enver Dolar, M.; Özpınar, H. Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk. J. Gastroenterol. 2018, 30, 242–253. [Google Scholar] [CrossRef] [PubMed]
  95. Matsuoka, K.; Uemura, Y.; Kanai, T.; Kunisaki, R.; Suzuki, Y.; Yokoyama, K.; Yoshimura, N.; Hibi, T. Efficacy of Bifidobacterium breve Fermented Milk in Maintaining Remission of Ulcerative Colitis. Dig. Dis. Sci. 2018, 63, 1910–1919. [Google Scholar] [CrossRef]
  96. Yoshimatsu, Y.; Yamada, A.; Furukawa, R.; Sono, K.; Osamura, A.; Nakamura, K.; Aoki, H.; Tsuda, Y.; Hosoe, N.; Takada, N.; et al. Effectiveness of probiotic therapy for the prevention of relapse in patients with inactive ulcerative colitis. World J. Gastroenterol. 2015, 21, 5985–5994. [Google Scholar] [CrossRef]
  97. Oliva, S.; Di Nardo, G.; Ferrari, F.; Mallardo, S.; Rossi, P.; Patrizi, G.; Cucchiara, S.; Stronati, L. Randomised clinical trial: The effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment. Pharmacol. Ther. 2012, 35, 327–334. [Google Scholar] [CrossRef]
  98. Tursi, A.; Brandimarte, G.; Papa, A.; Giglio, A.; Elisei, W.; Giorgetti, G.M.; Forti, G.; Morini, S.; Hassan, C.; Pistoia, M.A.; et al. Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: A double-blind, randomized, placebo-controlled study. Am. J. Gastroenterol. 2010, 105, 2218–2227. [Google Scholar] [CrossRef]
  99. Miele, E.; Pascarella, F.; Giannetti, E.; Quaglietta, L.; Baldassano, R.N.; Staiano, A. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am. J. Gastroenterol. 2009, 104, 437–443. [Google Scholar] [CrossRef]
  100. Zocco, M.A.; dal Verme, L.Z.; Cremonini, F.; Piscaglia, A.C.; Nista, E.C.; Candelli, M.; Novi, M.; Rigante, D.; Cazzato, I.A.; Ojetti, V.; et al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment. Pharmacol. Ther. 2006, 23, 1567–1574. [Google Scholar] [CrossRef]
  101. Kato, K.; Mizuno, S.; Umesaki, Y.; Ishii, Y.; Sugitani, M.; Imaoka, A.; Otsuka, M.; Hasunuma, O.; Kurihara, R.; Iwasaki, A.; et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment. Pharmacol. Ther. 2004, 20, 1133–1141. [Google Scholar] [CrossRef] [PubMed]
  102. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, types, sources, mechanisms, and clinical applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef]
  103. Wilson, B.; Whelan, K. Prebiotic inulin-type fructans and galacto-oligosaccharides: Definition, specificity, function, and application in gastrointestinal disorders. J. Gastroenterol. Hepatol. 2017, 32, 64–68. [Google Scholar] [CrossRef]
  104. Hughes, R.L.; Alvarado, D.A.; Swanson, K.S.; Holscher, H.D. The Prebiotic Potential of Inulin-Type Fructans: A Systematic Review. Adv. Nutr. 2022, 13, 492–529. [Google Scholar] [CrossRef] [PubMed]
  105. Rose, E.C.; Odle, J.; Blikslager, A.T.; Ziegler, A.L. Probiotics, Prebiotics and Epithelial Tight Junctions: A Promising Approach to Modulate Intestinal Barrier Function. Int. J. Mol. Sci. 2021, 22, 6729. [Google Scholar] [CrossRef]
  106. Shokryazdan, P.; Faseleh Jahromi, M.; Navidshad, B.; Liang, J.B. Effects of prebiotics on immune system and cytokine expression. Med. Microbiol. Immunol. 2017, 206, 1–9. [Google Scholar] [CrossRef]
  107. Masanetz, S.; Preißinger, W.; Meyer, H.H.; Pfaffl, M.W. Effects of the prebiotics inulin and lactulose on intestinal immunology and hematology of preruminant calves. Animal 2011, 5, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  108. Yang, C.; Du, Y.; Li, Q.; Liu, L.; Zhao, L.; Gao, C.; Tang, Z.; Zhang, X.; Zhao, Y.; Yang, X. Fructo-oligosaccharides Alleviated Ulcerative Colitis via Gut Microbiota-Dependent Tryptophan Metabolism in Association with Aromatic Hydrocarbon Receptor Activation in Mice. J. Agric. Food Chem. 2024, 72, 27912–27922. [Google Scholar] [CrossRef]
  109. Capitán-Cañadas, F.; Ocón, B.; Aranda, C.J.; Anzola, A.; Suárez, M.D.; Zarzuelo, A.; de Medina, F.S.; Martínez-Augustin, O. Fructooligosaccharides exert intestinal anti-inflammatory activity in the CD4+ CD62L+ T cell transfer model of colitis in C57BL/6J mice. Eur. J. Nutr. 2016, 55, 1445–1454. [Google Scholar] [CrossRef]
  110. Lindsay, J.O.; Whelan, K.; Stagg, A.J.; Gobin, P.; Al-Hassi, H.O.; Rayment, N.; Kamm, M.A.; Knight, S.C.; Forbes, A. Clinical, microbiological, and immunological effects of fructo-oligosaccharide in patients with Crohn’s disease. Gut 2006, 55, 348–355. [Google Scholar] [CrossRef]
  111. Casellas, F.; Borruel, N.; Torrejón, A.; Varela, E.; Antolin, M.; Guarner, F.; Malagelada, J.R. Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Aliment. Pharmacol. Ther. 2007, 25, 1061–1067. [Google Scholar] [CrossRef] [PubMed]
  112. Ikegami, S.; Nakamura, M.; Honda, T.; Yamamura, T.; Maeda, K.; Sawada, T.; Ishikawa, E.; Yamamoto, K.; Furune, S.; Ishikawa, T.; et al. Efficacy of 1-kestose supplementation in patients with mild to moderate ulcerative colitis: A randomised, double-blind, placebo-controlled pilot study. Aliment. Pharmacol. Ther. 2023, 57, 1249–1257. [Google Scholar] [CrossRef]
  113. Valcheva, R.; Koleva, P.; Martínez, I.; Walter, J.; Gänzle, M.G.; Dieleman, L.A. Inulin-type fructans improve active ulcerative colitis associated with microbiota changes and increased short-chain fatty acids levels. Gut Microbes 2019, 10, 334–357. [Google Scholar] [CrossRef]
  114. Zhang, T.; Zhang, W.; Feng, C.; Kwok, L.Y.; He, Q.; Sun, Z. Stronger gut microbiome modulatory effects by postbiotics than probiotics in a mouse colitis model. NPJ Sci. Food 2022, 6, 53. [Google Scholar] [CrossRef]
  115. Facchin, S.; Vitulo, N.; Calgaro, M.; Buda, A.; Romualdi, C.; Pohl, D.; Perini, B.; Lorenzon, G.; Marinelli, C.; D’Incà, R.; et al. Microbiota changes induced by microencapsulated sodium butyrate in patients with inflammatory bowel disease. Neurogastroenterol. Motil. 2020, 32, e13914. [Google Scholar] [CrossRef]
  116. Henn, M.R.; O’Brien, E.J.; Diao, L.; Feagan, B.G.; Sandborn, W.J.; Huttenhower, C.; Wortman, J.R.; McGovern, B.H.; Wang-Weigand, S.; Lichter, D.I.; et al. A Phase 1b Safety Study of SER-287, a Spore-Based Microbiome Therapeutic, for Active Mild to Moderate Ulcerative Colitis. Gastroenterology 2021, 160, 115–127.e30. [Google Scholar] [CrossRef] [PubMed]
  117. Ivanovska, T.P.; Mladenovska, K.; Zhivikj, Z.; Pavlova, M.J.; Gjurovski, I.; Ristoski, T.; Petrushevska-Tozi, L. Synbiotic loaded chitosan-Ca-alginate microparticles reduces inflammation in the TNBS model of rat colitis. Int. J. Pharm. 2017, 527, 126–134. [Google Scholar] [CrossRef]
  118. Wang, Y.N.; Meng, X.C.; Dong, Y.F.; Zhao, X.H.; Qian, J.M.; Wang, H.Y.; Li, J.N. Effects of probiotics and prebiotics on intestinal microbiota in mice with acute colitis based on 16S rRNA gene sequencing. Chin. Med. J. 2019, 132, 1833–1842. [Google Scholar] [CrossRef] [PubMed]
  119. Furrie, E.; Macfarlane, S.; Kennedy, A.; Cummings, J.H.; Walsh, S.V.; O’neil, D.A.; Macfarlane, G.T. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: A randomised controlled pilot trial. Gut 2005, 54, 242–249. [Google Scholar] [CrossRef]
  120. Liu, Z.; Liu, F.; Wang, W.; Sun, C.; Gao, D.; Ma, J.; Hussain, M.A.; Xu, C.; Jiang, Z.; Hou, J. Study of the alleviation effects of a combination of Lactobacillus rhamnosus and inulin on mice with colitis. Food Funct. 2020, 11, 3823–3837. [Google Scholar] [CrossRef]
  121. Yu, F.; Hu, X.; Ren, H.; Wang, X.; Shi, R.; Guo, J.; Chang, J.; Zhou, X.; Jin, Y.; Li, Y.; et al. Protective effect of synbiotic combination of Lactobacillus plantarum SC-5 and olive oil extract tyrosol in a murine model of ulcerative colitis. J. Transl. Med. 2024, 22, 308. [Google Scholar] [CrossRef]
  122. Quaranta, G.; Guarnaccia, A.; Fancello, G.; Agrillo, C.; Iannarelli, F.; Sanguinetti, M.; Masucci, L. Fecal Microbiota Transplantation and Other Gut Microbiota Manipulation Strategies. Microorganisms 2022, 10, 2424. [Google Scholar] [CrossRef]
  123. Varga, A.; Kocsis, B.; Sipos, D.; Kása, P.; Vigvári, S.; Pál, S.; Dembrovszky, F.; Farkas, K.; Péterfi, Z. How to Apply FMT More Effectively, Conveniently and Flexible—A Comparison of FMT Methods. Front. Cell. Infect. Microbiol. 2021, 11, 657320. [Google Scholar] [CrossRef] [PubMed]
  124. Verdier, C.; Denis, S.; Gasc, C.; Boucinha, L.; Uriot, O.; Delmas, D.; Dore, J.; Le Camus, C.; Schwintner, C.; Blanquet-Diot, S. An Oral FMT Capsule as Efficient as an Enema for Microbiota Reconstruction Following Disruption by Antibiotics, as Assessed in an In Vitro Human Gut Model. Microorganisms 2021, 9, 358. [Google Scholar] [CrossRef] [PubMed]
  125. van Nood, E.; Vrieze, A.; Nieuwdorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.; Tijssen, J.G.; et al. Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef] [PubMed]
  126. Berg, D.; Clemente, J.C.; Colombel, J.F. Can inflammatory bowel disease be permanently treated with short-term interventions on the microbiome? Expert Rev. Gastroenterol. Hepatol. 2015, 9, 781–795. [Google Scholar] [CrossRef]
  127. Deleu, S.; Caenepeel, C.; Vazquez Castellanos, J.F.; Arnauts, K.; Braekeleire, S.; Machiels, K.; Baert, F.; Mana, F.; Pouillon, L.; Hindryckx, P.; et al. DOP48 Faecal microbiota transplantation in active Ulcerative Colitis: Key lessons from a randomized controlled trial halted for futility. J. Crohn’s Colitis 2024, 18, 159–160. [Google Scholar] [CrossRef]
  128. Haifer, C.; Paramsothy, S.; Kaakoush, N.O.; Saikal, A.; Ghaly, S.; Yang, T.; Luu, L.D.W.; Borody, T.J.; Leong, R.W. Lyophilised oral faecal microbiota transplantation for ulcerative colitis (LOTUS): A randomised, double-blind, placebo-controlled trial. Lancet. Gastroenterol. Hepatol. 2022, 7, 141–151. [Google Scholar] [CrossRef]
  129. Shabat, S.C.; Scaldaferri, F.; Zittan, E.; Hirsch, A.; Mentella, M.C.; Musca, T.; Cohen, N.A.; Ron, Y.; Fliss Isakov, N.; Pfeffer, J.; et al. Use of Faecal Transplantation with a Novel Diet for Mild to Moderate Active Ulcerative Colitis: The CRAFT UC Randomised Controlled Trial. J. Crohn’s Colitis 2022, 16, 369–378. [Google Scholar] [CrossRef]
  130. Crothers, J.W.; Chu, N.D.; Nguyen, L.T.T.; Phillips, M.; Collins, C.; Fortner, K.; Del Rio-Guerra, R.; Lavoie, B.; Callas, P.; Velez, M.; et al. Daily, oral FMT for long-term maintenance therapy in ulcerative colitis: Results of a single-center, prospective, randomized pilot study. BMC Gastroenterol. 2021, 21, 281. [Google Scholar] [CrossRef]
  131. Costello, S.P.; Hughes, P.A.; Waters, O.; Bryant, R.V.; Vincent, A.D.; Blatchford, P.; Katsikeros, R.; Makanyanga, J.; Campaniello, M.A.; Mavrangelos, C.; et al. Effect of Fecal Microbiota Transplantation on 8-Week Remission in Patients with Ulcerative Colitis: A Randomized Clinical Trial. JAMA 2019, 321, 156–164. [Google Scholar] [CrossRef] [PubMed]
  132. Paramsothy, S.; Kamm, M.A.; Kaakoush, N.O.; Walsh, A.J.; van den Bogaerde, J.; Samuel, D.; Leong, R.W.L.; Connor, S.; Ng, W.; Paramsothy, R.; et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet 2017, 389, 1218–1228. [Google Scholar] [CrossRef] [PubMed]
  133. Moayyedi, P.; Surette, M.G.; Kim, P.T.; Libertucci, J.; Wolfe, M.; Onischi, C.; Armstrong, D.; Marshall, J.K.; Kassam, Z.; Reinisch, W.; et al. Fecal Microbiota Transplantation Induces Remission in Patients with Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology 2015, 149, 102–109.e6. [Google Scholar] [CrossRef] [PubMed]
  134. Rossen, N.G.; Fuentes, S.; van der Spek, M.J.; Tijssen, J.G.; Hartman, J.H.; Duflou, A.; Löwenberg, M.; van den Brink, G.R.; Mathus-Vliegen, E.M.; de Vos, W.M.; et al. Findings from a Randomized Controlled Trial of Fecal Transplantation for Patients with Ulcerative Colitis. Gastroenterology 2015, 149, 110–118.e4. [Google Scholar] [CrossRef]
  135. Lopetuso, L.R.; Laterza, L.; Petito, V.; Pecere, S.; Quaranta, G.; Del Chierico, F.; Puca, P.; Schiavoni, E.; Napolitano, D.; Poscia, A.; et al. Serial Fecal Microbiota Infusions via Colonoscopy for Active Ulcerative Colitis: A Feasibility, Safety, and Translational Monocentric Italian Study. Microorganisms 2023, 11, 2536. [Google Scholar] [CrossRef]
  136. Feng, J.; Chen, Y.; Liu, Y.; Lin, L.; Lin, X.; Gong, W.; Xia, R.; He, J.; Sheng, J.; Cai, H.; et al. Efficacy and safety of fecal microbiota transplantation in the treatment of ulcerative colitis: A systematic review and meta-analysis. Sci. Rep. 2023, 13, 14494. [Google Scholar] [CrossRef]
  137. Zhang, J.T.; Zhang, N.; Dong, X.T.; Wang, X.R.; Ma, H.W.; Liu, Y.D.; Li, M.R. Efficacy and safety of fecal microbiota transplantation for treatment of ulcerative colitis: A post-consensus systematic review and meta-analysis. World J. Clin. Cases 2024, 12, 4691–4702. [Google Scholar] [CrossRef]
  138. Xiang, L.; Ding, X.; Li, Q.; Wu, X.; Dai, M.; Long, C.; He, Z.; Cui, B.; Zhang, F. Efficacy of faecal microbiota transplantation in Crohn’s disease: A new target treatment? Microb. Biotechnol. 2020, 13, 760–769. [Google Scholar] [CrossRef]
  139. 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. [Google Scholar] [CrossRef]
  140. Zhou, S.; Cui, Y.; Zhang, Y.; Zhao, T.; Cong, J. Fecal microbiota transplantation for induction of remission in Crohn’s disease: A systematic review and meta-analysis. Int. J. Color. Dis. 2023, 38, 62. [Google Scholar] [CrossRef]
  141. Fehily, S.R.; Basnayake, C.; Wright, E.K.; Kamm, M.A. Fecal microbiota transplantation therapy in Crohn’s disease: Systematic review. J. Gastroenterol. Hepatol. 2021, 36, 2672–2686. [Google Scholar] [CrossRef] [PubMed]
  142. Kahan, T.; Chandan, S.; Khan, S.R.; Deliwala, S.; Chang, S.; Axelrad, J.; Shaukat, A. Safety and Efficacy of Fecal Microbiota Transplant in Chronic Pouchitis—A Systematic Review with Meta-Analysis. Gastro Hep. Adv. 2023, 2, 843–852. [Google Scholar] [CrossRef] [PubMed]
  143. Zaman, S.; Akingboye, A.; Mohamedahmed, A.Y.Y.; Peterknecht, E.; Bhattacharya, P.; El-Asrag, M.E.; Iqbal, T.H.; Quraishi, M.N.; Beggs, A.D. Faecal Microbiota Transplantation [FMT] in the Treatment of Chronic Refractory Pouchitis: A Systematic Review and Meta-analysis. J. Crohn’s Colitis 2024, 18, 144–161. [Google Scholar] [CrossRef] [PubMed]
  144. Ezzatpour, S.; Mondragon Portocarrero, A.D.C.; Cardelle-Cobas, A.; Lamas, A.; López-Santamarina, A.; Miranda, J.M.; Aguilar, H.C. The Human Gut Virome and Its Relationship with Nontransmissible Chronic Diseases. Nutrients 2023, 15, 977. [Google Scholar] [CrossRef]
  145. Ng, R.W.; Dharmaratne, P.; Wong, S.; Hawkey, P.; Chan, P.; Ip, M. Revisiting the donor screening protocol of faecal microbiota transplantation (FMT): A systematic review. Gut 2024, 73, 1029–1031. [Google Scholar] [CrossRef]
  146. Cammarota, G.; Ianiro, G.; Kelly, C.R.; Mullish, B.H.; Allegretti, J.R.; Kassam, Z.; Putignani, L.; Fischer, M.; Keller, J.J.; Costello, S.P. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut 2019, 68, 2111–2121. [Google Scholar] [CrossRef]
  147. Bénard, M.V.; de Bruijn, C.M.A.; Fenneman, A.C.; Wortelboer, K.; Zeevenhoven, J.; Rethans, B.; Herrema, H.J.; van Gool, T.; Nieuwdorp, M.; Benninga, M.A.; et al. Challenges and costs of donor screening for fecal microbiota transplantations. PLoS ONE 2022, 17, e0276323. [Google Scholar] [CrossRef]
  148. Breyner, N.M.; Michon, C.; de Sousa, C.S.; Vilas Boas, P.B.; Chain, F.; Azevedo, V.A.; Langella, P.; Chatel, J.M. Microbial anti-inflammatory molecule (MAM) from Faecalibacterium prausnitzii shows a protective effect on DNBS and DSS-induced colitis model in mice through inhibition of NF-κB pathway. Front. Microbiol. 2017, 8, 114. [Google Scholar] [CrossRef]
  149. Olsson, L.M.; Boulund, F.; Nilsson, S.; Khan, M.T.; Gummesson, A.; Fagerberg, L.; Engstrand, L.; Perkins, R.; Uhlén, M.; Bergström, G.; et al. Dynamics of the normal gut microbiota: A longitudinal one-year population study in Sweden. Cell Host Microbe 2022, 30, 726–739.e3. [Google Scholar] [CrossRef]
  150. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef]
  151. Quévrain, E.; Maubert, M.A.; Michon, C.; Chain, F.; Marquant, R.; Tailhades, J.; Miquel, S.; Carlier, L.; Bermúdez-Humarán, L.G.; Pigneur, B.; et al. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s disease. Gut 2016, 65, 415–425. [Google Scholar] [CrossRef] [PubMed]
  152. Laval, L.; Martin, R.; Natividad, J.N.; Chain, F.; Miquel, S.; Desclée de Maredsous, C.; Capronnier, S.; Sokol, H.; Verdu, E.F.; van Hylckama Vlieg, J.E.; et al. Lactobacillus rhamnosus CNCM I-3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrier hyper-permeability in mice. Gut Microbes 2015, 6, 1–9. [Google Scholar] [CrossRef]
  153. Miquel, S.; Martín, R.; Rossi, O.; Bermúdez-Humarán, L.G.; Chatel, J.M.; Sokol, H.; Thomas, M.; Wells, J.M.; Langella, P. Faecalibacterium prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013, 16, 255–261. [Google Scholar] [CrossRef] [PubMed]
  154. Chen, J.; Vitetta, L. The Role of Butyrate in Attenuating Pathobiont-Induced Hyperinflammation. Immune Netw. 2020, 20, e15. [Google Scholar] [CrossRef]
  155. Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef]
  156. Machiels, K.; Joossens, M.; Sabino, J.; De Preter, V.; Arijs, I.; Eeckhaut, V.; Ballet, V.; Claes, K.; Van Immerseel, F.; Verbeke, K.; et al. A decrease of the butyrate-producing species roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 2014, 63, 1275–1283. [Google Scholar] [CrossRef]
  157. 116 Martín, R.; Chain, F.; Miquel, S.; Lu, J.; Gratadoux, J.J.; Sokol, H.; Verdu, E.F.; Bercik, P.; Bermúdez-Humarán, L.G.; Langella, P. The commensal bacterium Faecalibacterium prausnitzii is protective in DNBS-induced chronic moderate and severe colitis models. Inflamm. Bowel Dis. 2014, 20, 417–430. [Google Scholar] [CrossRef]
  158. Jian, H.; Liu, Y.; Wang, X.; Dong, X.; Zou, X. Akkermansia muciniphila as a Next-Generation Probiotic in Modulating Human Metabolic Homeostasis and Disease Progression: A Role Mediated by Gut–Liver–Brain Axes? Int. J. Mol. Sci. 2023, 24, 3900. [Google Scholar] [CrossRef]
  159. Pellegrino, A.; Coppola, G.; Santopaolo, F.; Gasbarrini, A.; Ponziani, F.R. Role of Akkermansia in Human Diseases: From Causation to Therapeutic Properties. Nutrients 2023, 15, 1815. [Google Scholar] [CrossRef]
  160. Shin, N.R.; Lee, J.C.; Lee, H.Y.; Kim, M.S.; Whon, T.W.; Lee, M.S.; Bae, J.W. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 2014, 63, 727–735. [Google Scholar] [CrossRef]
  161. Dunn, K.A.; Moore-Connors, J.; MacIntyre, B.; Stadnyk, A.W.; Thomas, N.A.; Noble, A.; Mahdi, G.; Rashid, M.; Otley, A.R.; Bielawski, J.P.; et al. Early changes in microbial community structure are associated with sustained remission after nutritional treatment of pediatric Crohn’s disease. Inflamm. Bowel Dis. 2016, 22, 2853–2862. [Google Scholar] [CrossRef]
  162. Grander, C.; Adolph, T.E.; Wieser, V.; Lowe, P.; Wrzosek, L.; Gyongyosi, B.; Ward, D.V.; Grabherr, F.; Gerner, R.R.; Pfister, A.; et al. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut 2018, 67, 891–901. [Google Scholar] [CrossRef] [PubMed]
  163. Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef]
  164. Hänninen, A.; Toivonen, R.; Pöysti, S.; Belzer, C.; Plovier, H.; Ouwerkerk, J.P.; Emani, R.; Cani, P.D.; De Vos, W.M. Akkermansia muciniphila induces gut microbiota remodelling and controls islet autoimmunity in NOD mice. Gut 2018, 67, 1445–1453. [Google Scholar] [CrossRef]
  165. Zheng, M.; Han, R.; Yuan, Y.; Xing, Y.; Zhang, W.; Sun, Z.; Liu, Y.; Li, J.; Mao, T. The role of Akkermansia muciniphila in inflammatory bowel disease: Current knowledge and perspectives. Inflamm. Bowel Dis. 2023, 13, 1089600. [Google Scholar] [CrossRef] [PubMed]
  166. Png, C.W.; Lindén, S.K.; Gilshenan, K.S.; Zoetendal, E.G.; McSweeney, C.S.; Sly, L.I.; McGuckin, M.A.; Florin, T.H. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am. J. Gastroenterol. 2010, 105, 2420–2428. [Google Scholar] [CrossRef]
  167. Kump, P.; Wurm, P.; Gröchenig, H.P.; Wenzl, H.; Petritsch, W.; Halwachs, B.; Wagner, M.; Stadlbauer, V.; Eherer, A.; Hoffmann, K.M.; et al. The taxonomic composition of the donor intestinal microbiota is a major factor influencing the efficacy of faecal microbiota transplantation in therapy refractory ulcerative colitis. Aliment. Pharmacol. Ther. 2018, 47, 67–77. [Google Scholar] [CrossRef]
  168. 127Earley, H.; Lennon, G.; Balfe, Á.; Coffey, J.C.; Winter, D.C.; O’Connell, P.R. The abundance of Akkermansia muciniphila and its relationship with sulphated colonic mucins in health and ulcerative colitis. Sci. Rep. 2019, 9, 15683. [Google Scholar] [CrossRef]
  169. Vigsnæs, L.K.; Brynskov, J.; Steenholdt, C.; Wilcks, A.; Licht, T.R. Gram-negative bacteria account for main differences between faecal microbiota from patients with ulcerative colitis and healthy controls. Benef. Microbes 2012, 3, 287–297. [Google Scholar] [CrossRef]
  170. Wang, L.; Tang, L.; Feng, Y.; Zhao, S.; Han, M.; Zhang, C.; Yuan, G.; Zhu, J.; Cao, S.; Wu, Q.; 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, 1988–1997. [Google Scholar] [CrossRef]
  171. Maria, H.S. Safety aspects of next generation probiotics. Curr. Opin. Food Sci. 2019, 30, 8–13. [Google Scholar] [CrossRef]
  172. Merenstein, D.; Pot, B.; Leyer, G.; Ouwehand, A.C.; Preidis, G.A.; Elkins, C.A.; Hill, C.; Lewis, Z.T.; Shane, A.L.; Zmora, N.; et al. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes 2023, 15, 2185034. [Google Scholar] [CrossRef] [PubMed]
  173. D’Agostin, M.; Squillaci, D.; Lazzerini, M.; Barbi, E.; Wijers, L.; Da Lozzo, P. Invasive Infections Associated with the Use of Probiotics in Children: A Systematic Review. Children 2021, 8, 924. [Google Scholar] [CrossRef]
  174. Tiwari, A.; Krisnawati, D.I.; Susilowati, E.; Mutalik, C.; Kuo, T.R. Next-Generation Probiotics and Chronic Diseases: A Review of Current Research and Future Direction. J. Agric. Food Chem. 2024, 72, 27679–27700. [Google Scholar] [CrossRef]
  175. Turck, D.; Bresson, J.L.; Burlingame, B.; Dean, T.; Fairweather-Tait, S.; Heinonen, M.; Hirsch-Ernst, K.I.; Mangelsdorf, I.; McArdle, H.; Naska, A.; et al. Guidance on the preparation and submission of an application for authorisation of a novel food in the context of Regulation (EU) 2015/22831. EFSA J. 2016, 19, e06555. [Google Scholar] [CrossRef]
  176. Zhang, J.; Huang, Y.J.; Yoon, J.Y.; Kemmitt, J.; Wright, C.; Schneider, K.; Sphabmixay, P.; Hernandez-Gordillo, V.; Holcomb, S.J.; Bhushan, B.; et al. Primary human colonic mucosal barrier crosstalk with super oxygen-sensitive Faecalibacterium prausnitzii in continuous culture. Med 2021, 2, 74–98.e9. [Google Scholar] [CrossRef]
  177. Bag, S.; Ghosh, T.S.; Das, B. Complete genome sequence of Faecalibacterium prausnitzii isolated from the gut of a healthy Indian adult. Genome Announc. 2017, 5, 10–1128. [Google Scholar] [CrossRef]
  178. Machado, D.; Barbosa, J.C.; Domingos, M.; Almeida, D.; Andrade, J.C.; Freitas, A.C.; Gomes, A.M. Revealing antimicrobial resistance profile of the novel probiotic candidate Faecalibacterium prausnitzii DSM 17677. Int. J. Food Microbiol. 2022, 363, 109501. [Google Scholar] [CrossRef]
  179. Hu, W.; Gao, W.; Liu, Z.; Fang, Z.; Zhao, J.; Zhang, H.; Lu, W.; Chen, W. Biodiversity and Physiological Characteristics of Novel Faecalibacterium prausnitzii Strains Isolated from Human Feces. Microorganisms 2022, 10, 297. [Google Scholar] [CrossRef]
  180. Li, Z.; Zhu, L.; Hu, G.; Sun, Z.; Zhan, X.; Gao, M. Akkermansia muciniphila fermentation culture based on a novel bionic large intestine dynamic digestion model. Food Biosci. 2021, 43, 101260. [Google Scholar] [CrossRef]
  181. Dubourg, G.; Lagier, J.C.; Armougom, F.; Robert, C.; Audoly, G.; Papazian, L.; Raoult, D. High-level colonisation of the human gut by Verrucomicrobia following broad-spectrum antibiotic treatment. Int. J. Antimicrob. Agents 2013, 41, 149–155. [Google Scholar] [CrossRef] [PubMed]
  182. Machado, D.; Barbosa, J.C.; Almeida, D.; Andrade, J.C.; Freitas, A.C.; Gomes, A.M. Insights into the antimicrobial resistance profile of a next generation probiotic Akkermansia muciniphila DSM 22959. Int. J. Environ. Res. Public Health 2022, 19, 9152. [Google Scholar] [CrossRef]
  183. Vallianou, N.G.; Kounatidis, D.; Tsilingiris, D.; Panagopoulos, F.; Christodoulatos, G.S.; Evangelopoulos, A.; Karampela, I.; Dalamaga, M. The role of next-generation probiotics in obesity and obesity-Associated disorders: Current knowledge and future perspectives. Int. J. Mol. Sci. 2023, 24, 6755. [Google Scholar] [CrossRef]
  184. O’Toole, P.W.; Marchesi, J.R.; Hill, C. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol. 2017, 2, 17057. [Google Scholar] [CrossRef]
  185. Zhang, S.; Merino, N.; Okamoto, A.; Gedalanga, P. Interkingdom microbial consortia mechanisms to guide biotechnological applications. Microb. Biotechnol. 2018, 11, 833–847. [Google Scholar] [CrossRef] [PubMed]
  186. Walana, W.; Ye, Y.; Li, M.; Wang, J.; Wang, B.; Cheng, J.W.; Gordon, J.R.; Li, F. IL-8 antagonist, CXCL8(3-72)K11R/G31P coupled with probiotic exhibit variably enhanced therapeutic potential in ameliorating ulcerative colitis. Biomed. Pharmacother. 2018, 103, 253–261. [Google Scholar] [CrossRef] [PubMed]
  187. Zhou, C.; Wang, Y.; Li, C.; Xie, Z.; Dai, L. Amelioration of Colitis by a Gut Bacterial Consortium Producing Anti-Inflammatory Secondary Bile Acids. Microbiol. Spectr. 2023, 11, e0333022. [Google Scholar] [CrossRef]
  188. Xu, L.; Liu, B.; Huang, L.; Li, Z.; Cheng, Y.; Tian, Y.; Pan, G.; Li, H.; Xu, Y.; Wu, W.; et al. Probiotic Consortia and Their Metabolites Ameliorate the Symptoms of Inflammatory Bowel Diseases in a Colitis Mouse Model. Microbiol. Spectr. 2022, 10, e0065722. [Google Scholar] [CrossRef]
  189. van der Lelie, D.; Oka, A.; Taghavi, S.; Umeno, J.; Fan, T.J.; Merrell, K.E.; Watson, S.D.; Ouellette, L.; Liu, B.; Awoniyi, M.; et al. Rationally designed bacterial consortia to treat chronic immune-mediated colitis and restore intestinal homeostasis. Nat. Commun. 2021, 12, 3105. [Google Scholar] [CrossRef]
  190. Polonsky, O.; Meshner, S.; Eshar, S.; Ben-Shabat, S.K.; Tirosh, O.; Haber, E.; Ringel, Y. BMC322- a rationally-designed live bacterial consortium based on microbiome functional genomic analysis for treatment of ibd. Inflamm. Bowel Dis. 2021, 27, S38. [Google Scholar] [CrossRef]
  191. Guthrie, L.; Kelly, L. Bringing microbiome-drug interaction research into the clinic. EBioMedicine 2019, 44, 708–715. [Google Scholar] [CrossRef] [PubMed]
  192. Lopetuso, L.R.; Deleu, S.; Puca, P.; Abreu, M.T.; Armuzzi, A.; Barbara, G.; Caprioli, F.; Chieng, S.; Costello, S.P.; Damiani, A.; et al. Guidance for Fecal Microbiota Transplantation Trials in Ulcerative Colitis: The Second ROME Consensus Conference. Inflamm. Bowel Dis. 2025, izaf013, advance online publication. [Google Scholar] [CrossRef] [PubMed]
  193. Nobrega, F.L.; Vlot, M.; de Jonge, P.A.; Dreesens, L.L.; Beaumont, H.J.E.; Lavigne, R.; Dutilh, B.E.; Brouns, S.J.J. Targeting mechanisms of tailed bacteriophages. Nat. Rev. Microbiol. 2018, 16, 760–773. [Google Scholar] [CrossRef] [PubMed]
  194. Balcha, F.B.; Neja, S.A. CRISPR-Cas9 mediated phage therapy as an alternative to antibiotics. Anim. Dis. 2023, 3, 4. [Google Scholar] [CrossRef]
  195. Hodyra-Stefaniak, K.; Miernikiewicz, P.; Drapała, J.; Drab, M.; Jończyk-Matysiak, E.; Lecion, D.; Kaźmierczak, Z.; Beta, W.; Majewska, J.; Harhala, M.; et al. Mammalian Host-Versus-Phage immune response determines phage fate in vivo. Sci. Rep. 2015, 5, 14802. [Google Scholar] [CrossRef]
  196. Park, H.; Laffin, M.R.; Jovel, J.; Millan, B.; Hyun, J.E.; Hotte, N.; Kao, D.; Madsen, K.L. The success of fecal microbial transplantation in Clostridium difficile infection correlates with bacteriophage relative abundance in the donor: A retrospective cohort study. Gut Microbes 2019, 10, 676–687. [Google Scholar] [CrossRef]
  197. Machuca, P.; Daille, L.; Vinés, E.; Berrocal, L.; Bittner, M. Isolation of a novel bacteriophage specific for the periodontal pathogen Fusobacterium nucleatum. Appl. Environ. Microbiol. 2010, 76, 7243–7250. [Google Scholar] [CrossRef]
  198. 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. [Google Scholar] [CrossRef]
  199. Federici, S.; Kredo-Russo, S.; Valdés-Mas, R.; Kviatcovsky, D.; Weinstock, E.; Matiuhin, Y.; Silberberg, Y.; Atarashi, K.; Furuichi, M.; Oka, A.; et al. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell 2022, 185, 2879–2898.e24. [Google Scholar] [CrossRef]
  200. Speck, P.; Smithyman, A. Safety and efficacy of phage therapy via the intravenous route. FEMS Microbiol. Lett. 2016, 363, fnv242. [Google Scholar] [CrossRef]
  201. Malik, D.J.; Sokolov, I.J.; Vinner, G.K.; Mancuso, F.; Cinquerrui, S.; Vladisavljevic, G.T.; Clokie, M.R.J.; Garton, N.J.; Stapley, A.G.F.; Kirpichnikova, A. Formulation, stabilisation and encapsulation of bacteriophage for phage therapy. Adv. Colloid Interface Sci. 2017, 249, 100–133. [Google Scholar] [CrossRef]
  202. Hyman, P.; Abedon, S.T. Bacteriophage host range and bacterial resistance. Adv. Appl. Microbiol. 2010, 70, 217–248. [Google Scholar] [CrossRef] [PubMed]
  203. Gill, J.J.; Hyman, P. Phage choice, isolation, and preparation for phage therapy. Curr. Pharm. Biotechnol. 2010, 11, 2–14. [Google Scholar] [CrossRef] [PubMed]
  204. Carascal, M.B.; Dela Cruz-Papa, D.M.; Remenyi, R.; Cruz, M.C.B.; Destura, R.V. Phage Revolution Against Multidrug-Resistant Clinical Pathogens in Southeast Asia. Front. Microbiol. 2022, 13, 820572. [Google Scholar] [CrossRef] [PubMed]
  205. Tsonos, J.; Vandenheuvel, D.; Briers, Y.; De Greve, H.; Hernalsteens, J.P.; Lavigne, R. Hurdles in bacteriophage therapy: Deconstructing the parameters. Vet. Microbiol. 2014, 171, 460–469. [Google Scholar] [CrossRef]
  206. Lin, J.; Du, F.; Long, M.; Li, P. Limitations of Phage Therapy and Corresponding Optimization Strategies: A Review. Molecules 2022, 27, 1857. [Google Scholar] [CrossRef]
  207. Federici, S.; Kviatcovsky, D.; Valdés-Mas, R.; Elinav, E. Microbiome-phage interactions in inflammatory bowel disease. Clin. Microbiol. Infect. 2023, 29, 682–688. [Google Scholar] [CrossRef]
  208. Tetz, G.V.; Ruggles, K.V.; Zhou, H.; Heguy, A.; Tsirigos, A.; Tetz, V. Bacteriophages as potential new mammalian pathogens. Sci. Rep. 2017, 7, 7043. [Google Scholar] [CrossRef]
  209. Champagne-Jorgensen, K.; Luong, T.; Darby, T.; Roach, D.R. Immunogenicity of bacteriophages. Trends Microbiol. 2023, 31, 1058–1071. [Google Scholar] [CrossRef]
  210. Pesce, M.; Seguella, L.; Del Re, A.; Lu, J.; Palenca, I.; Corpetti, C.; Rurgo, S.; Sanseverino, W.; Sarnelli, G.; Esposito, G. Next-Generation Probiotics for Inflammatory Bowel Disease. Int. J. Mol. Sci. 2022, 23, 5466. [Google Scholar] [CrossRef]
  211. Aggarwal, N.; Breedon, A.M.E.; Davis, C.M.; Hwang, I.Y.; Chang, M.W. Engineering probiotics for therapeutic applications: Recent examples and translational outlook. Curr. Opin. Biotechnol. 2020, 65, 171–179. [Google Scholar] [CrossRef]
  212. Kan, A.; Gelfat, I.; Emani, S.; Praveschotinunt, P.; Joshi, N.S. Plasmid Vectors for in Vivo Selection-Free Use with the Probiotic E. coli Nissle 1917. ACS Synth. Biol. 2021, 10, 94–106. [Google Scholar] [CrossRef]
  213. Zhou, S.; Zhao, L.; Zuo, W.; Zheng, Y.; Zhang, P.; Sun, Y.; Wang, Y.; Du, G.; Kang, Z. Minimizing endogenous cryptic plasmids to construct antibiotic-free expression systems for Escherichia coli Nissle 1917. Synth. Syst. Biotechnol. 2024, 9, 165–175. [Google Scholar] [CrossRef]
  214. Cook, D.P.; Gysemans, C.; Mathieu, C. Lactococcus lactis As a Versatile Vehicle for Tolerogenic Immunotherapy. Front. Immunol. 2018, 8, 1961. [Google Scholar] [CrossRef] [PubMed]
  215. Berlec, A.; Škrlec, K.; Kocjan, J.; Olenic, M.; Štrukelj, B. Single plasmid systems for inducible dual protein expression and for CRISPR-Cas9/CRISPRi gene regulation in lactic acid bacterium Lactococcus lactis. Sci. Rep. 2018, 8, 1009. [Google Scholar] [CrossRef] [PubMed]
  216. Wang, L.; Liao, Y.; Yang, R.; Zhu, Z.; Zhang, L.; Wu, Z.; Sun, X. An engineered probiotic secreting Sj16 ameliorates colitis via Ruminococcaceae/butyrate/retinoic acid axis. Bioeng. Transl. Med. 2021, 6, e10219. [Google Scholar] [CrossRef]
  217. Cui, M.; Pang, G.; Zhang, T.; Sun, T.; Zhang, L.; Kang, R.; Xue, X.; Pan, H.; Yang, C.; Zhang, X.; et al. Optotheranostic Nanosystem with Phone Visual Diagnosis and Optogenetic Microbial Therapy for Ulcerative Colitis At-Home Care. ACS Nano 2021, 15, 7040–7052. [Google Scholar] [CrossRef]
  218. Esposito, G.; Pesce, M.; Seguella, L.; Lu, J.; Corpetti, C.; Del Re, A.; De Palma, F.D.E.; Esposito, G.; Sanseverino, W.; Sarnelli, G. Engineered Lactobacillus paracasei Producing Palmitoylethanolamide (PEA) Prevents Colitis in Mice. Int. J. Mol. Sci. 2021, 22, 2945. [Google Scholar] [CrossRef]
  219. Esposito, G.; Capoccia, E.; Turco, F.; Palumbo, I.; Lu, J.; Steardo, A.; Cuomo, R.; Sarnelli, G.; Steardo, L. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation. Gut 2014, 63, 1300–1312. [Google Scholar] [CrossRef]
  220. Kim, W.K.; Jang, Y.J.; Seo, B.; Han, D.H.; Park, S.; Ko, G.P. Administration of Lactobacillus paracasei strains improves immunomodulation and changes the composition of gut microbiota leading to improvement of colitis in mice. J. Funct. Foods 2019, 5, 565–575. [Google Scholar] [CrossRef]
  221. Andriantsoanirina, V.; Allano, S.; Butel, M.J.; Aires, J. Tolerance of Bifidobacterium human isolates to bile, acid and oxygen. Anaerobe 2013, 21, 39–42. [Google Scholar] [CrossRef]
  222. Liu, M.; Li, S.; Zhang, Q.; Xu, Z.; Wang, J.; Sun, H. Oral engineered Bifidobacterium longum expressing rhMnSOD to suppress experimental colitis. Int. Immunopharmacol. 2018, 57, 25–32. [Google Scholar] [CrossRef]
  223. Wei, X.; Yan, X.; Chen, X.; Yang, Z.; Li, H.; Zou, D.; He, X.; Wang, S.; Cui, Q.; Liu, W.; et al. Proteomic analysis of the interaction of Bifidobacterium longum NCC2705 with the intestine cells Caco-2 and identification of plasminogen receptors. J. Proteom. 2014, 108, 89–98. [Google Scholar] [CrossRef] [PubMed]
  224. Avila-Calderón, E.D.; Ruiz-Palma, M.D.S.; Aguilera-Arreola, M.G.; Velázquez-Guadarrama, N.; Ruiz, E.A.; Gomez-Lunar, Z.; Witonsky, S.; Contreras-Rodríguez, A. Outer Membrane Vesicles of Gram-Negative Bacteria: An Outlook on Biogenesis. Front. Microbiol. 2021, 12, 557902. [Google Scholar] [CrossRef] [PubMed]
  225. Carvalho, A.L.; Fonseca, S.; Miquel-Clopés, A.; Cross, K.; Kok, K.S.; Wegmann, U.; Gil-Cordoso, K.; Bentley, E.G.; Al Katy, S.H.M.; Coombes, J.L.; et al. Bioengineering commensal bacteria-derived outer membrane vesicles for delivery of biologics to the gastrointestinal and respiratory tract. J. Extracell. Vesicles 2019, 8, 1632100. [Google Scholar] [CrossRef]
  226. Fábrega, M.J.; Rodríguez-Nogales, A.; Garrido-Mesa, J.; Algieri, F.; Badía, J.; Giménez, R.; Gálvez, J.; Baldomà, L. Intestinal anti-inflammatory effects of outer membrane vesicles from Escherichia coli Nissle 1917 in DSS-experimental colitis in mice. Front. Microbiol. 2017, 8, 1274. [Google Scholar] [CrossRef]
  227. Zhang, B.; Liu, Y.; Lan, X.; Xu, X.; Zhang, X.; Li, X.; Zhao, Y.; Li, G.; Du, C.; Lu, S.; et al. Oral Escherichia coli expressing IL-35 meliorates experimental colitis in mice. J. Transl. Med. 2018, 16, 71. [Google Scholar] [CrossRef]
  228. Hanson, M.L.; Hixon, J.A.; Li, W.; Felber, B.K.; Anver, M.R.; Stewart, C.A.; Janelsins, B.M.; Datta, S.K.; Shen, W.; McLean, M.H.; et al. Oral delivery of il-27 recombinant bacteria attenuates immune colitis in mice. Gastroenterology 2014, 146, 210–221.e13. [Google Scholar] [CrossRef]
  229. Xia, J.Y.; Hepler, C.; Tran, P.; Waldeck, N.J.; Bass, J.; Prindle, A. Engineered calprotectin-sensing probiotics for IBD surveillance in humans. Proc. Natl. Acad. Sci. USA 2023, 120, e2221121120. [Google Scholar] [CrossRef]
  230. Nguyen, T.L.; Vieira-Silva, S.; Liston, A.; Raes, J. How informative is the mouse for human gut microbiota research? Dis. Model. Mech. 2015, 8, 1–16. [Google Scholar] [CrossRef]
  231. Murali, S.K.; Mansell, T.J. Next generation probiotics: Engineering live biotherapeutics. Biotechnol. Adv. 2024, 72, 108336. [Google Scholar] [CrossRef]
  232. Gulliver, E.L.; Young, R.B.; Chonwerawong, M.; D’Adamo, G.L.; Thomason, T.; Widdop, J.T.; Rutten, E.L.; Rossetto Marcelino, V.; Bryant, R.V.; Costello, S.P.; et al. Review article: The future of microbiome-based therapeutics. Aliment. Pharmacol. Ther. 2022, 56, 192–208. [Google Scholar] [CrossRef] [PubMed]
  233. Pols, T.; Sikkema, H.R.; Gaastra, B.F.; Frallicciardi, J.; Śmigiel, W.M.; Singh, S.; Poolman, B. A synthetic metabolic network for physicochemical homeostasis. Nat. Commun. 2019, 10, 4239. [Google Scholar] [CrossRef] [PubMed]
  234. Schiering, C.; Wincent, E.; Metidji, A.; Iseppon, A.; Li, Y.; Potocnik, A.J.; Omenetti, S.; Henderson, C.J.; Wolf, C.R.; Nebert, D.W.; et al. Feedback control of AHR signalling regulates intestinal immunity. Nature 2017, 542, 242–245. [Google Scholar] [CrossRef]
  235. McAleer, J.P.; Fan, J.; Roar, B.; Primerano, D.A.; Denvir, J. Cytokine Regulation in Human CD4 T Cells by the Aryl Hydrocarbon Receptor and Gq-Coupled Receptors. Sci. Rep. 2018, 8, 10954. [Google Scholar] [CrossRef]
  236. Baricza, E.; Tamási, V.; Marton, N.; Buzás, E.I.; Nagy, G. The emerging role of aryl hydrocarbon receptor in the activation and differentiation of Th17 cells. Cell. Mol. Life Sci. 2016, 73, 95–117. [Google Scholar] [CrossRef]
  237. Cervenka, I.; Agudelo, L.Z.; Ruas, J.L. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science 2017, 357, eaaf9794. [Google Scholar] [CrossRef]
  238. Nikolaus, S.; Schulte, B.; Al-Massad, N.; Thieme, F.; Schulte, D.M.; Bethge, J.; Rehman, A.; Tran, F.; Aden, K.; Häsler, R.; et al. Increased Tryptophan Metabolism Is Associated With Activity of Inflammatory Bowel Diseases. Gastroenterology 2017, 153, 1504–1516.e2. [Google Scholar] [CrossRef]
  239. Liu, J.Z.; van Sommeren, S.; Huang, H.; Ng, S.C.; Alberts, R.; Takahashi, A.; Ripke, S.; Lee, J.C.; Jostins, L.; Shah, T. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015, 47, 979–986. [Google Scholar] [CrossRef]
  240. Alharbi, R.S.; Shaik, N.A.; Almahdi, H.; ElSokary, H.A.; Jamalalail, B.A.; Mosli, M.H.; Alsufyani, H.A.; Al-Aama, J.Y.; Elango, R.; Saadah, O.I.; et al. Genetic association study of NOD2 and IL23R amino acid substitution polymorphisms in Saudi Inflammatory Bowel Disease patients. J. King Saud Univ.-Sci. 2022, 34, 101726. [Google Scholar] [CrossRef]
  241. Michaudel, C.; Danne, C.; Agus, A.; Magniez, A.; Aucouturier, A.; Spatz, M.; Lefevre, A.; Kirchgesner, J.; Rolhion, N.; Wang, Y.; et al. Rewiring the altered tryptophan metabolism as a novel therapeutic strategy in inflammatory bowel diseases. Gut 2023, 72, 1296–1307. [Google Scholar] [CrossRef] [PubMed]
  242. Zhao, L.P.; Wu, J.; Quan, W.; Zhou, Y.; Hong, H.; Niu, G.Y.; Li, T.; Huang, S.B.; Qiao, C.M.; Zhao, W.J.; et al. DSS-induced colitis activates the kynurenine pathway in serum and brain by affecting IDO-1 and gut microbiota. Front. Immunol. 2023, 13, 1089200. [Google Scholar] [CrossRef]
  243. Yang, S.; Li, W.; Bai, X.; Di Nunzio, G.; Fan, L.; Zhao, Y.; Ren, L.; Zhao, R.; Bian, S.; Liu, M.; et al. Ginseng-derived nanoparticles alleviate inflammatory bowel disease via the TLR4/MAPK and p62/Nrf2/Keap1 pathways. J. Nanobiotechnol. 2024, 22, 48. [Google Scholar] [CrossRef] [PubMed]
  244. Zhang, X.; Yuan, Z.; Wu, J.; He, Y.; Lu, G.; Zhang, D.; Zhao, Y.; Wu, R.; Lv, Y.; Cai, K.; et al. An Orally-Administered Nanotherapeutics with Carbon Monoxide Supplying for Inflammatory Bowel Disease Therapy by Scavenging Oxidative Stress and Restoring Gut Immune Homeostasis. ACS Nano 2023, 17, 21116–21133. [Google Scholar] [CrossRef] [PubMed]
  245. Min, D.K.; Kim, Y.E.; Kim, M.K.; Choi, S.W.; Park, N.; Kim, J. Orally Administrated Inflamed Colon-Targeted Nanotherapeutics for Inflammatory Bowel Disease Treatment by Oxidative Stress Level Modulation in Colitis. ACS Nano 2023, 17, 24404–24416. [Google Scholar] [CrossRef]
  246. Barani, M.; Rahdar, A.; Sargazi, S.; Amiri, M.S.; Sharma, P.; Bhalla, N. Nanotechnology for inflammatory bowel disease management: Detection, imaging and treatment. Sens. Bio-Sens. Res. 2021, 32, 100417. [Google Scholar] [CrossRef]
Figure 1. Microbial biotherapeutics as new therapeutic tools in gut microbiota modulation. The effectiveness of most new microbiota modulation therapies has been demonstrated in preclinical models and the early stages of disease in human models. Fecal microbiota transplantation has also shown effectiveness in human models, both in the early and advanced stages of disease.
Figure 1. Microbial biotherapeutics as new therapeutic tools in gut microbiota modulation. The effectiveness of most new microbiota modulation therapies has been demonstrated in preclinical models and the early stages of disease in human models. Fecal microbiota transplantation has also shown effectiveness in human models, both in the early and advanced stages of disease.
Ijms 26 03059 g001
Table 1. Efficacy of probiotics in UC patients with active or quiescent colitis.
Table 1. Efficacy of probiotics in UC patients with active or quiescent colitis.
Probiotic TestedReferenceDisease
Activity
(UC vs. CD)
Trial DesignOutcomes
L. rhamnosus NCIMB 30174, L. plantarum NCIMB 30173, L. acidophilus NCIMB 30175 and E. faecium NCIMB 30176Bjarnason et al., 2019 [90]Quiescent UC and CDSingle center, randomized, double-blind, placebo-controlled trialNo significant differences in IBD-QoL scores;
reduced intestinal inflammation in UC patients
E. coli Nissle 1917Park et al., 2022 [91]Mild to moderate active UCMulticenter, double-blind, randomized, placebo-controlled studyPrevents exacerbations of IBD-QoL scores;
achieves clinical response and endoscopic remission
L. paracasei (A234), L. gasseri (A237), L. rhamnosus (A119), L. acidophilus (A118), L. plantarum (A138), L. casei (A179), L. reuteri (A113), L. lactis (A328), B. animalis subsp lactis (A026), B. breve (A055), B. longum subsp. longum (A027), B. bifidum (A058), B. longum subsp. infantis (A041) species.Agraib et al., 2022 [92]Mild to moderate UCRandomized, double-blind, placebo-controlled, parallel-arms, multicenter studyInduces clinical (partial Mayo score) and biochemical remission in UC patients
E. coli Nissle 1917Oh et al., 2021 [93]Quiescent UCUncontrolled, observational, retrospective studyAdditional administration of E. coli Nissle 1917 may improve UC symptoms
Kefir (L. pentosus, L. brevis, L. plantarum, L. fermentum, L. kefiri, and L. lindneri)Yilmaz et al., 2019 [94]Active UC and CDsingle-center, prospective, open-label randomized controlled trialKefir consumption may modulate gut microbiota and enhance short-term quality of life
B. breve strain Yakult, L. AcidophilusMatsuoka et al., 2018 [95]Quiescent UCMulticenter, randomized, placebo-controlled, double-blind parallel-group studyNo effect on relapse timing
S. faecalis T-110, C. butyricum TO-A, B. mesentericus TO-AYoshimatsu et al., 2015 [96]Quiescent UCRandomized, double-blind, placebo-controlled studySupports clinical remission mantainance
L. reuteri ATCC55730Oliva et al., 2012 [97]Mild to moderate active distal UC (children)Prospective randomized, placebo-controlled studyReduces Mayo score and histological scores;
modulates mucosal cytokine expression
S. thermophilus BT01, B. breve BB02, B. longum * BL03, B. infantis * BI04, L. acidophilus BA05, L. plantarum BP06, L. paracasei BP07, L. delbrueckii subsTursi et al., 2010 [98]Relapsing mild-moderate UCMulticenter, double-blind, randomized placebo-controlled, parallel studyDecreases UCDAI scores;
improves rectal bleeding;
may reinduce remission after 8 weeks of treatment
L. acidophilus, L. plantarum, L. casei, L. delbruecki subspecies bulgaricus, B. breve, B. longum, B. infantis, S. salivarius subspecies thermophilusMiele et al., 2009 [99]Active UC (children)Randomized, placebo-controlled, double-blind studyMaintains clinical and endoscopic remission
Lactobacillus GGZocco et al., 2006 [100]Quiescent UCSingle center, prospective, open-label randomized trialMaintains remission compared to mesalazine;
delays UC relapse
B. breve strain Yakult, B. bifidum strain Yakult, L. acidophillus strainKato et al., 2004 [101]Mild to moderate active UCrandomized placebo-controlled clinical trialReduces clinical activity index; significantly improves post-treatment endoscopic activity index and histological score
UC: ulcerative colitis. CD: Crohn’s disease. IBD-QoL: Inflammatory Bowel Disease Quality of Life Questionnaire. UCDAI: Ulcerative Colitis Disease Activity Index.
Table 2. Randomized controlled trials testing the role of FMT in UC.
Table 2. Randomized controlled trials testing the role of FMT in UC.
Way of AdministrationReferenceDisease
Activity
Trial DesignResults Summary
FMT by colonoscopyDeleu et al., 2024 [127]Moderate to severe UCMulti-centric, double-blind, sham-controlled randomized trialFailure to achieve steroid-free clinical remission at week 8
Lyophilized oral FMTHaifer. et al., 2022 [128]Mild to moderate UCDouble-blind, randomized, placebo-controlled trialInduction of clinical remission with endoscopic remission or
response at week 8;
maintenance of clinical, endoscopic, and histological remission at week 56
FMT by colonoscopy and enemasShabat et al., 2022 [129]Moderate to severe active UCSingle, blinded, randomized, controlled trialUC exclusion diet (UCED) leads to higher clinical remission and mucosal healing than single donor FMT, with or without diet
Encapsuled oral FMTCrothers et al., 2021 [130]Mild to moderate UCsingle center, double-blinded, placebo-controlled, randomized control trialProlonged durability of FMT-induced changes in gut bacterial community structure; association between MAIT cell cytokine production and clinical response
FMT by colonoscopy and enemasCostello et al., 2019 [131]Mild to moderate UCDouble blind, randomized, clinical trialA 1-week treatment with anaerobically prepared donor FMT results in higher remission rates at week 8 compared to autologous FMT
FMT by colonoscopy and enemasParamsothy et al., 2017 [132]Mild to moderate UCMulticenter, double-blind, randomized, placebo-controlled trialInduction of clinical remission and endoscopic improvement
FMT via enemaMoayyedi et al., 2015 [133]Mild to severe UCDouble-blind randomized controlled trialInduction of remission in a significantly higher percentage of patients with active UC; greater microbial diversity
FMT via nasoduodenal tubeRossen et al., 2015 [134]Mild to moderate UCSingle-center, double-blind, placebo-controlled, randomized, proof-of-concept phase 2 trialNo significant difference in clinical and endoscopic remission between patients receiving FMT from healthy donors and those receiving autologous FMT
UC: ulcerative colitis. FMT: fecal microbiota transplantation. MAIT: mucosal-associated invariant T cells.
Table 3. The role of bacterial consortia tested in animal colitis models.
Table 3. The role of bacterial consortia tested in animal colitis models.
Bacterial ConsortiaReferenceTrial DesignResults Summary
BAC (bile acid consortium, made up of Clostridium AP sp000509125, Bacteroides ovatus, and Eubacterium limosum)Zhou et al., 2023 [187]DSS-induced colitis mice modelIncreases secondary bile acids (Ursodeoxycholic acid (UDCA) and Lithocholic acid (LCA)) in vitro; exerts protective effects against colitis (reduces weight loss, increases colon length, strengthens the intestinal barrier)
Lactobacillus reuteri, Lactobacillus gasseri, Lactobacillus acidophilus (Lactobacillus spp.), and Bifidobacterium lactis (Bifidobacterium spp.)Xu et al., 2022 [188]DSS-induced colitis mice modelAlleviates disease phenotype; restores the composition and structure of the gut microbiota
GUT 103 (strains of genera Bacteroides, Akkermansia, Clostridium, Faecalibacterium) and GUT108 (strains of Clostridium, Intestinimonas, Bitterella, Barneseilla)Van der Leile et al., 2021 [189]Immune-mediated colitis in germ free miceGUT-103 and GUT-108:
Correct the dysbiotic microbiome environment; activate IL-10-producing immune cells; reduce inflammatory responses; restore bacterial metabolic profiles to levels observed in healthy individuals’ stool samples
BMC332Polonsky et al., 2021 [190]DSS-induced colitis in miceExhibits anti-inflammatory properties; maintains intestinal barrier integrity
DSS: dextran sulfate sodium. BAC: bile acid consortium. UDCA: ursodeoxycholic acid. LCA: lithocholic acid.
Table 4. Summary of recent studies on engineered probiotics.
Table 4. Summary of recent studies on engineered probiotics.
Engineered ProbioticReferenceTrial DesignResults Summary
E. coliWang et al., 2021 [216]DSS-induced colitis in mice
-
Improvement in colitis by the modulation of microbiota composition (protein Sj16)
E. coliCui et al., 2021 [217]DSS-treated colitis in mice
-
Suppression of intestinal inflammatory response (IL-10)
-
Protection of mucosa against injury
L. paracasei (KBL382,384,385)Kim et al., 2019 [220]DSS-induced colitis in mice
-
Strong protective effects; improved colitis symptoms
B. longumLiu et al., 2016 [222]DSS-induced colitis in mice
-
Reduction of inflammation (TNF-α, IL-1β, IL-6, and IL-8)
B. longumWei et al., 2016 [223]DSS-induced colitis in mice
-
Expression of α-MSH and significant anti-inflammatory effect
OMV of Bacteroides thetaiotaomicron (Bt)Carvalho et al., 2019 [225]DSS-induced colitis in mice
-
KGF-2 reduced macroscopic and microscopic inflammation
OMV from E. coli Nissle 1917Fabrega et al., 2017 [226]DSS-induced colitis in mice
-
Intestinal anti-inflammatory effects
E. coliZhang et al., 2018 [227]DSS-induced experimental colitis in mice
-
Reduction of inflammation (IL-35)
L. lactisHanson et al., 2014 [228]DSS-induced colitis in mice
-
Reduction of inflammation (IL-10)
DSS: dextran sulfate sodium. IL: interleukin. KGF-2: keratinocyte growth factor-2.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Murgiano, M.; Bartocci, B.; Puca, P.; di Vincenzo, F.; Del Gaudio, A.; Papa, A.; Cammarota, G.; Gasbarrini, A.; Scaldaferri, F.; Lopetuso, L.R. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int. J. Mol. Sci. 2025, 26, 3059. https://doi.org/10.3390/ijms26073059

AMA Style

Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. International Journal of Molecular Sciences. 2025; 26(7):3059. https://doi.org/10.3390/ijms26073059

Chicago/Turabian Style

Murgiano, Marco, Bianca Bartocci, Pierluigi Puca, Federica di Vincenzo, Angelo Del Gaudio, Alfredo Papa, Giovanni Cammarota, Antonio Gasbarrini, Franco Scaldaferri, and Loris Riccardo Lopetuso. 2025. "Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools" International Journal of Molecular Sciences 26, no. 7: 3059. https://doi.org/10.3390/ijms26073059

APA Style

Murgiano, M., Bartocci, B., Puca, P., di Vincenzo, F., Del Gaudio, A., Papa, A., Cammarota, G., Gasbarrini, A., Scaldaferri, F., & Lopetuso, L. R. (2025). Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. International Journal of Molecular Sciences, 26(7), 3059. https://doi.org/10.3390/ijms26073059

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

Article Metrics

Back to TopTop