Skip to content
BY 4.0 license Open Access Published by De Gruyter Open Access April 5, 2024

Probiotics for inflammatory bowel disease: Is there sufficient evidence?

  • Yueying Ma , Dandan Yang , Jin Huang , Kunli Liu , Huirong Liu , Huangan Wu EMAIL logo and Chunhui Bao EMAIL logo
From the journal Open Life Sciences

Abstract

Inflammatory bowel disease (IBD) refers to chronic inflammatory disorders of the gut. Ulcerative colitis (UC) and Crohn’s disease (CD) are two subtypes of IBD. Evidence suggests that the intestinal microbiota plays a role in the pathogenesis of IBD, so probiotics have garnered a lot of interest as a potential treatment or prevention for IBD. However, clinical evidence of the efficacy of probiotics is still debatable. We performed a literature review. An advanced search considered clinical studies on probiotic for IBD from inception to 2023 in PubMed, Embase, Cochrane Library, and Web of Science. In the treatment of UC with probiotics, only Escherichia coli Nissle 1917 for maintenance treatment of UC in remission, and Bifidobacterium and VSL#3 for induction of remission in patients with mild to moderately active UC have shown strong evidence. Currently, there are no definitive conclusions regarding the effectiveness of probiotics in CD. The mechanism of probiotic treatment for IBD may be related to reducing oxidative stress, repairing the intestinal barrier, regulating intestinal flora balance, and modulating intestinal immune response. Differences in the benefits of probiotics between CD and UC may be attributable to the different lesion extent and immune-mediated pathophysiology. More robust randomized clinical trials are required to validate the efficacy and safety of diverse probiotic strains in IBD.

1 Introduction

Inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn’s disease (CD), is a chronic inflammatory disorder of the gastrointestinal tract with a disease course characterized by frequent relapses. The clinical manifestations of IBD include (hemorrhagic) diarrhea, abdominal pain, weight loss and/or fatigue. Some patients also exhibit extra-intestinal manifestations such as skin lesions, pulmonary symptoms, or arthritis [1,2].

Initially, IBD was perceived as a condition predominantly prevalent in developed regions. However, an increasing number of cases have been observed across the world and the prevalence rates have shown an increasing trend [3]. The precise etiology of IBD is not well characterized. Host genetics, luminal environment, and the external environment have all been implicated in its causation [4,5,6]. A wide body of evidence from clinical and experimental studies suggests that dysbiosis of the intestinal bacteria characterized by structural and functional alterations of the gut microbiome may contribute to the development of IBD [7]. Some studies have shown that the emerging pathogenic bacteria may lead to increased incidence and severity of IBD in genetically susceptible individuals [8]. Most of the human IBD microbiome research conducted so far has focused on microbial composition rather than function [9]. Increased inflammation may be caused by the presence of bacteria which are not normally resident on the mucosal surface [10]. Owing to the presumed cause-and-effect relation between gut microbial dysbiosis and the development of IBD, various microbial-based therapies, such as probiotics [3], prebiotics [11], fecal microbiota transplantation [12], and nutritional supplements [13], are now available for the treatment of this disease.

Probiotics are defined as live microorganisms that confer a health benefit to the host by favorably altering the gut microflora after oral intake in adequate amounts [14]. Owing to their ease of administration and low cost, probiotics stand out among the many treatment therapies. They are often the most-used additional therapy in gastrointestinal diseases and are frequently recommended by physicians [15,16]. Microorganisms must meet several criteria to qualify as probiotic. They should be identified at the level of genus, species, and strain and should have demonstrable safety for clinical use, and the ability to survive intestinal transit. Most importantly, their clinical health effects should be validated in at least one phase 2 study [17]. However, although probiotics are widely used and generally regarded as safe [18], there is no clear consensus on the efficacy and safety of probiotic use in IBD. According to a meta-analysis, IBD patients receiving probiotics showed a higher risk of total adverse effects and gastrointestinal symptoms than those taking placebo; however, only abdominal pain showed statistically significant difference [19].

The strategy for manipulation of the microbial composition targeting the gut microbiota in IBD has been a research hotspot in recent decades. Each probiotic strain may have different immunoregulatory properties, and probiotics can modulate the intestinal immune response indirectly or directly [20]. Studies have shown that it is possible to modify the intestinal environment of patients with IBD by oral intake of probiotics [21,22].

2 Methodology

Owing to the conflicting evidence regarding the effectiveness of probiotics for IBD and the usage of several different kinds of probiotics, we conducted a literature review to summarize the results and conclusions of the clinical trials of probiotics in IBD. Advanced searches were performed in PubMed, Embase, Cochrane Library, and Web of Science. The search was performed by applying the following keywords alone or in combination: “Probiotic,” “Bifidobacterium,” “Escherichia coli Nissle 1917,” “Lactobacillus,” “Bifid Triple Viable,” “Saccharomyces,” “Inflammatory bowel disease,” “ulcerative colitis,” and “Crohn’s disease.” We considered clinical studies on probiotics for IBD from inception to 2023. A total of 55 articles were included. In particular, we sought to clarify the role of probiotics in the induction or maintenance of remission of IBD. In addition, we explored the potential mechanisms of the role of probiotics. This review may help inform the clinical use of probiotics in IBD.

3 Probiotics in the treatment of IBD

The clinical course of IBD is characterized by episodes of exacerbation and remission. Current treatment strategies for this disease include induction of remission first, followed by possibly long-term maintenance of remission [23]. We reviewed a total of 53 clinical trials of oral probiotics including clinical trials in IBD conducted over the last two decades. We organized these studies around the different subtypes of IBD (UC or CD) and the different disease stages (active or remission). The purpose of this review is to provide a broad overview of the studies on probiotics and their impact on IBD, as well as to discuss the effects of different probiotic strains.

3.1 Probiotics in UC

3.1.1 Induction of remission in active UC

Studies that investigated the induction of remission in active UC and evaluated the clinical outcomes are summarized in Table 1.

Table 1

Probiotics in UC for the induction of remission

Probiotic Author/year of publication/country Study design Methods Results Summary
Bifidobacteria-fermented milk Kato et al. 2004 [24] RPCT Patients: mild to moderate active UC Decrease in the CAI; reduce in the endoscopic activity index and histological score; increase in fecal butyrate, propionate, and short-chain fatty acid concentrations A beneficial effect was observed
Treatment period: 12 weeks
T( n = 10): bifidobacteria-fermented milk, 100 mL/day; conventional treatment (sulfasalazine or mesalazine)
Japan
C( n = 10): placebo; conventional treatment (sulfasalazine or mesalazine)
Bifidobacteria-fermented milk Ishikawa et al. 2011 [25] OL Patients: mild to moderate active UC Decrease in the endoscopic score, the MPO amounts, the fecal number of Bacteroidaceae, and the fecal pH A beneficial effect was observed
Treatment period: 1 year
T( n = 21): 1 g of the freeze-dried powder containing probiotic B. breve strain Yakult (109 CFU/g) thrice a day and 5.5 g of galacto-oligosac charide once a day
Japan
C( n = 20): none
B. infantis 35624 Groeger et al. 2013 [29] DB, RPCT Patients: mild to moderate active UC Reduction in the levels of CRP and TNF-α A beneficial effect was observed
Treatment period: 6 weeks
T( n = 13): sachets containing 1 × 1010 CFU viable B. infantis 35264
Ireland
C( n = 9): placebo
B. longum 536 Tamaki et al. 2016 [27] DB, RPCT Patients: mild-to-moderate active UC Reduction in UCDAI scores, EI and Mayo subscores, and rectal bleeding; clinical remission A beneficial effect was observed
Treatment period: 8 weeks
T( n = 28): 2–3 × 1011 freeze-dried viable B. longum 5,363 times daily
Japan
C( n = 28): placebo
B. longum Furrie 2005 [28] DB, RPCT Patients: active UC Reduce in sigmoidoscopy scores, mRNA levels for human beta defensins 2, 3, and 4, TNF-α, and IL-1α A beneficial effect was observed
Treatment period: 4 weeks
T( n = 9): 2 × 1011 freeze-dried viable B. longum in a gelatin capsule and a sachet containing 6 g of prebiotic fructo-oligosaccharide/inulin mix
United Kingdom
C( n = 9): placebo
B. longum Takeda et al. 2009 [30] OL Patients: mild to moderate active UC Decrease in the mean CAI at 8, 12, and 24 weeks A beneficial effect was observed
Treatment period: 24 weeks
T( n = 14): 2–3 × 1011 freeze-dried viable B. longum
No control group
Japan
Bifidobacterium Nagasaki et al. 2010 [26] Case report Patients: active UC Improve in physical condition and colonoscopic findings; possible to reduce the steroid dose without relapse A beneficial effect was observed
Treatment period: 1 week
Case Report( n = 1): Bifidobacterium 6 mg/day
Japan
E. coli Nissle 1917 Petersen et al. 2014 [32] DB, RPCT Patients: active UC No support for the use of E. coli Nissle as an add-on treatment to conventional therapies for active UC colitis No benefit was observed
Treatment period: 7 weeks
T1( n = 25): ciprofloxacin (500 mg twice daily) for 1 week followed by E. coli Nissle 1917 for 7 weeks (100 mg × 1 for 4 days followed by 100 mg × 2 daily for the rest of the period)
Denmark
T2( n = 25): ciprofloxacin for 1 week followed by placebo for 7 weeks
T3( n = 25): placebo for 1 week followed by E. coli Nissle 1917 for 7 weeks
T4( n = 25): placebo for 1 week followed by placebo for 7 weeks
E. coli Nissle 1917 Park et al. 2022 [34] DB, RCT Patients: using 5-ASA and presenting mild-to-moderate active UC Safe and effective in preventing the exacerbation of IBDQ scores and achieving clinical responses and endoscopic remission in patients with mild-to-moderate UC A beneficial effect was observed
Treatment period: a maximum of 12 months
T( n = 67): E. coli Nissle 1917 (2.5 × 109 CFU per capsule) one capsule/day from day 1 to day 4 and two capsules/day from day 5 for 8 weeks
Korea C( n = 66): placebo
E coli Nissle 1917 Rembacken et al. 1999 [33] DB, DD, RCT Patients: mild, moderate, and severe active UC An equivalent effect to mesalazine in inducing remission of UC A beneficial effect was observed
Treatment period: a maximum of 12 months
T( n = 57): gentamicin 80 mg three times daily for 1 week followed by E. coli Nissle 1917 at a dose of two capsules twice daily (2.5 × 1010 viable bacteria per capsule) for 12 weeks
United Kingdom
C( n = 59): gentamicin 80 mg for 1 week followed by mesalazine 800 mg three times daily
Lactobacillus delbruekii, L. fermentum Hegazy and El-Bedewy 2010 [35] RCT Patients: mild-to-moderate UC Decrease the colonic concentration of IL-6, expression of TNF-α and NF-κB p65, leukocyte recruitment (demonstrated by a decrease in colonic MPO activity), and the level of fecal calprotectin A beneficial effect was observed
Treatment period: 8 weeks
T( n = 15): 1010 CFU/day; sulfasalazine 2,400 mg/day
C( n = 15): sulfasalazine 2,400 mg/day
Egypt
L. casei DG D’Inca et al. 2011 [37] RCT Patients: mild left-sided UC Eight weeks of oral mesalazine did not provoke significant changes in the mucosa-associated microbiota in UC patients, nor did have a significant effect on the counts of Enterobacteriaceae spp. or of Lactobacillus spp No benefit was observed
Treatment period: 8 weeks
T( n = 8): oral mesalazine (2.4 g/day) plus oral L. casei DG (8 × 108 CFU) twice daily
Italy
C( n = 7): oral mesalazine (2.4 g/day)
L. reuteri ATCC 55730 Oliva et al. 2012 [36] RPCT Patients: mild-to-moderate distal UCTreatment period: 8 weeks Decrease in the Mayo score and the behavior of the histological score A beneficial effect was observed
T( n = 20): before bedtime an enema solution containing 1010 CFU of L. reuteri ATCC 55730
Italy
C( n = 20): placebo
L. rhamnosus GG, ATCC 5 03 Meini et al. 2015 [38] Case report Patients: active UC Bacteremia An adverse event was observed
Treatment period: 13 days
Case Report( n = 1): L. rhamnosus GG 6 × 109 CFU once daily; corticosteroids and mesalazine
Italy
Lactobacillus and Bifidobacterium Agraib et al. 2022 [39] DB, RPCT Patients: mild-to-moderate active UC Improvement in the Partial Mayo score, stool frequency, global assessment, and total PMS score; reduction in CRP and IgA level; increase in hemoglobin, hematocrit, RBC levels, and IL-10 levels A beneficial effect was observed
Treatment period: 6 weeks
T( n = 15): 3 × 1010 of probiotic product “probiotic 10 billion active cells®” (Jamieson Laboratories, Canada N8W5B5) capsules (containing nine Lactobacillus and five Bifidobacterium species) daily
Jordan
C( n = 15): placebo
Canada N8W5B5 Rayyan et al. 2023 [40] DB, RPCT Patients: mild-to-moderate UC Improvement in the scores of the systemic domain, social domain, bowel domain, emotional domain, and total SIBDQ A beneficial effect was observed
Treatment period: 6 weeks
T( n = 16): 3 × 1010 probiotic capsules containing nine Lactobacillus and five Bifidobacterium species 3 times daily
C( n = 14): placebo
Jordan
Lactobacillus salivarius, L. acidophilus, and Bifidobacterium bifidus strain BGN4 Palumbo et al. 2016 [41] RCT Patients: moderate-to-severe UC Ameliorate the clinical response; shorten the time of recovery; improve the stool frequency and intestinal mucosa aspect in the endoscopic picture A beneficial effect was observed
Treatment period: 2 years
T( n = 30): a double daily administration of a probiotic; mesalazine 1,200 mg
C( n = 30): mesalazine 1,200 mg
Italy
VSL#3 Tursi et al. 2004 [42] RCT Patients: mild-to-moderate UC The balsalazide/VSL#3 combination was faster in obtaining remission; better in improving well-being, bowel frequency, endoscopic, and histological scores A beneficial effect was observed
Treatment period: 8 weeks
T( n = 30): 2.25 g balsalazide daily as capsules containing 750 mg of balsalazide disodium, plus 3 g VSL#3 daily as 1 g bags containing 3 × 1011 viable lyophilized bacteria per gram
Italy
C1( n = 30): 4.50 g balsalazide
C1( n = 30): 2.4 g mesalazine
VSL#3 Miele et al. 2009 [46] DB, RPCT Patients: mild-to-moderate active UC; children age between 1.7 and 16.1 Maintained remission; decrease in endoscopic and histological scores A beneficial effect was observed
Treatment period: 1 year
Italy T( n = 14): VSL#3 4.5 × 1011 – 1.8 × 1012 (based on weight) bacteria/day; concomitant steroid induction and mesalamine
C( n = 15): placebo; mesalazine
VSL#3 Sood et al. 2009 [43] DB, RPCT Patients: mild-to-moderate UC Decrease in UCDAI scores; improvement in stool frequency score, blood in the stool score, mucosal appearance, and physician’s global assessment A beneficial effect was observed
Treatment period: 12 weeks
T( n = 77): 3.6 × 1012 CFU VSL#3 twice daily
C( n = 70): placebo
India
VSL#3 Tursi et al. 2010 [44] DB, RPCT Patients: mild-to-moderate UC Decrease in UCDAI scores; reduction in rectal bleeding A beneficial effect was observed
Treatment period: 8 weeks
T( n = 71): VSL#3 twice daily at a dose of 3.6 × 1012 CFU/day
Italy
C( n = 73): placebo
VSL#3 Ng et al. 2010 [45] DB, RPCT Patients: mild-to-moderately active UC Increase in IL-10 and IL-12p40; decrease in DC TLR-2 expression and IL-12p40 production A beneficial effect was observed
Treatment period: 8 weeks
T( n = 14): 2 sachets twice/day of VSL#3 (3.6 × 1012 bacteria)
United Kingdom
C( n = 14): placebo
VSL#3 Huynh et al. 2009 [47] OL Patients: children age between 3 and 17; mild to moderate acute exacerbation UC; had a duration of exacerbated symptoms lasting less than 4 weeks Decrease in SCCAI; improvement in the mean Mayo endoscopic score, TNF-α, IFN-γ, CRP, and ESR A beneficial effect was observed
Treatment period: 8 weeks
Canada
T( n = 18): a dose of probiotic based on their age (from one-half sachet to two & one-half sachets twice daily
No control group
VSL#3 Bibiloni et al. 2005 [48] OL Patients: mild-to-moderate active UC Decrease in UCDAI in patients entering remission or responding; clinical remission A beneficial effect was observed
Treatment period: 6 weeks
T( n = 34): VSL#3 (3.6 × 1012 bacteria) daily in two divided doses
Canada, Italy, USA
No control group
VSL#3 Soo et al. 2008 [49] OL Patients: mild-to-moderate active or quiescent UC colitis Decrease in UCDAI; increase in mucosal alkaline sphingomyelinase activity A beneficial effect was observed
Treatment period: 5 weeks
Canada T( n = 15): one sachet of VSL#3 (containing 9 × 1011 lyophilized bacteria) orally two times per day
No control group
BIO-THREE Tsuda et al. 2007 [51] OL Patients: mild-to-moderate distal UC Decrease in UCDAI score in patients who achieved either remission or response; clinical and endoscopic improvement accompanying changes of the intestinal microflora pattern A beneficial effect was observed
Treatment period: 4 weeks
T( n = 20): BIO-THREE tablets (2 mg S. faecalis T-110, 10 mg C. butyricum TO-A and 10 mg B. mesentericus TO-A) per day
No control group
Japan
Bifid Triple Viable Li et al. 2012 [52] RCT Patients: mild-to-moderate active UC Decrease in the clinical symptom score, colon mucosa inflammation score, and IL-1β expression; increase in IL-10 and IgA expressions in colon mucosa and the ratio of peripheral blood CD4+ T cell to CD8+ T cell A beneficial effect was observed
Treatment period: 2 months
T( n = 41): Bifid Triple Viable 2 capsules three times daily and mesalazine 1 g two times daily
China
C( n = 41): mesalazine
Bifid Triple Viable Cui et al. 2004 [53] RPCT Patients: active UC Increase in the concentration of fecal lactobacilli and bifidobacterial; improvement in the activation of NF-κB and the expression of IL-10; decrease in the expressions of TNF-α and IL-1β; helpful in maintaining remission and preventing relapse of UC A beneficial effect was observed
Treatment period: 8 weeks
T( n = 30): BIFICO 1.26 g/day
C( n = 30): placebo
China
S. boulardii Guslandi et al. 2003 [54] OL Patients: mild to moderate clinical flare-up of UC Reduction in the clinical index score; clinical remission confirmed by sigmoidoscopy A beneficial effect was observed
Treatment period: 4 weeks
T( n = 25): S. boulardii 250 mg three times a day; mesalazine
Italy
No control group

OL: open label; DB: double blind; DD: double dummy; RPCT: randomized placebo-controlled trial; RCT: randomized controlled trial; C: control group; T: treatment group.

3.1.1.1 Bifidobacterium

For a long time, fermented dairy products have been utilized to treat gastrointestinal disorders. For the treatment of active UC, bifidobacteria-fermented milk containing live bifidobacteria, Yakult strains of Bifidobacterium breve, Bifidobacterium bifidum, and Lactobacillus acidophilus may be safe and more effective than conventional treatment (sulfasalazine or mesalazine) alone. In a randomized controlled trial (RCT) [24] of bifidobacteria-fermented milk, the probiotic group showed significantly lower clinical activity index (CAI) than the placebo group, as well as greater concentrations of fecal butyrate, propionate, and short-chain fatty acids. In an uncontrolled trial by Ishikawa et al. [25], bifidobacteria-fermented milk was found to be more effective than placebo in reducing fecal myeloperoxidase (MPO) levels, the number of bacillus-like bacteria, and fecal pH. Moreover, bifidobacteria-fermented milk was found to lower the endoscopic activity index in patients with active UC in both trials. Nagasaki et al. [26] reported a 71-year-old patient with active UC who was administered Bifidobacteria 1 week after the failure of several treatments and showed improvement in physical condition and colonoscopy findings, with the possibility of reducing the steroid dose without relapse. Three randomized placebo-controlled trials [27,28,29] also showed the effect of Bifidobacterium in inducing remission in patients with active UC. Administration of Bifidobacterium longum for 8 weeks [27] resulted in greater reduction in the UC disease activity index (UCDAI) scores, endoscopic index, Mayo subscores, and rectal bleeding, as well as induced clinical remission. Administration of B. longum for 4 weeks [28] resulted in better reduction in sigmoidoscopy scores, mRNA levels of human beta-defensins 2, 3, and 4, tumor necrosis factor-α (TNF-α), and interleukin (IL)-1α. Administration of Bifidobacteria infantis 35624 for 6 weeks [29] resulted in better reduction in the levels C-reactive protein (CRP) and TNF-α. In an uncontrolled study by Takeda et al. [30], administration of B. longum for 24 weeks decreased the mean CAI.

3.1.1.2 E. coli Nissle 1917

E. coli Nissle 1917, a non-pathogenic strain of E. coli, is probably the most intensively investigated bacterial strain among the Gram-negative microorganisms [31]. Three RCTs that investigated the effect of E. coli Nissle 1917 in active UC yielded inconsistent results. Petersen et al. [32] found no benefit of use of E. coli Nissle 1917 as an add-on treatment with Ciprofloxacin for active UC. However, in a study of 116 patients with active UC conducted by Rembacken et al. [33], the effect of 1-week treatment with gentamicin followed by E. coli Nissle 1917 was equivalent to that of mesalazine in inducing UC remission. A study of 133 patients also reported the safety and efficacy of E. coli Nissle 1917 in preventing the exacerbation of IBDQ scores and achieving clinical responses and endoscopic remission in patients with mild-to-moderate UC [34]. The effect of E. coli Nissle 1917 on inducing remission in UC could not be clarified because the three studies used different drugs as the underlying treatment.

3.1.1.3 Lactobacillus

Three RCTs have investigated the effect of Lactobacillus in UC. Hegazy and El-Bedewy [35] found that Lactobacillus not only decreased the colonic concentration of IL-6 and protein expressions of TNF-α and NF-κB p65 but also reduced the leukocyte recruitment (demonstrated by a decrease in colonic MPO activity) and the level of fecal calprotectin in patients with active UC. A decrease in the Mayo score and the behavior of the histological score were also found by Oliva et al. [36]. In contrast, D’Inca et al. [37] found no effect of 5-ASA coupled with oral Lactobacillus casei DG on the colonic flora and TLR expression, but when coupled with rectally administered L. casei DG, it modified colonic microbiota by increasing Lactobacillus spp. and mucosal IL-10, while reducing Enterobacteriaceae, TLR-4, and interleukin (IL)-1b mRNA levels. Meini et al. [38] documented a case of Lactobacillus-induced bacteremia after oral administration, which was resistant to vancomycin. A combination of Lactobacillus and Bifidobacterium was also shown to induce remission in UC patients [39] and improve the quality of life in mild to moderate UC patients [40]. A combination of Lactobacillus with other strains investigated by Palumbo et al. [41] reported a positive effect used in conjunction with mesalazine in moderate-to-severe active UC as demonstrated by achievement of clinical response, shortened time to recovery, and improved stool frequency and endoscopic intestinal mucosa scores. The efficacy of Lactobacillus for UC appears to be unclear and needs to be supported by more high-quality evidence.

3.1.1.4 VSL#3

VSL#3 is very diverse probiotic containing multiple different strains of Lactobacillus, Bifidobacterium, and Streptococcus genera. There are eight trials, including six adult studies and two pediatric studies. All these studies showed a beneficial effect of VSL#3 in mild-to-moderate active UC. Tursi et al. [42] compared VSL#3 combined with low-dose balsalazide to medium-dose balsalazide or mesalazine alone and found that the combination helped achieve faster remission and better-improved well-being, bowel frequency, endoscopic, and histological scores. Treatment with VSL#3 in combination with commonly used anti-inflammatory drugs seems to be more effective than treatment with anti-inflammatory drugs alone. Four double-blind RCTs [43,44,45,46] were performed with VSL#3. The dose of 3.6 × 1012 VSL#3 CFU per day in adults was found to be sufficient to achieve a good clinical response. Two large studies [43,44] found greater decrease in UCDAI scores and significantly more patients with active UC improved at least 50% in CAI after VSL#3 treatment for 12 and 8 weeks than placebo, respectively. Similar results were also obtained in three open-label studies. Huynh et al. [47] found a decrease in simple clinical colitis activity index (SCCAI) and improvement in the mean Mayo endoscopic score, TNF-α, interferon-gamma (IFN-γ), CRP, and erythrocyte sedimentation rate (ESR). Bibiloni et al. [48] found a decrease in UCDAI. Soo et al. [49] found a decrease in UCDAI and an increase in mucosal alkaline sphingomyelinase activity. According to the 4th Yale/Harvard Workshop on Probiotic Recommendations in 2015 [50], VSL#3 was rated as grade B recommendations in inducing UC remission. None of the studies on VSL#3 found any side effects or ineffectiveness. These studies demonstrated the potential of VSL#3 as a stand-alone or additional treatment for active UC.

3.1.1.5 Other probiotics

Several other probiotic stains have also been studied, including BIO-THREE, Bifid Triple Viable, and Saccharomyces boulardii. In the study by Tsuda et al. [51], BIO-THREE was found to decrease UCDAI score, alter the intestinal microflora pattern, and improve clinical and endoscopic conditions. Li et al. [52] found that Bifid Triple Viable administered with mesalazine was better than mesalazine alone in decreasing the clinical symptom score, colon mucosa inflammation score, and IL-1β expression and in increasing IL-10 and IgA expressions in colon mucosa and the ratio of peripheral blood CD4+ T cell to CD8+ T cell. An RCT of Bifid Triple Viable [53] showed similar results, which suggests that use of Bifid Triple Viable alone may also achieve the same effect. In an uncontrolled study by Guslandi et al. [54], S. boulardii reduced the clinical index score and achieved clinical remission, which was confirmed by sigmoidoscopy. Although all these studies observed a beneficial effect, more robust studies are required to provide more definitive evidence of the effectiveness of these probiotics for the treatment of active UC.

3.1.2 Maintenance of remission in UC

Studies that investigated the maintenance of remission in UC and evaluated clinical outcomes are summarized in Table 2.

Table 2

Probiotics in UC for the maintenance of remission

Probiotic Author/year of publication/country Study design Methods Results Summary
Bifidobacteria-fermented milk Ishikawa et al. 2003 [55] RCT Patients: diagnosed with UC at least 1 year previously Success in maintaining remission and preventive effects on the relapse of UC A beneficial effect was observed
Treatment period: 1 year
T( n = 11): Bifidobacteria-fermented milk product (which contained at least 1010 per 100 mL bottle) 100 mL each day
Japan
C( n = 10): none
Bifidobacteria-fermented milk Matsuoka et al. 2018 [56] DB, RPCT Patients: quiescent UC No differences were observed between the BFM and placebo groups The study was discontinued for lack of efficacy
Treatment period: 48 weeks
Japan T( n = 98): Bifidobacteria-fermented milk (B. breve strain Yakult (1010 bacteria) and L. acidophilus (109 bacteria) )100 mL/day
C( n = 97): placebo
E. coli Nissle 1917 Rembacken et al. 1999 [33] DB, DD, RCT Patients: UC in remission An equivalent effect to mesalazine in maintaining remission of UC A beneficial effect was observed
Treatment period: a maximum of 12 months
United Kingdom
T( n = 44): E. coli Nissle 1917 at a dose of two capsules daily (2.5 × 1010 viable bacteria per capsule)
C( n = 39): mesalazine 400 mg three times daily
E. coli Nissle 1917 Kruis et al. 2004 [57] DB, DD, RCT Patients: UC in remission (CAI ≦ 4, EI ≦ 4, no signs of acute inflammation on histological examination); at least two acute attacks of UC prior to the study; a duration of the current remission of no longer than 12 months Promising behavior in sustaining the remission phase, prevention from inflammatory state; the probiotic EcN provides significantly equivalent efficacy in preventing relapses of UC and is not inferior to the established gold standard mesalazine A beneficial effect was observed
Germany
Treatment period: 12 months
T( n = 162): 200 mg/day (2.5–25 × 109 viable bacteria)
C( n = 165): mesalazine
E. coli Nissle 1917 Kruis et al. 1997 [58] DB, DD, RCT Patients: UC in remission An equivalent effect to mesalazine in maintaining remission of UC A beneficial effect was observed
Treatment period: 12 weeks
T( n = 50): 200 mg/day (day 1–4, only 100 mg/day) of Mutaflor (100 mg contains 2.5 × 1010 viable E. coli Nissle 1917)
Germany
C( n = 53): mesalazine
E. coli Nissle 1917 Henker et al. 2008 [59] RCT Patients: children aged between 11 and 18; in remission for at least 3 months An equivalent effect to mesalazine in maintaining remission of UC A beneficial effect was observed
Treatment period: 4 weeks
T( n = 24): 2 capsules of E. coli Nissle 1917 (2.5 × 1010 viable bacteria per capsule) while tapering off mesalazine
Germany
C( n = 10): mesalazine
VSL#3 Venturi et al. 1999 [60] OL Patients: UC in remission; intolerant or allergic to mesalazine Increase in fecal concentrations of S. salivarius ssp. thermophilus, lactobacilli, and bifidobacterial; maintaining the remission A beneficial effect was observed
Treatment period: 12 months
T( n = 20): VSL#3 (containing 5 × 1011 cells/g) 2 doses of 3 g
Italy
No control group
VSL#3 Miele et al. 2009 [46] DB, RPCT Patients: mild-to-moderate active UC; children age between 1.7 and 16.1 Maintained remission A beneficial effect was observed
Treatment period: 1 year
Italy T( n = 14): VSL#3 4.5 × 1011 – 1.8 × 1012 (based on weight) bacteria/day; concomitant steroid induction and mesalamine
C( n = 15): placebo; mesalazine
Lactobacillus GG Zocco et al. 2006 [61] RCT Patients: UC Clinical remission; more effective than mesalazine in prolonging the relapse-free time A beneficial effect was observed
Treatment period: 12 months
T1( n = 65): Lactobacillus GG 18 × 109 viable bacteria/day
T2( n = 62): Lactobacillus GG 18 × 109 viable bacteria/day; mesalazine 2,400 mg/day
Italy
C( n = 60): mesalazine 2,400 mg/day
L. acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 (Probio-Tec AB-25) Wildt et al. 2011 [63] DB, RPCT Patients: left-sided UC colitis in remission No effect on maintenance of remission in patients with UC could be demonstrated; no significant clinical benefit concerning a number of days until relapse could be demonstrated No benefit was observed
Treatment period: 52 weeks
T( n = 20): Probio-Tec AB-25 1.5 × 1011 CFU daily (two capsules three times daily)
Denmark
C( n = 12): placebo
Lactobacillus salivarius subsp. salivarius UCC118 or B. infantis 35624 Shanahan et al. 2006 [64] DB, RPCT Patients: remission of UC No difference in relapse time between probiotics and placebo No benefit was observed
Treatment period: 1 year
T( n = 52-53): 109 probiotics daily
C( n = 52-53): placebo
NC
Bio-Three Yoshimatsu et al. 2015 [62] DB, RPCT Patients: UC in remission Lower relapse rates than placebo A beneficial effect was observed
Treatment period: 12 months
T( n = 23): 9 Bio-Three tablets/day
C( n = 23): placebo
Japan
Multi-strain probiotic Bjarnason et al. 2019 [66] DB, RPCT Patients: stabile UC in remission Reduce the fecal calprotectin levels A beneficial effect was observed
Treatment period: 4 weeks
T( n = 40): 1 mL/kg Symprove (L. rhamnosus NCIMB 30174, L. plantarum NCIMB 30173, L. acidophilus NCIMB 30175, and Enterococcus faecium NCIMB 30176) each morning on a fasting stomach
United Kingdom
C( n = 41): placebo
Biotop capsule Lee et al. 2022 [65] OS Patients: UC whose IBS-like symptoms persisted during endoscopic remission Improvement of bowel-related symptoms and quality of life A beneficial effect was observed
Treatment period: 1 month
T( n = 43): Biotop capsule (L. acidophilus, 75 mg; C. butyricum TO-A, 25 mg; B. mesentericus TO-A, 25 mg; and S. faecalis T-110, 5 mg) 3 times daily
Korea

OL: open label; DB: double blind; DD: double dummy; RPCT: randomized placebo-controlled trial; RCT: randomized controlled trial; OS: observational study; C: control group; T: treatment group; NC: not clear.

3.1.2.1 Bifidobacterium

Bifidobacteria-fermented milk was proven to be effective in maintaining remission and preventing the relapse of UC by Ishikawa et al. [55]. However, Matsuoka et al. [56] did not corroborate this conclusion. The trial was stopped because there were no differences in preventing relapse or Sutherland UCDAI scale scores between the Bifidobacteria-fermented milk and placebo arms. The authors opined that the lack of effectiveness may be attributable to the mode of delivery or dose of Bifidobacterium species, rather than to the lack of efficacy of the bacterial culture itself. Due to the lack of endoscopic analysis in the trial, the effect of Bifidobacteria-fermented milk on patient mucosal status could not be determined.

3.1.2.2 E. coli Nissle 1917

Three large RCTs compared E. coli Nissle 1917 with mesalazine [33,57,58]. No significant differences were found between the two interventions with respect to relapse rate or side effects. One pediatric trial [59] also confirmed this conclusion. E. coli Nissle 1917 was rated as grade A recommendations [50] for maintaining UC remission, as it showed a similar effect to mesalazine and with good safety and tolerance profile.

3.1.2.3 VSL#3

In an open-label study by Venturi et al. [60], VSL#3 was found to increase the fecal concentrations of Streptococcus salivarius ssp. Thermophilus, lactobacilli, and bifidobacterial in remission patients. Another study also demonstrated the role of VSL#3 in inducing and maintaining remission in active UC [46]. VSL#3 was recommended as grade A for the maintenance of UC remission by the fourth Triennial Yale/Harvard Workshop [50].

3.1.2.4 Other probiotics

Several clinical trials have investigated Lactobacillus, Bio-Three, and multi-strain probiotics. Zocco et al. [61] enrolled 187 patients and found Lactobacillus GG had an equivalent effect to mesalazine in terms of relapse rate but was more effective than mesalazine in terms of lengthening relapse-free duration. In the study by Yoshimatsu et al. [62], Bio-Three arm had a lower relapse rate than the placebo arm. Two RCTs [63,64] found no difference in UC remission maintenance between Lactobacillus plus Bifidobacterium and placebo, but an observational study found that a combination of L. acidophilus, Clostridium butyricum, Bacillus mesentericus, and Streptococcus faecalis [65]. Bjarnason et al. [66] found that multi-strain probiotic was better than placebo in reducing the fecal calprotectin levels, but there were no significant differences with respect to IBD quality of life questionnaire scores between the two groups. These studies involving probiotics outside the mainstream provide only flimsy evidence of their beneficial role in maintaining remission of UC.

3.2 Probiotics in CD

3.2.1 Induction of remission in active CD

Few studies have investigated the use of probiotics in patients with active CD. Table 3 presents a summary of the clinical outcomes of these studies.

Table 3

Probiotics in CD for the induction of remission

Probiotic Author/year of publication/country Study design Methods Results Summary
Lactobacillus GG Gupta et al. 2000 [67] OL Patients: 4 male children with mild-to-moderately active CD Improvement in clinical activity, intestinal permeability, intestinal barrier function, and clinical status; decrease in CDAI score A beneficial effect was observed
Treatment period:6 months
T( n = 4): Lactobacillus GG (1010 colony-forming units [CFU]) in enterocoated tablets twice a day
United States
No control group
Lactobacillus GG (L. GG) Schultz et al. 2004 [68] DB, RPCT Patients: moderate-to-active CD No differences were observed between the L. GG and placebo groups. The benefit of L. GG in inducing or maintaining medically induced remission in CD was not demonstrated No benefit was observed
Treatment period: 6 months
T( n = 5): L. GG (2 × 109 CFU/day; CAG Functional Foods, Omaha, NE)
United States
C( n = 6): placebo
Combined probiotics (Bifidobacterium and Lactobacillus) Fujimori et al. 2007 [69] OL Patients: active CD patients without history of operation for CD Decrease in CDAI, IOIBD scores, incidence of daily diarrhea, and index of abdominal pain A beneficial effect was observed
Treatment period: 13.0 ± 4.5 months
Japan T( n = 10): a synbiotic therapy consisting of probiotics (7.5 × 1010 colony forming units CFU per day, including Bifidobacteria and Lactobacillus) and probiotics Yuan (9.9 g per day)
No control group
E. coli Nissle 1917 Malchow 1997 [71] DB, RPCT Patients: active colonic CD The use of E. coli strain Nissle 1917 as an adjuvant for patients with colonic CD can reduce the intake of prednisolone and stop using steroids during the trial A beneficial effect was observed
Treatment period: 1 year
Germany T( n = 16): capsules containing a preparation of viable, nonpathogenic E. coli Nissle 1917
C( n = 12): placebo
Synbiotic comprising B. longum and Synergy 1 Steed et al. 2010 [70] DB, RPCT Patients: active CD Decrease in CDAIs, histological scores and TNF-a expression; mucosal bifidobacteria proliferated A beneficial effect was observed
Treatment period: 6 months
T( n = 13): 2 × 1011 freeze-dried viable B. longum in a gelatin capsule and a sachet containing 6 g of Synergy I twice daily
United Kingdom
C( n = 11): placebo

OL: open label; DB: double blind; RPCT: randomized placebo-controlled trial; RCT: randomized controlled trial; C: control group; T: treatment group.

3.2.1.1 Lactobacillus GG

Two studies investigated the effect of Lactobacillus in patients with active CD. Gupta et al. [67] discovered that after taking enteric-coated pills containing Lactobacillus GG for 1 week, four children with mild-to-moderate CD showed significant improvement in clinical activity and continued to improve throughout 24 weeks. The median CD activity index (CDAI) score of children was 73% lower than baseline, and the intestinal permeability was also improved. Subsequently, Schultz et al. [68] conducted a double-blind RCT to investigate whether oral Lactobacillus GG can induce or maintain remission in CD. However, only five patients (5/11) completed the study, with two remissions in both the intervention and control groups and no significant between-group difference.

3.2.1.2 Other probiotics

Several other studies with different probiotic strains have been performed and the results are summarized in Table 3. In a single-arm trial by Fujimori et al. [69], high-dose combined-probiotics (Bifidobacterium and Lactobacillus) mixed with prebiotics (psyllium) (mean duration of intervention: 13.0 ± 4.5 months) were found to be safe and beneficial in the treatment of active CD. After treatment, seven of ten patients showed improvement in clinical symptoms and a significant decrease in the CDAI and International Organization for the Study of IBD (IOIBD) scores; two of these patients were able to stop using prednisolone, while four others reduced their dosage. In another placebo-controlled trial, CD patients treated with Synbiotic (comprised of B. longum and Synergy 1) showed a significant decrease in CDAI and histological scores, and improved proliferation of Bifidobacterium mucosae [70]. Malchow [71] found that use of E. coli strain Nissle 1917 as an adjuvant may help reduce the prednisolone dose in individuals with colonic CD. Despite these demonstrated benefits of probiotic strains in CD, the quality and the number of studies are limited. More placebo-controlled trials are required to provide robust evidence.

3.2.2 Maintenance of remission in CD

Studies that investigated the use of probiotics in the maintenance of remission in CD and the evaluated clinical outcomes are summarized in Table 4.

Table 4

Probiotics in CD for the maintenance of remission

Probiotic Author/year of publication/country Study design Methods Results Summary
S. boulardii Garcia et al. 2008 [72] RPCT Patients: CD in remission The intestinal permeability has been improved, the lactulose/mannitol ratio has decreased, and the barrier function has improved; even though complete normalization was not achieved A beneficial effect was observed
Treatment period:3 months
T( n = 14): S. boulardii every 8 h as an oral capsule formulation which contained 200 mg lyophilized S. boulardii-17 (about 4 × 108 cells), 6 mg sucrose, and 2.4 mg magnesium stearate
Brazil
C( n = 17): placebo every 8 h as an oral capsule containing 200 mg cellulose, 6 mg sucrose, and 2.4 mg magnesium stearate
S. boulardii Guslandi et al. 2000 [73] RCT Patients: CD in clinical remission The clinical recurrence of CD in patients receiving mesalazide plus Bulazyme maintenance treatment was significantly reduced A beneficial effect was observed
Treatment period: 6 months
T( n = 16): S. boulardii 500 mg two capsules in the morning; Pentasa 500 mg two capsules twice a day
Italy
C( n = 16): mesalazine 500 mg in a sustained-release preparation in ethylcellulose microgranules, two capsules three times a day
S. boulardii Bourreille et al. 2013 [74] DB, RPCT Patients: remission after treatment with steroids or salicylates There were no significant differences between the groups in terms of the average activity index score of CD, the red blood cell sedimentation rate or the median level of CRP, and the median time to relapse No benefit was observed
Treatment period: 52 weeks
France
T( n = 80): S. boulardii 1 g/day
C( n = 79): placebo
L. rhamnosus strain GG Bousvaros et al. 2005 [75] RPCT Patients: children age between 5 and 21; CD in remission The recurrence time and the proportion of patients in the LGG and placebo groups are basically the same. No reliable correlations were found between drug intake, clinical status, and lactic acid bacteria colonization in feces No benefit was observed
Treatment period: 2 years
United States
T( n = 39): L. rhamnosus strain GG 1 capsule (containing at least 1010 bacteria and 295 mg inulin) twice a day
C( n = 36): placebo
L. johnsonii Van Gossum et al. 2007 [76] DB, RPCT Patients: CD prior to elective ileo-cecal resection Oral probiotic LA1 has no protective effect on early endoscopic recurrence of CD patients undergoing ileocecal resection. The histological scores, serum inflammatory indexes, and clinical recurrence rates of the two groups were similar No benefit was observed
Treatment period:12 weeks
T( n = 34): L. johnsonii (LA1, Nestec) in freeze-dried form and blended with maltodextrin at 1010 colony-forming units (CFU)/day
C( n = 36): placebo
Belgium
L. johnsonii Marteau et al. 2006 [77] DB, RPCT Patients: patients had undergone surgical resection of <1 m, removing all macroscopic lesions within the past 21 days Endoscopic score distribution did not differ significantly between the LA1 and placebo groups. L. johnsonii LA1 (4 × 109 CFU/day) did not have a sufficient effect, if any, to prevent endoscopic recurrence of CD No benefit was observed
Treatment period: 6 months
France
T( n = 48): two packets per day of lyophilized LA1 (2 × 109 CFU; n = 48)
C( n = 50): placebo
Lactobacillus GG Prantera et al. 2002 [78] DB, RPCT Patients: patients operated on for CD in whom all of the diseased gut had been removed Lactobacillus GG seems neither to prevent endoscopic recurrence at 1 year nor reduce the severity of recurrent lesions No benefit was observed
Treatment period: 52 weeks
T( n = 23): 2.46 g bags containing Lactobacillus GG (Dicoflor 60; Dicofarm, Rome, Italy) 6 × 109 colony forming units (cfu) twice daily
Italy
C( n = 22): placebo
Synbiotic 2000 Chermesh et al. 2007 [81] DB, RPCT Patients: CD undergoing resection No differences were observed between the 2 groups in the clinical, laboratory, and endoscopic outcome No benefit was observed
Treatment period:1 year
T( n = 20): Synbiotic 2000 (contains prebiotics and probiotics, including 1010 Pediacoccus pentoseceus, 1010 L. raffinolactis, 1010 L. paracasei subsp. paracasei 19, 1010 L. plantarum 2,362; and 2.5 g β-glucans, 2.5 g inulin, 2.5 g pectin, and 2.5 g resistant starch) once daily
Israel
C( n = 10): placebo
VSL#3 Madsen et al. 2008 [80] DB, RPCT Patients: CD patients who had a recent ileal-colonic resection with a small intestine to colon anastomosis Patients receiving VSL#3 had significantly reduced levels of ileal mucosal pro-inflammatory cytokines, IL-1β, TNFα, and IFN-γ, and increased levels of the immunomodulatory cytokine, TGFβ (p < 0.05), compared to patients receiving placebo, and less severe endoscopic recurrence (Rutgeerts Grades 3 or 4) A beneficial effect was observed
Treatment period: 90 days
T( n = 58): one sachet of VSL#3 (9 × 1011 bacteria) twice daily
NC
C( n = 62): placebo
Multi-strain probiotic Bjarnason et al. 2019 [66] DB, RPCT Patients: stabile CD in remission No significant changes were seen in CD No benefit was observed
Treatment period:4 weeks
T( n = 33): treatment with Multi-strain probiotic 1 mL/kg/day
United Kingdom
C( n = 29): placebo

OL: open label; DB: double blind; RPCT: randomized placebo-controlled trial; RCT: randomized controlled trial; C: control group; T: treatment group; NC: not clear.

3.2.2.1 S. boulardii

There are three RCTs associated with S. boulardii. The duration and dose of these trials were radically different, and the final results were likewise somewhat conflicting. Garcia Villela et al. [72] discovered that compared to placebo, S. boulardii improved the intestinal permeability of CD patients in remission as well as decreased the lactulose/mannitol ratio and altered intestinal mucosal barrier integrity while maintaining the baseline drug treatments (mesalazine, azathioprine, prednisone, metronidazole, and/or thalidomide) unchanged. Guslandi et al. [73] reported that 32 CD patients receiving mesalamine combined with S. boulardii had a significantly lower 6-month clinical recurrence rate (6.25%) than patients receiving standard mesalamine therapy (37.5%). In addition, there was improvement in diarrhea, abdominal pain, overall health, CDAI, and hematocrit levels. Nevertheless, Bourreille et al. [74] reported contrary results. They found no significant differences in mean CDAI scores, median ESR, CRP levels, or median time to relapse between patients receiving S. boulardii or placebo. Because of the inconsistent effects of trials of the probiotic yeast S. boulardii, more studies are still required to assess its effectiveness and safety for patients with CD in remission.

3.2.2.2 Lactobacillus

We found four studies related to Lactobacillus, none of which showed its effectiveness in maintaining the remission of CD. Bousvaros et al. [75] tracked 75 CD adolescents for 2 years and found that the relapse time and proportion of patients in the Lactobacillus intervention and placebo arms were essentially the same, with no significant differences in medication intake, clinical state, or fecal lactate. There was no strong link between bacterial colonization and health. Three other studies [76,77,78] examined the effect of Lactobacillus on the time to recurrence in patients after surgery, with endoscopic relapse as the primary outcome. In the study by Van Gosum et al. [76], oral Lactobacillus johnsonii intervention in 70 CD patients after ileocecal resection did not prevent early recurrence. Marteau et al. [77] investigated CD patients who received 2 sachets of lyophilized L. johnsonii (2 × 109 CFU) or placebo per day for 6 months and concluded that L. johnsonii was ineffective in preventing endoscopic microscopic inspection of CD recurrence following CD bowel resection. Prantera et al. [78] studied the effects of oral Lactobacillus GG in CD patients in whom the diseased part of the gut was surgically removed for a year; they found that it did not prevent endoscopic recurrence, nor did it diminish the incidence of relapse and severity. Moreover, a meta-analysis [79] found that using Lactobacillus GG as a maintenance medication may increase the recurrence rate of CD when compared to placebo and that Lactobacillus GG was less successful in lowering the relapse rate.

3.2.2.3 Other probiotics

A small number of clinical trials have also investigated the effects of Synbiotic 2000, VSL#3, and multi-strain probiotics, most of which were on post-operative patients. Patients receiving VSL#3 showed significantly lower levels of ileal mucosal pro-inflammatory cytokines IL-1β, TNF-α, and IFN-γ, higher levels of TGF-β, and milder endoscopic recurrence than placebo [80]. Chermesh et al. [81] reported that daily administration of Synbiotic 2000 (a cocktail rich in four probiotics and four prebiotics) showed no effect on postoperative recurrence in CD patients. Multiple strains of probiotics that reduced intestinal inflammation in UC patients showed no therapeutic effect on CD [66]. More research is needed to evaluate whether these probiotics can lower the occurrence of clinical relapse.

Overall, based on the available results, E. coli Nissle 1917 may be available for maintenance treatment of UC in remission; in addition, Bifidobacterium and VSL#3 may be available for the induction of remission in mild to moderately active UC. Definitive conclusions on the effectiveness of probiotics in CD cannot yet be formed. All clinical trials have shown the effectiveness of E. coli Nissle 1917 as a maintenance treatment for UC in remission. In addition, both Bifidobacterium and VSL#3 have been shown to induce remission in mild-to-moderately active UC. However, there is inconsistent/insufficient evidence of the effectiveness of other probiotics in inducing or maintaining remission of UC. Similarly, clinical trials of probiotics for CD have demonstrated their ineffectiveness or yielded inconsistent results.

4 Mechanism of probiotics in IBD

Probiotics alleviate or treat IBD by mitigating oxidative stress, repairing the intestinal barrier, regulating intestinal flora balance, and modulating intestinal immune response. A schematic illustration of their mechanism of action is shown in Figure 1.

Figure 1 
               Schematic illustration of the mechanism of action of probiotics in IBD.
Figure 1

Schematic illustration of the mechanism of action of probiotics in IBD.

4.1 Mitigation of oxidative stress

Studies have shown that oxidative stress plays a key role in the development of IBD and that the imbalance between reactive oxygen species (ROS) accumulation and antioxidant activity is closely related to the incidence and severity of IBD [82]. Low-to-moderate concentrations of ROS are associated with the maintenance of normal intestinal homeostasis; however, intestinal inflammation leads to the production of excessive ROS, resulting in damage to cell structure and function and increased intestinal permeability, further aggravating inflammation [83]. MPO, which is specific to neutrophils and monocytes, is an abundant granulocyte heme enzyme. ROS can be efficiently produced by MPO through halogenation or peroxidase cycle. Lactobacillus plantarum supplementation and E. coli Nissle 1917 can reduce MPO levels to optimize the immune barrier [84,85]. Superoxide dismutase (SOD) is an important antioxidant enzyme and a prime scavenger of oxidative free radicals in the body. The severity of IBD is related to the level of SOD. Bifidobacterium quadruplex combined with mesalazine for UC can increase the level of SOD [86]. L. plantarum and Bifidobacterium activate the Nrf2 pathway at the transcriptional level and upregulate antioxidant factors (e.g., SOD1, SOD2, GPX2), with significant effects on DSS-induced UC in mice [87]. The product of probiotics can also modulate oxidative stress-related pathways. Γ-Glutamylcysteine, an antioxidant secreted by Bifidobacterium, can inhibit endoplasmic reticulum stress-mediated ROS [88]. The tryptophan metabolite indole-3-lactic acid produced by B. infantis metabolism induces increased mRNA expression of SOD2 and NAD(P)H dehydrogenase [89]. In summary, probiotics can reduce the intestinal inflammatory response by inhibiting the production of oxidants, promoting the production of antioxidants, and regulating oxidative stress-related pathways.

4.2 Repair of intestinal barrier

The intestinal mucosal barrier is the first line of defense in the intestine against bacteria and viruses, which can be divided into four parts: mechanical barrier, microbial barrier, chemical barrier, and immune barrier [90]. IBD is characterized by varying degrees of intestinal mucosal injury, and studies have shown that probiotics can improve the damaged mucosal epithelial barrier. The tight junctions surrounding the apical side of the intestinal epithelium are the structural basis for maintaining the mechanical barrier. E. coli Nissle 1917 was shown to increase the expression of zonula occludins-1 in mouse intestinal epithelial cells and provide protection against the increased permeability of luminal material in the mucosa-associated with DSS colitis [91]. B. longum and VSL#3 gavage were shown to increase the expression of zonula occludins-1, ocludin, and claudin-1 in the colon tissue in a mouse model of TNBS-induced colitis [92]. The intestinal mucus layer in the outermost layer of the intestinal mucosal barrier is the first line of defense against harmful substances and pathogens in contact with the intestinal internal environment and is also a major part of the chemical barrier. In a study, Lactobacillus rhamnosus CNCM I-3690 physically maintained the regulated phagocytes and mucus layer, while counteracting changes in local and systemic lymphocytes [93]. Several genes involved in mucus layer production, including Muc2 (LFC 2.2), Muc6 (LFC 3.7), Muc5b (LFC 2.9), and Muc4 (LFC 1.24), were significantly upregulated in the B. breve UCC2003 experimental group [94]. VSL#3 inhibits pro-inflammatory chemokine KC, monocyte chemoattractant protein-1, and macrophage inflammatory protein-2 and upregulates tissue regenerative growth factor transforming growth factor-β, fibroblast growth factor-1, and vascular endothelial growth factor-A, resulting in accelerated relief of colitis symptoms in Muc2-deficient mice [95]. Overall, probiotics repair the intestinal barrier by repairing the mucosal epithelium and promoting the production of the Intestinal mucus layer.

4.3 Regulation of intestinal flora balance

Probiotics, which are indispensable for human health, play an important role in regulating intestinal flora. Probiotics are involved in synthesizing various vitamins, digesting food, promoting intestinal peristalsis, inhibiting the growth of pathogenic flora, and decomposing harmful and toxic substances [96]. Probiotics can inhibit the growth of pathogenic bacteria by producing bacteriocins and short-chain fatty acids and inhibit the multiplication and invasion of pathogenic bacteria by competing with pathogenic bacteria for nutrients [97]. Probiotics form a microbial barrier after they colonize the intestine, preventing pathogenic bacteria from adhering to and invading the intestine [98]. Selenium-enriched B. longum DD98 was found to improve the diversity of UC intestinal flora and promote the abundance of beneficial bacteria, including Lachnospiraceae, Lactobacillaceae, and Prevotellaceae at family level [99]. Compared to DSS-induced colitis mice, gavaged L. rhamnosus GG mice had higher relative abundance of two clades, Firmicutes and Bacteroidetes, and lower relative abundance of Proteobacteria and Epsilonbacteraeota. At the genus/species level, groups of Muribaculacea, Rikenellaceae_RC9_gut_group, and Lachnospiraceae_NK4A136_group were reduced, while Helicobacter and Escherichia–Shigella were significantly increased in mice with DSS-induced colitis [100]. In different chemical-induced IBD mice models, chitosan/sodium alginate-coated E. coli Nissle 1917 alleviated inflammation, repaired the colonic epithelial barrier, modulated the intestinal microbial community, and improved the abundance of Lachnospiraceae_NK4A136 and Odoribacter in the intestinal flora [101]. The formation of a flora of beneficial bacteria in the intestinal tract helps maintain the balance of intestinal flora and stops the onset and development of IBD.

4.4 Regulation of intestinal immune response

IBD is a chronic immune-mediated inflammatory disease. Probiotics and their metabolites can activate innate immunity and induce adaptive immunity to regulate immune cells (such as macrophages, T lymphocytes, DCs), bind Toll-like receptors, and activate signaling pathways, such as NF-κB, JAK/STAT, and MAPK [102,103,104]. IBD inflammation can be reduced by modulating the immune response, reducing the production of inflammatory factors, and promoting the secretion of anti-inflammatory factors. Treatment of colitis with E. coli Nissle 1917 resulted in restoration of secretory immunoglobulin A levels and reduction of IL-5, IL-13, TNF-α, and IFN-γ levels [105]. Lactobacillus spp. downregulated JAK, TIRAP, IRAK4, NEMO, and RIP genes in the NF-κB pathway, with different STAT gene expressions, as well as reduced IL-6 and IL-1β production [106]. L. plantarum ZS62 downregulated the serum levels of IL-1β, IL-6, IL-12, TNF-α, and IFN-γ, and the relative mRNA and protein expression of IL-1β, IL-12, and TNF-α in colonic tissues of IBD mice, with upregulation of serum and relative mRNA and protein expression levels of IL-10 [107]. Probiotics can alleviate IBD symptoms by reducing the degree of intestinal inflammation in the gut.

5 Discussion and conclusions

Despite the availability of a diverse range of biological agents and molecular-targeted therapies, primary and subsequent treatment failure rates for IBD continue to be high. Thus, development of novel therapeutic targets and calibration of the existing therapies are key imperative to improve the effectiveness, safety, and tolerability [108]. Despite the relative safety of probiotics, some patients tend to perceive these as health supplements rather than a treatment. Probiotics with demonstrable efficacy have great potential to move from supplemental to therapeutic agents for IBD in the future. Indeed, probiotics should be used more often as an alternative or as a supplement to conventional treatment in patients with IBD [109].

All trials have demonstrated the benefits of Bifidobacterium and VSL#3 in inducing remission of mild-to-moderate active UC with no side effects. Moreover, the efficacy of E. coli Nissle 1917 in maintaining UC remission has been found to be comparable to that of mesalazine. These are consistent with the positive results of three meta-analyses [110,111,112]. Moreover, probiotics may play a role in promoting mucosal healing, as mucosal performance assessment has become a standard part of IBD trials [113]. However, clinical trials investigating the effectiveness of other probiotics in the treatment of UC have yielded contradictory findings. The effect of probiotics on CD has been disappointing so far, with most studies showing ineffectiveness and conflicting evidence; thus, there is a lack of sufficient evidence to recommend their usage [110,114,115].

Available evidence suggests that probiotics are effective in relieving or treating IBD by alleviating oxidative stress, repairing the intestinal barrier, regulating the balance of intestinal flora, and modulating the intestinal immune response. The disparities in the benefits of probiotics between CD and UC may reflect the complexities of the probiotic (bacteria)–host interactions. The lesions of UC involve the mucosal and submucosal layers, whereas CD typically involves the entire intestinal wall, which may make intestinal repair more difficult in CD than in UC. Activation of the intestinal mucosal response is the direct cause of the onset and development of intestinal inflammation in IBD, where the most important cells are Th (Th1, Th2, Th17) and Tregs. CD is mediated by Th1, while UC is mediated by Th2. B. infantis was shown to promote Th1 and suppress Th2 immune responses [116]. Lactobacillus fermentum resulted in decreased levels of Th1, Th2, and Th17-related cytokines and increased IL-10 in the colon [117]. Bifidobacterium improved Th1/Th2 balance in mice, increased Th1 cytokine levels, and decreased Th2 cytokine levels in splenocytes [118]. VSL#3 retargeted allergen-specific Th2-polarized immune responses to Th1-T regulatory responses [119]. This may be the reason why probiotics have no significant relief and limited therapeutic effect in CD but are effective in UC. Compared to 24 kinds of probiotics, L. rhamnosus has the best effect in relieving weight loss and improving the Shannon index in the UC model; Lactobacillus reuteri has the best effect in reducing the UCDAI; L. acidophilus has the best effect in increasing the expression of tight junction protein ZO-1; and Lactobacillus coryniformis has the best effect in reducing the content of serum pro-inflammatory factor TNF-α [120]. Even with the same probiotic, different subtypes can show different effects, so the conditions they can treat may differ.

However, there are some limitations in these trials. First, many of these trials have not been sufficiently evaluated in terms of effectiveness, dose, or duration of administration. Second, the small sample size in these trials limits the generalizability of the findings. Third, most studies have adopted specific doses of probiotics without investigating the link between dose and response, making it difficult to compare results even for the same strains. Fourth, nearly all trials examining the effectiveness of Bifidobacterium in the treatment of IBD were conducted in Japan. Trials of E. coli Nissle 1917 for the maintenance treatment of UC in remission were conducted in Germany, while most of the trials examining VSL#3 were conducted in Italy. Regional heterogeneity may lead to bias in the effectiveness of various types of probiotics. Subgroup analyses of probiotic efficacy by geography, age, and gender were not performed in the available studies. Fifth, the western diet and its components affect the abundance, colonization, and phenotypic behavior of E. coli in the gut, which may trigger or contribute to intestinal inflammation. In contrast, the Mediterranean diet and specific dietary fibers can eliminate these effects and prevent inflammation [121]. The impact of diet on IBD pathogenesis and interactions with probiotics should be considered when studying the effectiveness of probiotics.

In the future, more RCTs are needed to investigate and validate the efficacy of single probiotic strains and combined probiotic applications for IBD. For probiotics with definite effects, it is recommended to investigate the bare minimum or precise probiotic required for specific advantages, which would help normalize the treatment [122]. For probiotics with unclear effects, it is recommended to investigate them from a mechanistic point of view and compare the differences in their effects to identify subsets and characteristics of IBD populations in whom probiotics are not treatment options. Use of probiotics as a preventive measure in individuals who are prone to IBD should be considered and the mechanism and course of action of probiotics should also be further understood. Moreover, probiotic engineering may be a promising new technology for the future treatment of IBD. Probiotic engineering uses suitable bacterial strains, such as L. rhamnosus [123], to form robust probiotic strains with enhanced functional properties that not only target the control of gut pathogenic microorganisms but also provide specific interventions for IBD [124]. Covalent-organic-framework-based artificial probiotics have been invented to treat IBD by regulating intestinal flora, suppressing intestinal inflammation, protecting intestinal epithelial cells, and modulating immunity [125]. New probiotic delivery systems are also being developed that may protect probiotics from harsh gastrointestinal conditions, improve intestinal adhesion and reduce immunogenicity [126]. Probiotics should adhere to strict guidelines from manufacturing to storage to distribution, making potential health benefits be maximized, and consumer faith in these helpful microbes can be bolstered by adopting thorough quality management measures to ensure their safety, efficacy, and consistency [127]. In addition to oral administration, fecal microbiota transplantation may be a reliable option for future treatment to improve the condition of IBD patients [128]. Fecal microbiota transplantation has been proven to be a therapeutic intervention for inducing clinical remission in UC, but achieving endoscopic remission and maintaining long-term remission remains a challenge, and there are safety concerns [129].

In conclusion, based on the available results, the use of E. coli Nissle 1917 for the maintenance treatment of UC in remission, and Bifidobacterium and VSL#3 for induction of remission of mild-to-moderately active UC is feasible. However, there is no definitive evidence of the effectiveness of other probiotics for the treatment of UC or probiotics for the treatment of CD. The mechanism of the therapeutic effect of probiotics in IBD may include reduced oxidative stress, repair of intestinal barrier, regulation of intestinal flora balance, and modulation of intestinal immune response. Differences in the benefits of probiotics between CD and UC may be attributable to the different lesion extent and immune-mediated pathophysiology in the two conditions.

Acknowledgements

We thank Mr. Li-Ming Chen for his suggestions on the revision of the manuscript.

  1. Funding information: This research was funded by the Natural Science Foundation of Shanghai (No. 22ZR1458300), the Special Clinical Research Project in the Health Industry of Shanghai Municipal Health Commission (No. 202340036), and the National Key Basic Research Program of China (No. 2015CB554500).

  2. Author contributions: Bao CH conceived the study; Ma YY, Yang DD, Huang J, and Liu KL performed the literature search; Liu HR and Wu HG performed the parsing of the literature; Ma YY and Yang DD performed the writing; and Bao CH edited and revised the manuscript.

  3. Conflict of interest: Authors state no conflict of interest.

  4. Data availability statement: Data sharing is not applicable to this article as no datasets were generated or analyzed during the current study.

References

[1] Ji XQ, Wang LX, Lu DG. Pulmonary manifestations of inflammatory bowel disease. World J Gastroenterol. 2014;20(37):13501–11.10.3748/wjg.v20.i37.13501Search in Google Scholar PubMed PubMed Central

[2] Annese V. A review of extraintestinal manifestations and complications of inflammatory bowel disease. Saudi J Med Med Sci. 2019;7(2):66–73.10.4103/sjmms.sjmms_81_18Search in Google Scholar PubMed PubMed Central

[3] Jakubczyk D, Leszczynska K, Gorska S. The effectiveness of probiotics in the treatment of inflammatory bowel disease (IBD)-A Critical Review. Nutrients. 2020;12(7):1973.10.3390/nu12071973Search in Google Scholar PubMed PubMed Central

[4] Niewiadomski O, Studd C, Wilson J, Williams J, Hair C, Knight R, et al. Influence of food and lifestyle on the risk of developing inflammatory bowel disease. Intern Med J. 2016;46(6):669–76.10.1111/imj.13094Search in Google Scholar PubMed

[5] Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–24.10.1038/nature11582Search in Google Scholar PubMed PubMed Central

[6] Ananthakrishnan AN. The exposome in inflammatory bowel disease. Trop Gastroenterol. 2014;35(3):135–40.10.7869/tg.200Search in Google Scholar PubMed

[7] Sartor RB, Wu GD. Roles for intestinal bacteria, viruses, and fungi in pathogenesis of inflammatory bowel diseases and therapeutic approaches. Gastroenterology. 2017;152(2):327–39.10.1053/j.gastro.2016.10.012Search in Google Scholar PubMed PubMed Central

[8] Leone V, Chang EB, Devkota S. Diet, microbes, and host genetics: The perfect storm in inflammatory bowel diseases. J Gastroenterol. 2013;48(3):315–21.10.1007/s00535-013-0777-2Search in Google Scholar PubMed PubMed Central

[9] Somineni HK, Kugathasan S. The microbiome in patients with inflammatory diseases. Clin Gastroenterol Hepatol. 2019;17(2):243–55.10.1016/j.cgh.2018.08.078Search in Google Scholar PubMed

[10] Lee M, Chang EB. Inflammatory bowel diseases (IBD) and the microbiome-searching the crime scene for clues. Gastroenterology. 2021;160(2):524–37.10.1053/j.gastro.2020.09.056Search in Google Scholar PubMed PubMed Central

[11] Bernstein CN. Antibiotics, probiotics and prebiotics in IBD. Nestle Nutr Inst Workshop Ser. 2014;79:83–100.10.1159/000360713Search in Google Scholar PubMed

[12] Ooijevaar RE, Terveer EM, Verspaget HW, Kuijper EJ, Keller JJ. Clinical application and potential of fecal microbiota transplantation. Annu Rev Med. 2019;70(1):335–51.10.1146/annurev-med-111717-122956Search in Google Scholar PubMed

[13] Malinowski B, Wicinski M, Sokolowska MM, Hill NA, Szambelan M. The rundown of dietary supplements and their effects on inflammatory bowel disease-a review. Nutrients. 2020;12(5):1423.10.3390/nu12051423Search in Google Scholar PubMed PubMed Central

[14] Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol. 2014;11(8):506–14.10.1038/nrgastro.2014.66Search in Google Scholar PubMed

[15] Cheifetz AS, Gianotti R, Luber R, Gibson PR. Complementary and alternative medicines used by patients with inflammatory bowel diseases. Gastroenterology. 2017;152(2):415–29.10.1053/j.gastro.2016.10.004Search in Google Scholar PubMed

[16] Cordina C, Shaikh I, Shrestha S, Camilleri-Brennan J. Probiotics in the management of gastrointestinal disease: Analysis of the attitudes and prescribing practices of gastroenterologists and surgeons. J Dig Dis. 2011;12(6):489–96.10.1111/j.1751-2980.2011.00534.xSearch in Google Scholar PubMed

[17] Borchers AT, Selmi C, Meyers FJ, Keen CL, Gershwin ME. Probiotics and immunity. J Gastroenterol. 2009;44(1):26–46.10.1007/s00535-008-2296-0Search in Google Scholar PubMed

[18] Sanders ME, Akkermans LM, Haller D, Hammerman C, Heimbach J, Hormannsperger G, et al. Safety assessment of probiotics for human use. Gut Microbes. 2010;1(3):164–85.10.4161/gmic.1.3.12127Search in Google Scholar PubMed PubMed Central

[19] Dore MP, Bibbo S, Fresi G, Bassotti G, Pes GM. Side effects associated with probiotic use in adult patients with inflammatory bowel disease: A systematic review and meta-analysis of randomized controlled trials. Nutrients. 2019;11(12):2913.10.3390/nu11122913Search in Google Scholar PubMed PubMed Central

[20] Macho FE, Pot B, Grangette C. Beneficial effect of probiotics in IBD: Are peptidogycan and NOD2 the molecular key effectors? Gut Microbes. 2011;2(5):280–6.10.4161/gmic.2.5.18255Search in Google Scholar PubMed

[21] Quigley EM. Prebiotics and probiotics: Their role in the management of gastrointestinal disorders in adults. Nutr Clin Pract. 2012;27(2):195–200.10.1177/0884533611423926Search in Google Scholar PubMed

[22] Mack DR. Probiotics in inflammatory bowel diseases and associated conditions. Nutrients. 2011;3(2):245–64.10.3390/nu3020245Search in Google Scholar PubMed PubMed Central

[23] Derikx LA, Dieleman LA, Hoentjen F. Probiotics and prebiotics in ulcerative colitis. Best Pract Res Clin Gastroenterol. 2016;30(1):55–71.10.1016/j.bpg.2016.02.005Search in Google Scholar PubMed

[24] Kato K, Mizuno S, Umesaki Y, Ishii Y, Sugitani M, Imaoka A, et al. Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Aliment Pharm Ther. 2004;20(10):1133–41.10.1111/j.1365-2036.2004.02268.xSearch in Google Scholar PubMed

[25] Ishikawa H, Matsumoto S, Ohashi Y, Imaoka A, Setoyama H, Umesaki Y, et al. Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: A randomized controlled study. Digestion. 2011;84(2):128–33.10.1159/000322977Search in Google Scholar PubMed

[26] Nagasaki A, Takahashi H, Iinuma M, Uchiyama T, Watanabe S, Koide T, et al. Ulcerative colitis with multidrug-resistant Pseudomonas aeruginosa infection successfully treated with bifidobacterium. Digestion. 2010;81(3):204–5.10.1159/000236042Search in Google Scholar PubMed

[27] Tamaki H, Nakase H, Inoue S, Kawanami C, Itani T, Ohana M, et al. Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: A randomized, double-blinded, placebo-controlled multicenter trial. Dig Endosc. 2016;28(1):67–74.10.1111/den.12553Search in Google Scholar PubMed

[28] Furrie E. Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: A randomised controlled pilot trial. Gut. 2005;54(2):242–9.10.1136/gut.2004.044834Search in Google Scholar PubMed PubMed Central

[29] Groeger D, O’Mahony L, Murphy EF, Bourke JF, Dinan TG, Kiely B, et al. Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes. 2013;4(4):325–39.10.4161/gmic.25487Search in Google Scholar PubMed PubMed Central

[30] Takeda Y, Nakase H, Namba K, Inoue S, Ueno S, Uza N, et al. Upregulation of T-bet and tight junction molecules by Bifidobactrium longum improves colonic inflammation of ulcerative colitis. Inflamm Bowel Dis. 2009;15(11):1617–8.10.1002/ibd.20861Search in Google Scholar PubMed

[31] Sonnenborn U. Escherichia coli strain Nissle 1917-from bench to bedside and back: History of a special Escherichia coli strain with probiotic properties. FEMS Microbiol Lett. 2016;363(19):fnw212.10.1093/femsle/fnw212Search in Google Scholar PubMed

[32] Petersen AM, Mirsepasi H, Halkjaer SI, Mortensen EM, Nordgaard-Lassen I, Krogfelt KA. Ciprofloxacin and probiotic Escherichia coli Nissle add-on treatment in active ulcerative colitis: A double-blind randomized placebo controlled clinical trial. J Crohns Colitis. 2014;8(11):1498–505.10.1016/j.crohns.2014.06.001Search in Google Scholar PubMed

[33] Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon AT. Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: A randomised trial. Lancet. 1999;354(9179):635–9.10.1016/S0140-6736(98)06343-0Search in Google Scholar

[34] Park S, Kang S, Kim S, Kim TO, Cha JM, Im JP, et al. Additive effect of probiotics (Mutaflor) on 5-aminosalicylic acid therapy in patients with ulcerative colitis. Korean J Intern Med. 2022;37(5):949–57.10.3904/kjim.2021.458Search in Google Scholar PubMed PubMed Central

[35] Hegazy SK, El-Bedewy MM. Effect of probiotics on pro-inflammatory cytokines and NF-kappaB activation in ulcerative colitis. World J Gastroenterol. 2010;16(33):4145–51.10.3748/wjg.v16.i33.4145Search in Google Scholar PubMed PubMed Central

[36] Oliva S, Di Nardo G, Ferrari F, Mallardo S, Rossi P, Patrizi G, et al. Randomised clinical trial: The effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Aliment Pharm Ther. 2012;35(3):327–34.10.1111/j.1365-2036.2011.04939.xSearch in Google Scholar PubMed

[37] D’Inca R, Barollo M, Scarpa M, Grillo AR, Brun P, Vettorato MG, et al. Rectal administration of Lactobacillus casei DG modifies flora composition and Toll-like receptor expression in colonic mucosa of patients with mild ulcerative colitis. Dig Dis Sci. 2011;56(4):1178–87.10.1007/s10620-010-1384-1Search in Google Scholar PubMed

[38] Meini S, Laureano R, Fani L, Tascini C, Galano A, Antonelli A, et al. Breakthrough Lactobacillus rhamnosus GG bacteremia associated with probiotic use in an adult patient with severe active ulcerative colitis: Case report and review of the literature. Infection. 2015;43(6):777–81.10.1007/s15010-015-0798-2Search in Google Scholar PubMed

[39] Agraib LM, Yamani MI, Tayyem R, Abu-Sneineh AT, Rayyan YM. 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;51:83–91.10.1016/j.clnesp.2022.08.020Search in Google Scholar PubMed

[40] Rayyan YM, Agraib LM, Alkhatib B, Yamani MI, Abu-Sneineh AT, Tayyem RF. Does probiotic supplementation improve quality of life in mild-to-moderately active ulcerative colitis patients in Jordan? A secondary outcome of the randomized, double-blind, placebo-controlled study. Eur J Nutr. 2023;62(7):3069–77.10.1007/s00394-023-03207-8Search in Google Scholar PubMed

[41] Palumbo VD, Romeo M, Marino GA, Carini F, Damiani P, Damiano G, et al. The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomed Pap. 2016;160(3):372–7.10.5507/bp.2016.044Search in Google Scholar PubMed

[42] Tursi A, Brandimarte G, Giorgetti GM, Forti G, Modeo ME, Gigliobianco A. Low-dose balsalazide plus a high-potency probiotic preparation is more effective than balsalazide alone or mesalazine in the treatment of acute mild-to-moderate ulcerative colitis. Med Sci Monit. 2004;10(11):126–31.Search in Google Scholar

[43] Sood A, Midha V, Makharia GK, Ahuja V, Singal D, Goswami P, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7(11):1202–9.e1.10.1016/j.cgh.2009.07.016Search in Google Scholar PubMed

[44] Tursi A, Brandimarte G, Papa A, Giglio A, Elisei W, Giorgetti GM, 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(10):2218–27.10.1038/ajg.2010.218Search in Google Scholar PubMed PubMed Central

[45] Ng SC, Plamondon S, Kamm MA, Hart AL, Al-Hassi HO, Guenther T, et al. Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis. Inflamm Bowel Dis. 2010;16(8):1286–98.10.1002/ibd.21222Search in Google Scholar PubMed

[46] Miele E, Pascarella F, Giannetti E, Quaglietta L, Baldassano RN, 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(2):437–43.10.1038/ajg.2008.118Search in Google Scholar PubMed

[47] Huynh HQ, DeBruyn J, Guan L, Diaz H, Li M, Girgis S, et al. Probiotic preparation VSL#3 induces remission in children with mild to moderate acute ulcerative colitis: A pilot study. Inflamm Bowel Dis. 2009;15(5):760–8.10.1002/ibd.20816Search in Google Scholar PubMed

[48] Bibiloni R, Fedorak RN, Tannock GW, Madsen KL, Gionchetti P, Campieri M, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100(7):1539–46.10.1111/j.1572-0241.2005.41794.xSearch in Google Scholar PubMed

[49] Soo I, Madsen KL, Tejpar Q, Sydora BC, Sherbaniuk R, Cinque B, et al. VSL#3 probiotic upregulates intestinal mucosal alkaline sphingomyelinase and reduces inflammation. Can J Gastroenterol. 2008;22(3):237–42.10.1155/2008/520383Search in Google Scholar PubMed PubMed Central

[50] Floch MH, Walker WA, Sanders ME, Nieuwdorp M, Kim AS, Brenner DA, et al. Recommendations for probiotic Use--2015 update: Proceedings and consensus opinion. J Clin Gastroenterol. 2015;49:69–73.10.1097/MCG.0000000000000420Search in Google Scholar PubMed

[51] Tsuda Y, Yoshimatsu Y, Aoki H, Nakamura K, Irie M, Fukuda K, et al. Clinical effectiveness of probiotics therapy (BIO-THREE) in patients with ulcerative colitis refractory to conventional therapy. Scand J Gastroenterol. 2007;42(11):1306–11.10.1080/00365520701396091Search in Google Scholar PubMed

[52] Li G, Zeng S, Liao W, Lv N. The effect of bifid triple viable on immune function of patients with ulcerative colitis. Gastroenterol Res Pract. 2012;2012:404752.10.1155/2012/404752Search in Google Scholar PubMed PubMed Central

[53] Cui HH, Chen CL, Wang JD, Yang YJ, Cun Y, Wu JB, et al. Effects of probiotic on intestinal mucosa of patients with ulcerative colitis. World J Gastroenterol. 2004;10(10):1521–5.10.3748/wjg.v10.i10.1521Search in Google Scholar PubMed PubMed Central

[54] Guslandi M, Giollo P, Testoni PA. A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur J Gastroenterol Hepatol. 2003;15(6):697–8.10.1097/00042737-200306000-00017Search in Google Scholar PubMed

[55] Ishikawa H, Akedo I, Umesaki Y, Tanaka R, Imaoka A, Otani T. Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis. J Am Coll Nutr. 2003;22(1):56–63.10.1080/07315724.2003.10719276Search in Google Scholar PubMed

[56] Matsuoka K, Uemura Y, Kanai T, Kunisaki R, Suzuki Y, Yokoyama K, et al. Efficacy of bifidobacterium breve fermented milk in maintaining remission of ulcerative colitis. Dig Dis Sci. 2018;63(7):1910–9.10.1007/s10620-018-4946-2Search in Google Scholar PubMed PubMed Central

[57] Kruis W, Fric P, Pokrotnieks J, Lukas M, Fixa B, Kascak M, et al. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut. 2004;53(11):1617–23.10.1136/gut.2003.037747Search in Google Scholar PubMed PubMed Central

[58] Kruis W, Schutz E, Fric P, Fixa B, Judmaier G, Stolte M. Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. Aliment Pharm Ther. 1997;11(5):853–8.10.1046/j.1365-2036.1997.00225.xSearch in Google Scholar PubMed

[59] Henker J, Muller S, Laass MW, Schreiner A, Schulze J. Probiotic Escherichia coli Nissle 1917 (EcN) for successful remission maintenance of ulcerative colitis in children and adolescents: An open-label pilot study. Z Gastroenterol. 2008;46(9):874–5.10.1055/s-2008-1027463Search in Google Scholar PubMed

[60] Venturi A, Gionchetti P, Rizzello F, Johansson R, Zucconi E, Brigidi P, et al. Impact on the composition of the faecal flora by a new probiotic preparation: Preliminary data on maintenance treatment of patients with ulcerative colitis. Aliment Pharm Ther. 1999;13(8):1103–8.10.1046/j.1365-2036.1999.00560.xSearch in Google Scholar PubMed

[61] Zocco MA, dal Verme LZ, Cremonini F, Piscaglia AC, Nista EC, Candelli M, et al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharm Ther. 2006;23(11):1567–74.10.1111/j.1365-2036.2006.02927.xSearch in Google Scholar PubMed

[62] Yoshimatsu Y, Yamada A, Furukawa R, Sono K, Osamura A, Nakamura K, et al. Effectiveness of probiotic therapy for the prevention of relapse in patients with inactive ulcerative colitis. World J Gastroenterol. 2015;21(19):5985–94.10.3748/wjg.v21.i19.5985Search in Google Scholar PubMed PubMed Central

[63] Wildt S, Nordgaard I, Hansen U, Brockmann E, Rumessen JJ. A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis. J Crohns Colitis. 2011;5(2):115–21.10.1016/j.crohns.2010.11.004Search in Google Scholar PubMed

[64] Shanahan F, Guarner F, Von Wright A, Vilpponen-Salmela T, O’Donoghue D, Kiely B, et al. A one year, randomised, double-blind, placebo controlled trial of a lactobacillus or a bidifobacterium probiotic for maintenance of steroid-induced remission of ulcerative colitis. AGA Abstr. 2006;130(4):A–44.Search in Google Scholar

[65] Lee J, Park SB, Kim HW, Lee HS, Jee SR, Lee JH, et al. Clinical efficacy of probiotic therapy on bowel-related symptoms in patients with ulcerative colitis during endoscopic remission: An observational study. Gastroenterol Res Pract. 2022;2022:1–5.10.1155/2022/9872230Search in Google Scholar PubMed PubMed Central

[66] Bjarnason I, Sission G, Hayee B. A randomised, double-blind, placebo-controlled trial of a multi-strain probiotic in patients with asymptomatic ulcerative colitis and Crohn’s disease. Inflammopharmacology. 2019;27(3):465–73.10.1007/s10787-019-00595-4Search in Google Scholar PubMed PubMed Central

[67] Gupta P, Andrew H, Kirschner BS, Guandalini S. Is lactobacillus GG helpful in children with Crohn’s disease? Results of a preliminary, open-label study. J Pediatr Gastr Nutr. 2000;31(4):453–7.10.1002/j.1536-4801.2000.tb07158.xSearch in Google Scholar

[68] Schultz M, Timmer A, Herfarth HH, Sartor RB, Vanderhoof JA, Rath HC. Lactobacillus GG in inducing and maintaining remission of Crohn’s disease. BMC Gastroenterol. 2004;4(1):5.10.1186/1471-230X-4-5Search in Google Scholar PubMed PubMed Central

[69] Fujimori S, Tatsuguchi A, Gudis K, Kishida T, Mitsui K, Ehara A, et al. High dose probiotic and prebiotic cotherapy for remission induction of active Crohn’s disease. J Gastroenterol Hepatol. 2007;22(8):1199–204.10.1111/j.1440-1746.2006.04535.xSearch in Google Scholar PubMed

[70] Steed H, Macfarlane GT, Blackett KL, Bahrami B, Reynolds N, Walsh SV, et al. Clinical trial: The microbiological and immunological effects of synbiotic consumption - a randomized double-blind placebo-controlled study in active Crohn’s disease. Aliment Pharm Ther. 2010;32(7):872–83.10.1111/j.1365-2036.2010.04417.xSearch in Google Scholar PubMed

[71] Malchow HA. Crohn’s disease and Escherichia coli. A new approach in therapy to maintain remission of colonic Crohn’s disease? J Clin Gastroenterol. 1997;25(4):653–8.10.1097/00004836-199712000-00021Search in Google Scholar PubMed

[72] Garcia Vilela E, De Lourdes DAFM, Oswaldo DGTH, Guerra PA, Carolina CAA, Paiva MF, et al. Influence of Saccharomyces boulardii on the intestinal permeability of patients with Crohn’s disease in remission. Scand J Gastroenterol. 2008;43(7):842–8.10.1080/00365520801943354Search in Google Scholar PubMed

[73] Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in maintenance treatment of Crohn’s disease. Dig Dis Sci. 2000;45(7):1462–4.10.1023/A:1005588911207Search in Google Scholar

[74] Bourreille A, Cadiot G, Le Dreau G, Laharie D, Beaugerie L, Dupas JL, et al. Saccharomyces boulardii does not prevent relapse of Crohn’s disease. Clin Gastroenterol Hepatol. 2013;11(8):982–7.10.1016/j.cgh.2013.02.021Search in Google Scholar PubMed

[75] Bousvaros A, Guandalini S, Baldassano RN, Botelho C, Evans J, Ferry GD, et al. A randomized, double-blind trial of Lactobacillus GG versus placebo in addition to standard maintenance therapy for children with Crohn’s disease. Inflamm Bowel Dis. 2005;11(9):833–9.10.1097/01.MIB.0000175905.00212.2cSearch in Google Scholar

[76] Van Gossum A, Dewit O, Louis E, de Hertogh G, Baert F, Fontaine F, et al. Multicenter randomized-controlled clinical trial of probiotics (Lactobacillus johnsonii, LA1) on early endoscopic recurrence of Crohn’s disease after lleo-caecal resection. Inflamm Bowel Dis. 2007;13(2):135–42.10.1002/ibd.20063Search in Google Scholar PubMed

[77] Marteau P, Lemann M, Seksik P, Laharie D, Colombel JF, Bouhnik Y, et al. Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn’s disease: A randomised, double blind, placebo controlled GETAID trial. Gut. 2006;55(6):842–7.10.1136/gut.2005.076604Search in Google Scholar PubMed PubMed Central

[78] Prantera C, Scribano ML, Falasco G, Andreoli A, Luzi C. Ineffectiveness of probiotics in preventing recurrence after curative resection for Crohn’s disease: A randomised controlled trial with Lactobacillus GG. Gut. 2002;51(3):405–9.10.1136/gut.51.3.405Search in Google Scholar PubMed PubMed Central

[79] Shen J, Ran HZ, Yin MH, Zhou TX, Xiao DS. Meta-analysis: The effect and adverse events of Lactobacilli versus placebo in maintenance therapy for Crohn disease. Intern Med J. 2009;39(2):103–9.10.1111/j.1445-5994.2008.01791.xSearch in Google Scholar PubMed

[80] Madsen K, Backer JL, Leddin D, Dieleman LA, Bitton A, Feagan B, et al. A Randomized Controlled Trial of VSL#3 for the Prevention of Endoscopic Recurrence Following Surgery for Crohn’s Disease. AGA Abstr. 2008;134(4):A–361.10.1016/S0016-5085(08)61682-0Search in Google Scholar

[81] Chermesh I, Tamir A, Reshef R, Chowers Y, Suissa A, Katz D, et al. Failure of synbiotic 2000 to prevent postoperative recurrence of Crohn’s disease. Dig Dis Sci. 2007;52(2):385–9.10.1007/s10620-006-9549-7Search in Google Scholar PubMed

[82] Guan G, Lan S. Implications of antioxidant systems in inflammatory bowel disease. Biomed Res Int. 2018;2018:1–7.10.1155/2018/1290179Search in Google Scholar PubMed PubMed Central

[83] Piechota-Polanczyk A, Fichna J. Review article: The role of oxidative stress in pathogenesis and treatment of inflammatory bowel diseases. Naunyn-Schmiedeberg’s Arch Pharmacol. 2014;387(7):605–20.10.1007/s00210-014-0985-1Search in Google Scholar PubMed PubMed Central

[84] Wu Y, Jha R, Li A, Liu H, Zhang Z, Zhang C, et al. Probiotics (Lactobacillus plantarum HNU082) supplementation relieves ulcerative colitis by affecting intestinal barrier functions, immunity-related gene expression, gut microbiota, and metabolic pathways in mice. Microbiol Spectr. 2022;10(6):e165122.10.1128/spectrum.01651-22Search in Google Scholar PubMed PubMed Central

[85] Li L, Liu T, Gu Y, Wang X, Xie R, Sun Y, et al. Regulation of gut microbiota-bile acids axis by probiotics in inflammatory bowel disease. Front Immunol. 2022;13:974395.10.3389/fimmu.2022.974305Search in Google Scholar PubMed PubMed Central

[86] Xie F, Li S, Fan Y, Li W, Lv Q, Sun X, et al. Efficacy and safety of bifidobacterium quadruple viable bacteria combined with mesalamine against uc management: A systematic review and meta-analysis. Oxid Med Cell Longev. 2022;2022:8272371.10.1155/2022/8272371Search in Google Scholar PubMed PubMed Central

[87] Wang Y, Guo Y, Chen H, Wei H, Wan C. Potential of Lactobacillus plantarum ZDY2013 and Bifidobacterium bifidum WBIN03 in relieving colitis by gut microbiota, immune, and anti-oxidative stress. Can J Microbiol. 2018;64(5):327–37.10.1139/cjm-2017-0716Search in Google Scholar PubMed

[88] Engevik MA, Herrmann B, Ruan W, Engevik AC, Engevik KA, Ihekweazu F, et al. Bifidobacterium dentium-derived y-glutamylcysteine suppresses ER-mediated goblet cell stress and reduces TNBS-driven colonic inflammation. Gut Microbes. 2021;13(1):1–21.10.1080/19490976.2021.1902717Search in Google Scholar PubMed PubMed Central

[89] Ehrlich AM, Pacheco AR, Henrick BM, Taft D, Xu G, Huda MN, et al. Indole-3-lactic acid associated with Bifidobacterium-dominated microbiota significantly decreases inflammation in intestinal epithelial cells. BMC Microbiol. 2020;20(1):357.10.1186/s12866-020-02023-ySearch in Google Scholar PubMed PubMed Central

[90] Liu L, Yin M, Gao J, Yu C, Lin J, Wu A, et al. Intestinal Barrier Function in the Pathogenesis of Nonalcoholic Fatty Liver Disease. J Clin Transl Hepatol. 2023;11(2):452–8.Search in Google Scholar

[91] Ukena SN, Singh A, Dringenberg U, Engelhardt R, Seidler U, Hansen W, et al. Probiotic Escherichia coli Nissle 1917 inhibits leaky gut by enhancing mucosal integrity. Plos One. 2007;2(12):e1308.10.1371/journal.pone.0001308Search in Google Scholar PubMed PubMed Central

[92] Chen X, Fu Y, Wang L, Qian W, Zheng F, Hou X. Bifidobacterium longum and VSL#3((R)) amelioration of TNBS-induced colitis associated with reduced HMGB1 and epithelial barrier impairment. Dev Comp Immunol. 2019;92:77–86.10.1016/j.dci.2018.09.006Search in Google Scholar PubMed

[93] Martin R, Chamignon C, Mhedbi-Hajri N, Chain F, Derrien M, Escribano-Vazquez U, et al. The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci Rep-UK. 2019;9(1):5398.10.1038/s41598-019-41738-5Search in Google Scholar PubMed PubMed Central

[94] Kiu R, Treveil A, Harnisch LC, Caim S, Leclaire C, van Sinderen D, et al. Bifidobacterium breve UCC2003 induces a distinct global transcriptomic program in neonatal murine intestinal epithelial cells. IScience. 2020;23(7):101336.10.1016/j.isci.2020.101336Search in Google Scholar PubMed PubMed Central

[95] Kumar M, Kissoon-Singh V, Coria AL, Moreau F, Chadee K. Probiotic mixture VSL#3 reduces colonic inflammation and improves intestinal barrier function in Muc2 mucin-deficient mice. Am J Physiol-Gastrointest Liver Physiol. 2017;312(1):G34–45.10.1152/ajpgi.00298.2016Search in Google Scholar PubMed

[96] de Oliveira G, Leite AZ, Higuchi BS, Gonzaga MI, Mariano VS. Intestinal dysbiosis and probiotic applications in autoimmune diseases. Immunology. 2017;152(1):1–12.10.1111/imm.12765Search in Google Scholar PubMed PubMed Central

[97] Lane ER, Zisman TL, Suskind DL. The microbiota in inflammatory bowel disease: Current and therapeutic insights. J Inflamm Res. 2017;10:63–73.10.2147/JIR.S116088Search in Google Scholar PubMed PubMed Central

[98] Yoon MY, Lee K, Yoon SS. Protective role of gut commensal microbes against intestinal infections. J Microbiol. 2014;52(12):983–9.10.1007/s12275-014-4655-2Search in Google Scholar PubMed

[99] Hu Y, Jin X, Gao F, Lin T, Zhu H, Hou X, et al. Selenium-enriched Bifidobacterium longum DD98 effectively ameliorates dextran sulfate sodium-induced ulcerative colitis in mice. Front Microbiol. 2022;13:955112.10.3389/fmicb.2022.955112Search in Google Scholar PubMed PubMed Central

[100] Tong L, Zhang X, Hao H, Liu Q, Zhou Z, Liang X, et al. Lactobacillus rhamnosus GG derived extracellular vesicles modulate gut microbiota and attenuate inflammatory in DSS-induced colitis mice. Nutrients. 2021;13(10):3319.10.3390/nu13103319Search in Google Scholar PubMed PubMed Central

[101] Zhou J, Li M, Chen Q, Li X, Chen L, Dong Z, et al. Programmable probiotics modulate inflammation and gut microbiota for inflammatory bowel disease treatment after effective oral delivery. Nat Commun. 2022;13(1):3432.10.1038/s41467-022-31171-0Search in Google Scholar PubMed PubMed Central

[102] Herrera-deGuise C, Serra-Ruiz X, Lastiri E, Borruel N. JAK inhibitors: A new dawn for oral therapies in inflammatory bowel diseases. Front Med-Lausanne. 2023;10:1089099.10.3389/fmed.2023.1089099Search in Google Scholar PubMed PubMed Central

[103] Mishima Y, Oka A, Liu B, Herzog JW, Eun CS, Fan TJ, et al. Microbiota maintain colonic homeostasis by activating TLR2/MyD88/PI3K signaling in IL-10-producing regulatory B cells. J Clin Invest. 2019;129(9):3702–16.10.1172/JCI93820Search in Google Scholar PubMed PubMed Central

[104] Gomez-Bris R, Saez A, Herrero-Fernandez B, Rius C, Sanchez-Martinez H, Gonzalez-Granado JM. CD4 T-cell subsets and the pathophysiology of inflammatory bowel disease. Int J Mol Sci. 2023;24:3.10.3390/ijms24032696Search in Google Scholar PubMed PubMed Central

[105] Souza E, Campos C, Reis DC, Cassali GD, Generoso SV, Cardoso VN, et al. Beneficial effects resulting from oral administration of Escherichia coli Nissle 1917 on a chronic colitis model. Benef Microbes. 2020;11(8):779–90.10.3920/BM2020.0045Search in Google Scholar PubMed

[106] Aghamohammad S, Sepehr A, Miri ST, Najafi S, Pourshafie MR, Rohani M. Anti-inflammatory and immunomodulatory effects of Lactobacillus spp. as a preservative and therapeutic agent for IBD control. Immun Inflamm Dis. 2022;10(6):e635.10.1002/iid3.635Search in Google Scholar PubMed PubMed Central

[107] Pan Y, Ning Y, Hu J, Wang Z, Chen X, Zhao X. The preventive effect of Lactobacillus plantarum ZS62 on DSS-induced IBD by regulating oxidative stress and the immune response. Oxid Med Cell Longev. 2021;2021:9416794.10.1155/2021/9416794Search in Google Scholar PubMed PubMed Central

[108] Al-Bawardy B, Shivashankar R, Proctor DD. Novel and Emerging Therapies for Inflammatory Bowel Disease. Front Pharmacol. 2021;12:651415.10.3389/fphar.2021.651415Search in Google Scholar PubMed PubMed Central

[109] Mercer M, Brinich MA, Geller G, Harrison K, Highland J, James K, et al. How patients view probiotics: Findings from a multicenter study of patients with inflammatory bowel disease and irritable bowel syndrome. J Clin Gastroenterol. 2012;46(2):138–44.10.1097/MCG.0b013e318225f545Search in Google Scholar PubMed PubMed Central

[110] Jonkers D, Penders J, Masclee A, Pierik M. Probiotics in the management of inflammatory bowel disease: A systematic review of intervention studies in adult patients. Drugs. 2012;72(6):803–23.10.2165/11632710-000000000-00000Search in Google Scholar PubMed

[111] Ganji-Arjenaki M, Rafieian-Kopaei M. Probiotics are a good choice in remission of inflammatory bowel diseases: A meta-analysis and systematic review. J Cell Physiol. 2018;233(3):2091–103.10.1002/jcp.25911Search in Google Scholar PubMed

[112] Derwa Y, Gracie DJ, Hamlin PJ, Ford AC. Systematic review with meta-analysis: The efficacy of probiotics in inflammatory bowel disease. Aliment Pharm Ther. 2017;46(4):389–400.10.1111/apt.14203Search in Google Scholar PubMed

[113] Boal CP, Cotter J. Mucosal healing in ulcerative colitis: A comprehensive review. Drugs. 2017;77(2):159–73.10.1007/s40265-016-0676-ySearch in Google Scholar PubMed

[114] Rolfe VE, Fortun PJ, Hawkey CJ, Bath-Hextall F. Probiotics for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev. 2006;4:D4826.10.1002/14651858.CD004826.pub2Search in Google Scholar PubMed

[115] Doherty G, Bennett G, Patil S, Cheifetz A, Moss AC. Interventions for prevention of post-operative recurrence of Crohn’s disease. Cochrane Database Syst Rev. 2009;4:D6873.10.1002/14651858.CD006873.pub2Search in Google Scholar PubMed

[116] Wang W, Luo X, Zhang Q, He X, Zhang Z, Wang X. Bifidobacterium infantis Relieves Allergic Asthma in Mice by Regulating Th1/Th2. Med Sci Monit. 2020;26:e920583.10.12659/MSM.920583Search in Google Scholar PubMed PubMed Central

[117] Jang YJ, Kim WK, Han DH, Lee K, Ko G. Lactobacillus fermentum species ameliorate dextran sulfate sodium-induced colitis by regulating the immune response and altering gut microbiota. Gut Microbes. 2019;10(6):696–711.10.1080/19490976.2019.1589281Search in Google Scholar PubMed PubMed Central

[118] Takahashi N, Kitazawa H, Iwabuchi N, Xiao JZ, Miyaji K, Iwatsuki K, et al. Oral administration of an immunostimulatory DNA sequence from Bifidobacterium longum improves Th1/Th2 balance in a murine model. Biosci Biotech Biochem. 2006;70(8):2013–7.10.1271/bbb.60260Search in Google Scholar PubMed

[119] Schiavi E, Barletta B, Butteroni C, Corinti S, Boirivant M, Di Felice G. Oral therapeutic administration of a probiotic mixture suppresses established Th2 responses and systemic anaphylaxis in a murine model of food allergy. Allergy. 2011;66(4):499–508.10.1111/j.1398-9995.2010.02501.xSearch in Google Scholar PubMed

[120] Jin W, Ai H, Huang Q, Li C, He X, Jin Z, et al. Preclinical evidence of probiotics in ulcerative colitis: A systematic review and network meta-analysis. Front Pharmacol. 2023;14:1187911.10.3389/fphar.2023.1187911Search in Google Scholar PubMed PubMed Central

[121] Faqerah N, Walker D, Gerasimidis K. Review article: The complex interplay between diet and Escherichia coli in inflammatory bowel disease. Aliment Pharm Ther. 2023;58(10):984–1004.10.1111/apt.17720Search in Google Scholar PubMed

[122] Shanahan F, Quigley EM. Manipulation of the microbiota for treatment of IBS and IBD-challenges and controversies. Gastroenterology. 2014;146(6):1554–63.10.1053/j.gastro.2014.01.050Search in Google Scholar PubMed

[123] Mathipa-Mdakane MG, Thantsha MS. Lacticaseibacillus rhamnosus: A Suitable Candidate for the Construction of Novel Bioengineered Probiotic Strains for Targeted Pathogen Control. Foods. 2022;11(6):785.10.3390/foods11060785Search in Google Scholar PubMed PubMed Central

[124] Mishra J, Stubbs M, Kuang L, Vara N, Kumar P, Kumar N. Inflammatory bowel disease therapeutics: A focus on probiotic engineering. Mediat Inflamm. 2022;2022:9621668.10.1155/2022/9621668Search in Google Scholar PubMed PubMed Central

[125] Deng Q, Zhang L, Liu X, Kang L, Yi J, Ren J, et al. COF-based artificial probiotic for modulation of gut microbiota and immune microenvironment in inflammatory bowel disease. Chem Sci. 2023;14(6):1165–598.10.1039/D2SC04984HSearch in Google Scholar PubMed PubMed Central

[126] Han M, Lei W, Liang J, Li H, Hou M, Gao Z. The single-cell modification strategies for probiotics delivery in inflammatory bowel disease: A review. Carbohydr Polym. 2024;324:121472.10.1016/j.carbpol.2023.121472Search in Google Scholar PubMed

[127] Ahire JJ, Rohilla A, Kumar V, Tiwari A. Quality management of probiotics: Ensuring safety and maximizing health benefits. Curr Microbiol. 2023;81(1):1.10.1007/s00284-023-03526-3Search in Google Scholar PubMed

[128] Mirsepasi-Lauridsen HC. Therapy Used to Promote Disease Remission Targeting Gut Dysbiosis, in UC Patients with Active Disease. J Clin Med. 2022;11(24):7472.10.3390/jcm11247472Search in Google Scholar PubMed PubMed Central

[129] Feng J, Chen Y, Liu Y, Lin L, Lin X, Gong W, et al. Efficacy and safety of fecal microbiota transplantation in the treatment of ulcerative colitis: A systematic review and meta-analysis. Sci Rep-UK. 2023;13(1):14494.10.1038/s41598-023-41182-6Search in Google Scholar PubMed PubMed Central

Received: 2023-08-17
Revised: 2023-11-26
Accepted: 2023-12-11
Published Online: 2024-04-05

© 2024 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 30.4.2024 from https://www.degruyter.com/document/doi/10.1515/biol-2022-0821/html
Scroll to top button