The Montreal classification for Crohn's disease (CD) and ulcerative colitis (UC) [9-11].

The Montreal classification for Crohn's disease (CD) and ulcerative colitis (UC) [9-11].

Source publication
Article
Full-text available
Inflammatory bowel disease (IBD), which affects millions of people worldwide, includes two separate diseases: Crohn's disease (CD) and ulcerative colitis (UC). Although the background (chronic inflammatory state) and some of the symptoms of CD and UC are similar, both diseases differ from each other. It is becoming clear that a combination of many...

Context in source publication

Context 1
... Montreal classification for UC includes the criteria of severity and extensity. Both classifications are shown in Table 1. The occurrence of UC and CD can be compared to a sine wave, periods of symptom intensification interweave with a remission phase. ...

Similar publications

Article
Full-text available
Background and aims: Fecal calprotectin (FC) is a biomarker of gut inflammation, and Escherichia coli Nissle 1917 (EcN) is a probiotic strain able to reduce gut inflammation and maintain disease remission in patients with inflammatory bowel disease (IBD). The aim is to assess the effects of EcN administration in patients with IBD in clinical remis...
Article
Full-text available
Background: Probiotics have been widely used in the treatment of inflammatory bowel disease (IBD) due to their special conditioning effects on the intestinal flora, but their efficacy in inducing and maintaining remission is still controversial. In the present study, we analyzed randomized controlled trials (RCTs) on the treatment of probiotics in...

Citations

... Despite years of research on UC treatment, current clinical therapies for this disease remain insufficient [4]. Owing to the lack of a satisfactory therapeutic regimen, patients with UC frequently require lifelong treatment [5,6]. Previous studies identified UC as a risk factor for colon cancer [7][8][9]. ...
Article
Full-text available
Objective This study aimed to evaluate the effects of trilobatin (TLB) on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice and further explore the underlying mechanisms from the perspectives of signaling pathway and gut microbiota. Methods A mouse model of UC was established using DSS. Trilobatin was administered via oral gavage. Disease severity was assessed based on body weight, disease activity index (DAI), colon length, histological detection, inflammation markers, and colonic mucosal barrier damage. Alternations in the NF-κB and PI3K/Akt pathways were detected by marker proteins. High-throughput 16S rRNA sequencing was performed to investigate the gut microbiota of mice. Results In the DSS-induced UC mice, TLB (30 μg/g) treatment significantly increased the body weight, reduced the DAI score, alleviated colon length shortening, improved histopathological changes in colon tissue, inhibited the secretion and expression of inflammation factors (TNF-α, IL-1β, and IL-6), and increased the expression of tight-junction proteins (ZO-1 and occludin). Furthermore, TLB (30 μg/g) treatment significantly suppressed the activation of NF-κB pathway and altered the composition and diversity of the gut microbiota, as observed in the variations of the relative abundances of Proteobacteria, Actinobacteriota, and Bacteroidota, in UC mice. Conclusion TLB effectively alleviates DSS-induced UC in mice. Regulation of the NF-κB pathway and gut microbiota contributes to TLB-mediated therapeutic effects. Our study not only identified a novel drug candidate for the treatment of UC, but also enhanced our understanding of the biological functions of TLB.
... Probiotics can improve the microenvironment of intestinal epithelial cells and boost the resistance of the intestinal mucosa to pathogenic bacteria by stimulating immune cells to secrete substances, such as lysozyme, secretory phospholipase A2, and defensin [50,51]. Probiotics also play a vital role in reducing inflammation by decreasing epithelial cell apoptosis, increasing the number of anti-inflammatory bacteria (e.g., Butyricimonas and Prevotella), and maintaining intestinal barrier function [52][53][54][55]. Another way in which probiotic strains may improve barrier function is through probiotic-derived proteins, which can transactivate Epidermal Growth Factor Receptor signaling to ameliorate cytokine-induced apoptosis and mitigate disruption of the epithelial barrier and inflammation [52,56]. ...
Article
Full-text available
Maintaining homeostasis within the intestinal microbiota is imperative for assessing the health status of hosts, and dysbiosis within the intestinal microbiota is closely associated with canine intestinal diseases. In recent decades, the modulation of canine intestinal health through probiotics and prebiotics has emerged as a prominent area of investigation. Evidence indicates that probiotics and prebiotics play pivotal roles in regulating intestinal health by modulating the intestinal microbiota, fortifying the epithelial barrier, and enhancing intestinal immunity. This review consolidates literature on using probiotics and prebiotics for regulating microbiota homeostasis in canines, thereby furnishing references for prospective studies and formulating evaluation criteria.
... Many studies have confirmed that beneficial bacterium is essential in relieving or treating IBD. The beneficial bacteria have the capacity to reinforce the self-protective mechanism of the intestinal mucosa, counteract the pathogenic impact of harmful bacteria, regulate the release of inflammatory factors, and expedite the restoration of the intestinal flora within a brief timeframe (Jakubczyk et al., 2020;Martyniak et al., 2021) . ...
... Numerous studies have utilised probiotics to modify the gut microbiota and its functions. They have been employed in specific conditions such as antibiotic-associated diarrhoea and necrotising enterocolitis, and they seem to offer benefits in IBD [67][68][69]. The mechanism through which they exert their effects may involve not only altering the microbiota but also influencing microbial functions. ...
Article
Full-text available
Inflammatory Bowel Disease (IBD), encompassing Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic and relapsing inflammatory condition of the intestine that significantly impairs quality of life and imposes a heavy burden on healthcare systems globally. While the exact etiology of IBD is unclear, it is influenced by genetic, environmental, immunological, and microbial factors. Recent advances highlight the gut microbiome’s pivotal role in IBD pathogenesis. The microbial dysbiosis characteristic of IBD, marked by a decline in beneficial bacteria and an increase in pathogenic microbes, suggests a profound connection between microbial imbalance and disease mechanisms. This review explores diagnostic approaches to IBD that integrate clinical assessment with advanced microbiological analyses, highlighting the potential of microbiome profiling as a non-invasive diagnostic tool. In addition, it evaluates conventional and emerging treatments and discusses microbiome-targeted intervention prospects, such as probiotics, symbiotics, and faecal microbiota transplantation. The necessity for future research to establish their efficacy and safety is emphasised.
... A recent critical review has reported on various in vitro (laboratory-based cell culture investigations) and in vivo studies (i.e., animal models and humans) on the overall benefit that probiotics can provide to the inflammatory conditions of IBD [94]. The review investigated the value of different probiotic bacteria, especially those from the Bifidobacterium spp., in various considerations that could have possibly been involved in the etiology of IBD [94]. ...
... A recent critical review has reported on various in vitro (laboratory-based cell culture investigations) and in vivo studies (i.e., animal models and humans) on the overall benefit that probiotics can provide to the inflammatory conditions of IBD [94]. The review investigated the value of different probiotic bacteria, especially those from the Bifidobacterium spp., in various considerations that could have possibly been involved in the etiology of IBD [94]. The authors investigated and reported probiotic modulatory properties amid different study models (i.e., cell lines, animal models of colitis, clinical studies) and reported on the usefulness of administration in relation to the treatment, prevention, and remission of IBD diseases. ...
Article
Full-text available
Adverse intestinal microbiome profiles described as a dysbiotic gut are a complicit etiological operative factor that can progress and maintain inflammatory sequelae in the intestines. The disruption of the gut microbiome that ensues with intestinal dysbiosis is, for example, posited by decreases in the alpha-diversity of the gut microbiome, which is characterized by significant reductions in the abundance of bacterial members from the Bacteroidetes and Firmicutes phyla. Proteobacteria have often been recognized as gut microbial signatures of disease. For example, this happens with observed increases in abundance of the phyla Proteobacteria and Gammaproteobacteria, such as the adherent-invasive Escherichia coli strain, which has been significantly linked with maintaining inflammatory bowel diseases. Research on the administration of probiotics, often identified as gut-functional foods, has demonstrated safety, tolerability, and efficacy issues in treating inflammatory bowel diseases (IBDs). In this narrative review, we explore the efficacy of probiotics in treating IBDs with bacterial strain-and dose-specific characteristics and the association with multi-strain administration. Keywords: inflammatory bowel disease; ulcerative colitis; Crohn's disease; intestinal dysbiosis; gut microbiome; probiotics; prebiotics; functional foods
... A recent study provides compelling evidence that various functional food types such as probiotics, prebiotics, and herbs may have a favorable impact on the digestive tract [13]. Probiotics are likely to be beneficial in the prevention of IBD, whereas prebiotics include fibers that may inhibit the development of IBD [14][15][16]. Triangles, elm, and pie plant are herbs that may be associated with alleviating symptoms of IBDs. Mastic, herbal origin, may help manage heartburn, which is a common symptom of GERD [17]. ...
Article
Full-text available
This study explores the effects of the Mediterranean diet, herbal remedies, and their phytochemicals on various gastrointestinal conditions and reviews the global use of medicinal plants for common digestive problems. The review highlights key plants and their mechanisms of action and summarizes the latest findings on how plant-based products influence the digestive system and how they work. We searched various sources of literature and databases, including Google Scholar, PubMed, Science Direct, and MedlinePlus. Our focus was on gathering relevant papers published between 2013 and August 2023. Certain plants exhibit potential in preventing or treating digestive diseases and cancers. Notable examples include Curcuma longa, Zingiber officinale, Aloe vera, Calendula officinalis, Lavandula angustifolia, Thymus vulgaris, Rosmarinus officinalis, Ginkgo biloba, Cynodon dactylon, and Vaccinium myrtillus. The phytochemical analysis of the plants showed that compounds such as quercetin, anthocyanins, curcumin, phenolics, isoflavones glycosides, flavonoids, and saponins constitute the main active substances within these plants. These natural remedies have the potential to enhance the digestive system and alleviate pain and discomfort in patients. However, further research is imperative to comprehensively evaluate the benefits and safety of herbal medicines to use their active ingredients for the development of natural and effective drugs.
... However, the utilization of our native paraprobiotics exhibited a remarkable impact on these aforementioned indicators, clearly illustrating the beneficial anti-inflammatory characteristics of our native paraprobiotic. Therefore, we can verify the effectiveness of our native paraprobiotic strains possibility of any agent with positive outcomes that could extend the duration of the remission phase would undeniably have a direct influence on the overall well-being of patients suffering from IBD [19]. The health status of patients suffering from IBD may occasionally experience a decline and this deterioration is evident in both subtypes of IBD. ...
Article
Full-text available
Purpose Paraprobiotics are a non-viable form of probiotics that are reported to provide significant health benefits. Nevertheless, little is known about the beneficial effects of paraprobiotics on inflammatory bowel disease. Although probiotics show potential as therapeutic agents for a range of diseases, including inflammatory bowel disease (IBD), there are certain risks associated with their use. These risks include toxin production, hemolytic potential, antibiotic resistance, and the need to analyze metabolic activities. Hence Using paraprobiotic with the lower aforementioned risk would therefore be the preferable option. Here, we conducted an in vivo study to evaluate the preventive effect of our native paraprobiotic cocktail against dextran sulfate sodium (DSS)-induced murine colitis by affecting the autophagy signaling pathway. Methods Four-week-old male C57Bl/6 mice were randomly divided into three groups after a two-week acclimation period with normal standard laboratory food diet. Mice were administered PBS (PBS group as control), PBS along with DSS (DSS group, as a control), and a cocktail of paraprobiotics along with DSS (Para group). The severity of colitis, length and histopathology of the colon were evaluated. In addition, the expression of autophagy was assessed using real-time PCR. Results The results showed that administration of the paraprobiotic cocktail to DSS-treated mice inhibited the severity of colitis symptoms, as evidenced by the inhibition of weight loss and DAI, as well as histopathological scores in the study colon, as well as shortening of colon length caused by DSS. In contrast to the DSS group, the cocktail was able to modulate inflammation through upregulation of autophagy-related genes (becline 1, atg5, atg7, atg12, and atg13). Conclusion Although there are some limitations in our investigation, such as the dosage and duration of treatments, our native paraprobiotic blend effectively prevented the advancement of colitis. This suggests that it plays a vital role in regulating inflammation and preventing colitis by promoting the autophagy mechanism in cases where the consumption of probiotics may have negative consequences.
... 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. ...
... 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. ...
Article
Full-text available
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.
... Probiotics are live microorganisms that benefit the host when ingested adequately (Hill et al., 2014). The beneficial effects of these microorganisms are attributed to their ability to regulate the intestinal microbiota, increase short-chain fatty acids (SCFA) production (e.g., acetate, propionate, and butyrate), inhibit the proliferation of pathogenic bacteria, modulate the systemic and local immune responses, and to reinforce epithelial barrier via stimulation of tight junctions proteins expression and of mucus-producing goblet cells (Jonkers et al., 2012;Bellavia et al., 2013;Derikx et al., 2016;Jakubczyk et al., 2020). ...
... Probiotics, prebiotics, symbiotics, paraprobiotics, and postbiotics constitute promising therapeutic alternatives to ameliorate IBD (Martyniak et al., 2021). Selected strains of probiotic bacteria have already been demonstrated to modulate the immune response, intestinal barrier, and permeability in vivo (Bellavia et al., 2013;Batista et al., 2020;Jakubczyk et al., 2020;Wei et al., 2022). Among them, Lactococcus lactis is of great interest for probiotic applications. ...
Article
Full-text available
Introduction and objective p62 is a human multifunctional adaptor protein involved in key cellular processes such as tissue homeostasis, inflammation, and cancer. It acts as a negative regulator of inflammasome complexes. It may thus be considered a good candidate for therapeutic use in inflammatory bowel diseases (IBD), such as colitis. Probiotics, including recombinant probiotic strains producing or delivering therapeutic biomolecules to the host mucosal surfaces, could help prevent and mitigate chronic intestinal inflammation. The objective of the present study was to combine the intrinsic immunomodulatory properties of the probiotic Lactococcus lactis NCDO2118 with its ability to deliver health-promoting molecules to enhance its protective and preventive effects in the context of ulcerative colitis (UC). Material and methods This study was realized in vivo in which mice were supplemented with the recombinant strain. The intestinal barrier function was analyzed by monitoring permeability, secretory IgA total levels, mucin expression, and tight junction genes. Its integrity was evaluated by histological analyses. Regarding inflammation, colonic cytokine levels, myeloperoxidase (MPO), and expression of key genes were monitored. The intestinal microbiota composition was investigated using 16S rRNA Gene Sequencing. Results and discussion No protective effect of L. lactis NCDO2118 pExu:p62 was observed regarding mice clinical parameters compared to the L. lactis NCDO2118 pExu: empty. However, the recombinant strain, expressing p62, increased the goblet cell counts, upregulated Muc2 gene expression in the colon, and downregulated pro-inflammatory cytokines Tnf and Ifng when compared to L. lactis NCDO2118 pExu: empty and inflamed groups. This recombinant strain also decreased colonic MPO activity. No difference in the intestinal microbiota was observed between all treatments. Altogether, our results show that recombinant L. lactis NCDO2118 delivering p62 protein protected the intestinal mucosa and mitigated inflammatory damages caused by dextran sodium sulfate (DSS). We thus suggest that p62 may constitute part of a therapeutic approach targeting inflammation.
... These genera, well-regarded for their beneficial properties such as butyrate production and immune modulation, underscore the potential capability of XOS in modulating the gut microbiota. 67 Crucially, other clinical studies offer differing perspectives on the benefits of Bifidobacterium and Lactobacillus genera in active IBD phases. 68 Despite this, the benefits of XOS are limited by insufficient evidence in clinical IBD studies. ...
Article
Full-text available
Inflammatory bowel disease (IBD) is characterised by chronic inflammation in the gastrointestinal tract, with unclear aetiology but with known factors contributing to the disease, including genetics, immune responses, environmental factors and dysbiosis of the gut microbiota. Existing pharmacotherapies mainly target the inflammatory symptoms of disease, but recent research has highlighted the capacity for microbial-accessible carbohydrates that confer health benefits (ie, prebiotics) to selectively stimulate the growth of beneficial gut bacteria for improved IBD management. However, since prebiotics vary in source, chemical composition and microbiota effects, there is a clear need to understand the impact of prebiotic selection on IBD treatment outcomes. This review subsequently explores and contrasts the efficacy of prebiotics from various sources (β-fructans, galacto-oligosaccharides, xylo-oligosaccharides, resistant starch, pectin, β-glucans, glucomannans and arabinoxylans) in mitigating IBD symptomatology, when used as either standalone or adjuvant therapies. In preclinical animal colitis models, prebiotics have revealed type-dependent effects in positively modulating gut microbiota composition and subsequent attenuation of disease indicators and proinflammatory responses. While prebiotics have demonstrated therapeutic potential in animal models, clinical evidence for their precise efficacy remains limited, stressing the need for further investigation in human patients with IBD to facilitate their widespread clinical translation as microbiota-targeting IBD therapies.