Skip to main content

REVIEW article

Front. Cell. Infect. Microbiol., 08 December 2023
Sec. Intestinal Microbiome
Volume 13 - 2023 | https://doi.org/10.3389/fcimb.2023.1268041

Reviewing the potential of probiotics, prebiotics and synbiotics: advancements in treatment of ulcerative colitis

  • 1Herbal Medicine, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
  • 2Sai Ayurved Medical College, Maharashtra University of Health Sciences, Solapur, Maharashtra, India
  • 3Department of Zoology, Savitribai Phule Pune University, Pune, Maharashtra, India
  • 4Microbial Biotechnology, Rajiv Gandhi Institute of IT & Biotechnology, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India

Inflammatory bowel diseases (IBD) like Crohn’s and ulcerative colitis (UC) are multifactorial pathologies caused by environmental factors and genetic background. UC is a chronic inflammatory disorder that specifically targets the colon, resulting in inflammation. Various chemical interventions, including aminosalicylates, corticosteroids, immunomodulators, and biological therapies, have been extensively employed for the purpose of managing symptoms associated with UC. Nevertheless, it is important to note that these therapeutic interventions may give rise to undesirable consequences, including, but not limited to, the potential for weight gain, fluid retention, and heightened vulnerability to infections. Emerging therapeutic approaches for UC are costly due to their chronic nature. Alternatives like synbiotic therapy, combining prebiotics and probiotics, have gained attention for mitigating dysbiosis in UC patients. Prebiotics promote beneficial bacteria proliferation, while probiotics establish a balanced gut microbiota and regulate immune system functionality. The utilisation of synbiotics has been shown to improve the inflammatory response and promote the resolution of symptoms in individuals with UC through the stimulation of beneficial bacteria growth and the enhancement of intestinal barrier integrity. Hence, this review article aims to explore the potential benefits and underlying reasons for incorporating alternative approaches in the management of UC with studies performed using prebiotics, probiotics, and synbiotics to treat ulcerative colitis and to highlight safety and considerations in UC and future perspectives. This will facilitate the utilisation of novel treatment strategies for the safer and more efficacious management of patients with UC.

1 Introduction

Over the past decade, there has been an observed increase in the prevalence and incidence of Inflammatory Bowel Diseases (IBD), specifically Crohn’s Disease (CD) and Ulcerative Colitis (UC) (Molodecky et al., 2012; Roy and Dhaneshwar, 2023). According to a recent epidemiological study conducted by Ng et al. (2017), IBD affects over 0.3% of people in North America and Europe. It is a group of gastrointestinal disorders with chronic inflammation. The cause of IBD is not fully understood but is believed to be a combination of genetic and environmental factors. Factors such as diet, use of antibiotics, and socioeconomic status can influence persistent immune-mediated inflammation in the intestines. (Manichanh et al., 2012; Çavdar and Çavdar, 2023). UC is a chronic gastrointestinal disorder characterised by chronic inflammation of the colorectal mucosa, resulting in non-specific inflammatory changes. The aetiology of this condition remains uncertain, and its conversion into cancer is not easily treatable. Ungaro et al. (2017) conducted epidemiological research which substantiated the global increase in the prevalence of UC. It is a condition caused by a combination of genetic, immune, microbial, and environmental factors. It involves persistent inflammation of the mucosal lining, affecting the rectum and proximal colon segments. The exact mechanisms behind UC development are not fully understood, but it is widely recognised that environmental factors, host genetics, a dysregulated immune system, and microbial dysbiosis play crucial roles. (Ananthakrishnan, 2015).

According to the study conducted by Higuchi et al. (2012), there is evidence to suggest that the adoption of Westernised diets and lifestyles can heighten the susceptibility of genetically predisposed individuals to IBD. Environmental factors encompass various aspects, such as residing in industrialised nations, urbanised settings, and regions situated at higher latitudes. In addition to infections and smoking, other factors also play a role in the pathogenesis of IBD. Patients frequently need lifelong medication, which is the main purpose of therapy, both to persuade clinical remission and to conserve it for a long period. According to the Global Burden of Disease Study, the estimated global prevalence of Crohn’s disease was approximately 6.8 million individuals, while ulcerative colitis affected approximately 10 million individuals. (Alatab et al., 2020).

These data suggest that, in addition to finding effective therapeutic approaches to advance IBD patients’ quality of life and lower healthcare system costs, it is crucial to better understand the factors involved in the development of IBD. Costs of IBD management have increased significantly in the past five years due to different therapeutic interventions and disease characteristics. Patients face increased healthcare utilisation, out-of-pocket expenses, and reduced productivity, necessitating the implementation of cost-effective solutions. (Park et al., 2020) Recent research suggests that microbiota can be therapeutically effective in treating IBD. The gut microbiome influences host processes and can mitigate inflammation. Various therapeutic approaches, including prebiotics, probiotics, and synbiotics, have been developed to alter and restructure the gut microbiome. (Britton et al., 2021) The most common strains currently available as probiotics and possessing beneficial health effects are Enterococcus faecium, Bifidobacterium, Bacillus, Saccharomyces boulardii (S. boulardii) and Lactobacillus strains. Prebiotics like fructo-oligosaccharides (FOS), isomalto-oligosaccharides (IMO), and xylooligosaccharide (XOS) are studied for their potential benefits on stool volume, constipation relief, and faecal acidity, with easily metabolised by Bifidobacteria and other microorganisms. (Guarino et al., 2020).

2 Understanding ulcerative colitis

Ulcerative colitis is a chronic inflammatory disorder of the gastrointestinal tract characterised by a progressive decline in health. UC is characterised by inflammation of the mucosal lining, typically confined to the colon and rectum. On the other hand, CD involves inflammation that extends through the entire thickness of the gastrointestinal tract, with a predilection for the ileum, terminal ileum, and colon. The CD is often associated with additional inflammatory conditions outside of the intestines. According to Çavdar and Çavdar (2023), the medical characteristics encompass symptoms such as hemorrhagic diarrhoea, weight loss, fatigue, and abdominal pain. Additionally, certain individuals may develop extra-intestinal manifestations associated with IBD, such as primary sclerosing cholangitis, skin lesions, or joint complications (Judge and Lichtenstein, 2001). The impact of UC on a global scale has been evaluated, revealing that it affects a population of approximately 5 million individuals. UC is characterised by recurring episodes of inflammation in the mucosal layer of the colon, which alternate between periods of relapse and remission. In ulcerative colitis, inflammation is typically confined to the mucosal surface. The disorder originates in the rectum and typically progresses proximally throughout the entirety of the colon. To date, a conclusive therapeutic approach for UC remains elusive, and the currently available interventions primarily focus on sustaining remission (Roy and Dhaneshwar, 2023).

Conventional therapies for UC consist of pharmacological interventions involving the use of various agents, including azathioprine, aminosalicylates, and corticosteroids. These treatments aim to induce remission, prevent relapses, promote mucosal healing, and ultimately reduce the need for colectomy in individuals diagnosed with UC. In recent times, advancements in medical treatments have been influenced by the utilisation of agents such as monoclonal antibodies that specifically target proinflammatory cytokines, adhesion molecules, T-cell activation, and agents that promote the production of anti-inflammatory cytokines, such as IL-10 (Interleukin) and Transforming Growth Factor (TGF). However, the administration of these medications in the context of ulcerative colitis is associated with a distinct set of systemic and localised adverse effects experienced by individuals. These include but are not limited to, symptoms such as headache, abdominal pain, nausea, cramping, loss of appetite, vomiting, and rash. In addition, it is not recommended to use corticosteroids for an extended period due to their potential to induce glucose intolerance, osteoporosis, myopathy, and increased vulnerability to infections. These adverse effects raise significant concerns regarding the safety profile of corticosteroid usage (Mowat et al., 2011). Similarly, the expenses associated with these promising therapeutic approaches are substantial, considering the increasing prevalence and persistent nature of IBD. Histological examinations conducted on patients with UC have revealed the presence of impaired intestinal epithelial barrier function. However, the significance of this dysfunction with the pathology of UC remains uncertain. Patients diagnosed with UC exhibit changes in the function of the epithelial barrier, impaired function of tight junctions, and reduced secretion of mucin-2. Nevertheless, the exact relationship between these abnormalities and chronic inflammation, accompanied by heightened cytokine production, remains uncertain. Heller et al. (2005) conducted a study that yielded evidence indicating that the overexpression of IL-13 plays a pivotal role in the disruption of tight junctions within the intestinal mucosal layer of individuals who have been diagnosed with active UC. The dysfunction of the barrier function in individuals diagnosed with UC has been linked to a heightened absorption of luminal antigens and an elevated susceptibility to bacterial infiltration in the underlying mucosa. Extensive research has provided comprehensive insights into the dysregulation of the intestinal immune system in individuals afflicted with UC. Dendritic cells (DCs) are known to have a pivotal role in the augmentation of UC patients, thus signifying the substantial contribution of these cells in the initiation and perpetuation of inflammatory processes (Hart et al., 2005).

3 Conventional treatments for UC

Conventional therapies play a pivotal role in the management of ulcerative colitis and the mitigation of associated symptoms. The objective of these treatments is to manage inflammation, initiate and sustain remission, and enhance the overall quality of life for patients. The Selecting Therapeutic Targets in IBD (STRIDE)-II statement was revised by the International Organisation for the Study of IBD to enhance the treatment of IBD in adults and children. The revised statement identified several goals at different timeframes. In the short term, achieving symptomatic remission and normalising C-reactive protein levels were prioritised. In the intermediate term, reducing calprotectin levels was identified as a target. Lastly, in the long term, the goals included achieving endoscopic healing and standardising the quality of life for individuals with IBD (Im et al., 2018; Turner et al., 2021). The subsequent therapies listed are commonly employed conventional treatments for ulcerative colitis.

3.1 Aminosalicylates

Aminosalicylates, alternatively referred to as 5-aminosalicylic acid (5-ASA) drugs, encompass a class of pharmaceuticals that possess the capability to mitigate inflammation within the gastrointestinal tract. These therapeutic agents function by specifically targeting the mucosal lining of the colon and rectum, which is the site of inflammation. Aminosalicylates are offered in diverse formulations, encompassing oral pharmaceuticals, rectal suppositories, and enemas. Corticosteroids are frequently employed as an initial therapeutic approach for individuals with mild to moderate ulcerative colitis, effectively managing symptoms and sustaining periods of remission. The class of medications known as 5-ASA-based drugs includes sulfasalazine, olsalazine, Mesalazine and balsalazide. These medications are widely recognised for their efficacy, safety, and affordability in the treatment of IBD, particularly UC (Sood et al., 2019).

3.2 Corticosteroids

Corticosteroids, such as prednisone and budesonide, exhibit potent anti-inflammatory properties and have demonstrated efficacy in promptly alleviating symptoms associated with moderate to severe ulcerative colitis exacerbations. These medications function through the inhibition of the immune system and the mitigation of inflammation within the colon. Corticosteroids are commonly prescribed for a limited duration to induce remission or manage acute symptoms. The avoidance of prolonged corticosteroid use is typically recommended due to the potential for significant adverse effects. According to Wild et al. (2003) and Waljee et al. (2016), these interventions effectively decrease intestinal inflammation by promptly reducing intestinal permeability, suppressing the production of Tumour necrosis factor (TNF) and inhibiting Nuclear factor-κB (NF-κB).

3.3 Immunomodulators

Immunomodulators represent a category of pharmaceutical agents that exert a modulatory or regulatory effect on the immune system. Patients who exhibit poor responses to aminosalicylates or corticosteroids are frequently prescribed these medications. Additionally, they may be used as a maintenance therapy to mitigate the occurrence of flare-ups., mercaptopurine (MP), Azathioprine (AZT) and methotrexate (MTX) are among the immunomodulators commonly employed in the treatment of ulcerative colitis. These pharmaceutical agents function by inhibiting the immune response and diminishing inflammation within the colon. According to Singh et al. (2022), in cases where patients with IBD do not respond to 5-ASA drugs and are reliant on or unresponsive to corticosteroids, it is advised to utilise conventional immunomodulators such as 6-MP, AZT, and MTX to maintain remission.

3.4 Biologic therapies

Biologic therapies are a relatively new class of pharmaceutical interventions that specifically target specific molecules or cells involved in the inflammatory pathway of ulcerative colitis. These interventions are usually recommended for people with moderate-to-severe symptoms who have not responded to conventional treatment. Biological therapeutics, such as anti-tumour necrosis factor (TNF) agents (e.g., adalimumab & infliximab) or integrin receptor antagonists (e.g., vedolizumab), function by inhibiting specific proteins or cells involved in the inflammatory processes taking place in the gastrointestinal tract. In contemporary medical practice, infliximab and adalimumab are the prevailing anti-tumour necrosis factor-alpha (TNF- α) agents that are frequently employed. The administration of these medications occurs through intravenous and subcutaneous routes, respectively. According to Seo and Chae (2014), clinical trials have provided evidence supporting the efficacy of infliximab and adalimumab in the treatment of moderate-to-severe UC.

3.5 Surgical interventions

Surgical interventions are necessary for treating ulcerative colitis when severe symptoms arise, complications arise, or conventional treatments are ineffective. Proctocolectomy involves removing the colon and rectum, with or without an ostomy. This procedure has high efficacy in eradicating the afflicted colon, providing patients with a prolonged period of remission. Treatment selection depends on the severity and scope of ulcerative colitis, as well as individual patient factors and preferences. (Kuhn and Klar, 2015).

4 Antibiotics used in UC treatment

Rifaximin modifies gut microbiota for small intestinal bacterial overgrowth and hepatic encephalopathy, while antibiotics aim to manipulate intestinal flora, potentially disrupting IBD progression. In their study, Khan et al. (2011) conducted a meta-analysis that examined the findings of 10 randomised controlled trials (RCTs) focused on the utilisation of antibiotics in patients diagnosed with active CD. Additionally, the study also included nine RCTs that investigated the impact of antibiotic usage in individuals with UC. Antibiotics significantly impact remission and disease severity in UC and CD. Rifamycin derivatives are effective in treating active CD. However, results vary due to different trials involving rifaximin, fluoroquinolones, macrolides, and anti-tuberculosis antibiotics. (Steinhart et al., 2002).

5 The role of gut microbiota in UC

The gut microbiota, an important group of microorganisms that live in the digestive tract, is correlated to several diseases, including ulcerative colitis. The composition and function of the microbiota in the gut have both been shown to be susceptible to change, as recent research has shown. The gastrointestinal tract of a neonate does not initially contain any microorganisms but is eventually colonised by bacteria that come from the mother and the environment. (Dominguez-Bello et al., 2010). According to Vyas and Ranganathan (2012), the number of bacterial cells present on and within the entire body of an adult human is approximately ten times greater than the total number of human cells. The complexity and diversity of the human microbiome are considerable. The composition and quantity of anatomical features vary across different regions of the gastrointestinal tract, ranging from the nasal and oral cavities to the distal segments of the colon and rectum. The composition and complexity of the gut microbiota exhibit variability during the transition of infants from a liquid diet to solid foods. The alterations in dietary patterns during adulthood play a substantial role in shaping the composition of the gut microbiota. Metagenomic techniques based on 16S ribosomal RNA sequence have improved our understanding of microbial communities in the gastrointestinal tract. This approach reveals that 90% of the bacterial population is attributed to Bacteroidetes and Firmicutes. (Mariat et al., 2009; Hu and Gubatan, 2023).

5.1 Dysbiosis in UC

Dysbiosis in the gut microbiota is the root cause of ulcerative colitis. This condition is characterised by a decrease in commensal bacteria such as bifidobacteria and Lactobacilli and an increase in pathogenic bacteria such as Escherichia coli and Fusobacterium. The imbalance that can result from dysbiosis can put a person at risk for several different diseases, including IBD and irritable bowel syndrome. Additionally, it has the potential to bring about unanticipated outcomes, such as the activation of an HIV infection or the development of autoimmune diseases. (Liu et al., 2016).

5.2 Impaired gut barrier function

The gut microbiota and intestinal epithelial cells collaborate to establish a defensive barrier aimed at impeding the infiltration of pathogens and toxins into the bloodstream. The integrity of the barrier function in ulcerative colitis is compromised, resulting in heightened permeability of the intestinal lining. The presence of dysbiosis in individuals with ulcerative colitis has been observed to have a detrimental effect on the integrity of the gut barrier. This compromised barrier allows for the infiltration of pathogenic bacteria and their associated metabolic by-products into the intestinal mucosa, subsequently eliciting an immune response. This phenomenon gives rise to atypical immune-inflammatory responses, including inflammation, allergies, and autoimmune disorders, which are facilitated by molecular mimicry and dysregulated T-cell reactions. According to the study conducted by Barnaba and Sinigaglia, 1997, it was found that the maintenance of intestinal homeostasis and the differentiation between harmful pathogens and beneficial commensal microbes heavily rely on the Treg/TH17 ratio, which represents the equilibrium between regulatory T cells (Tregs) and T helper type 17 cells (TH17). This regulation is strongly linked to the gut microbiota, as commensal microorganisms, including Firmicutes, Bacteroides fragilis and Bifidobacterium infantis promote the expansion of Treg cells, specifically FOXP3-expressing Treg and IL-10-producing Treg lymphocytes. Foxp3+ regulatory T cells play a crucial role in gut immunity and physiology, inducing intestinal tolerance and defending the host against harmful dietary antigens, commensal microorganisms, and pathogens. They also facilitate local tissue repair and maintain the integrity of the epithelial barrier, making them essential non-immune cells in the gastrointestinal tract. (Cosovanu and Neumann, 2020).

5.3 Immune system dysregulation

The gut microbiota exerts a substantial impact on the maturation and modulation of the immune system. Individuals diagnosed with UC exhibit an atypical immune response towards the gut microbiota, which is distinguished by an intensified activation of the immune system and the persistence of inflammatory processes. The dysregulation of the immune system can sustain the ongoing cycle of inflammation and subsequent tissue damage in the colon, thereby playing a role in the advancement of ulcerative colitis. The dysregulation of the immune response in the intestinal region is a crucial factor in the development of IBD, involving various molecules such as cytokines. In a recent study conducted by Kmiec et al. (2017), it was demonstrated that there is a correlation between the innate immune response and the promotion of gut inflammation in patients with IBD. The mucosa of patients with UC exhibits an altered equilibrium between regulatory T-cells and effector T-cells, including Th1, Th2, and Th17 cells. Evidence suggests that there is a correlation between UC and an a typical type 2 immune response, which is mediated by non-classical natural killer T-cells that produce IL-5 and IL-13. Previous studies have demonstrated that IL-13, which is secreted by specific subsets of NKT cells, plays a crucial role in exerting cytotoxic effects on epithelial cells. These effects include the induction of apoptosis and alterations in the composition of tight junction proteins (Fuss and Strober, 2008; Heller et al., 2008). Poggi et al. (2019) demonstrated a correlation between the exacerbation of pathology and the presence of supplementary inflammatory cytokines, including TNF, IL-1, IL-6, and IL-9.

5.4 Role of short-chain fatty acids

Short-chain fatty acids (SCFAs) are one of many metabolites produced by the gut microbiota during the fermentation of dietary fibres,exhibit anti-inflammatory properties. These fatty acids, typically not exceeding six carbon atoms, are derived from prebiotic or microbially fermentable carbohydrates like inulin, polysaccharides, and resistant starch. (Dalile et al., 2019) Butyrate, acetate, and propionate are examples of SCFAs that play important roles in intestinal health.The observed effects include acceleration of the regeneration and healing process of intestinal epithelial cells, augmentation of mucus production, and maintenance of appropriate pH levels within the intestine. Moreover, they hinder the attachment of pathogenic microorganisms to enterocytes. Acetate is commonly utilised as a cellular energy substrate for the growth and maintenance of muscle tissue and colonic cells. Butyrate exhibits a variety of beneficial effects on the host, such as strengthening metabolism, modulating the host’s immune system, and facilitating anti-inflammatory mechanisms. As a result, it attracts considerable attention. (Guarino et al., 2020) One of the hallmarks of UC is a decrease in SCFAs production, which contributes to an already severe inflammatory response and jeopardises the GI tract’s overall health. Because of this decline in SCFAs producers, it is common to find lower levels of SCFAs in faeces taken from people with inflammatory IBD. In a study of people with UC, Machiels et al. (2013) found lower levels of acetate and propionate in their faeces but not butyrate. When studying faecal samples from people with IBD Huda-Faujan et al. (2010) found lower levels of butyrate and propionate. Recent research findings indicate notable disparities in gut microbial species, microbial diversity and metabolic pathways between individuals diagnosed with IBD and healthy individual. (Lavelle and Sokol, 2020)

5.5 Potential therapeutic strategies

Due to the significant impact of the gut microbiota on ulcerative colitis, there is a growing interest in the development of effective strategies aimed at modulating the microbiota to restore equilibrium and improve disease outcomes. This involves the utilisation of probiotics, which are live beneficial bacteria that can be employed to reinstate the equilibrium of microorganisms in the gastrointestinal tract. Prebiotics, classified as dietary fibres that possess the ability to selectively enhance the proliferation of advantageous bacteria, can additionally be employed to enhance the growth of beneficial microorganisms. In addition, the utilisation of synbiotics, a combination of probiotics and prebiotics, may exhibit synergistic outcomes in the restoration of equilibrium within the gut microbiota (Hu and Gubatan, 2023). The present review provides an overview of the existing microbiome-centred therapeutic strategies employed in the management of IBD, as outlined in Table 1.

TABLE 1
www.frontiersin.org

Table 1 Summary of current microbiome-based therapeutic strategies in IBD.

6 Probiotics: restoring balance in the gut

Probiotics can be traced back to ancient civilisations when humans first began eating fermented foods as part of their regular diet. It was hypothesised by Elie Metchnikoff that the beneficial bacteria found in yoghurt could improve the health of the gut microbiome. Mackowiak (2013) presented the concept of probiotics for the first time. These microorganisms can positively modulate the composition of the gut microflora and reduce the presence of pathogenic bacteria that release harmful compounds within the human gastrointestinal tract. In previous studies, a range of microorganisms have been employed for disease management, thereby giving rise to the term “probiotics”. Probiotics, derived from Greek, refer to living, non-pathogenic organisms with beneficial effects on hosts. Vergin, in 1954, coined the term “probiotic” to compare the harmful effects of antibiotics on intestinal microbiota with the beneficial effects of some beneficial bacteria. The term “probiotic” was subsequently redefined by Lilly and Stillwell as a biologically active product produced by a microorganism that enhances the growth of another microorganism. As a result, the term was subsequently defined by Fuller (1989) as non-pathogenic microorganisms that, when ingested, have a beneficial impact on the health or physiology of the host. The most recent definition proposed by the FDA and WHO in collaboration is that of a live microorganism that, when administered in adequate quantities, confers a health advantage to the host. Several probiotic microorganisms are commonly utilised, species of Bifidobacterium and Lactobacillus, have been reported to be the predominant and subdominant groups among gut microbes, respectively (Casellas et al., 2007; Markowiak and Śliżewska, 2017). Bacterial organisms capable of forming spores, primarily belonging to the genus Bacillus, are prevalent in this context (Derikx et al., 2016; Jin et al., 2023).

Probiotics are incorporated into various food products, particularly fermented dairy products, either individually or in conjunction with other strains. Continuously, novel genera and strains of probiotics are being discovered through increasingly sophisticated and targeted research endeavours. VSL#3 is a combination of eight distinct probiotic strains, exhibiting high strain specificity and variability in benefits across different patient groups. Probiotic products can consist of a solitary strain or a mixture of two or more strains, with the effectiveness of multi-strain probiotics demonstrated in controlled studies. (Chapman et al., 2011). The study of probiotics, particularly Lactobacilli, has experienced significant growth over the past twenty years. This is evident from the substantial increase in research articles published on the topic. Specifically, while there were approximately 180 research articles available between 1980 and 2000, the number of articles on the probiotic Lactobacillus exceeded 5700 between 2000 and 2014. According to a study published in PubMed in 2014, the effectiveness of the probiotic strain Lactobacillus was investigated. The characteristics of an ideal probiotic strain are depicted in Figure 1.

FIGURE 1
www.frontiersin.org

Figure 1 Characteristics of an ideal probiotic strain.

Probiotics act against intestinal diseases through various processes, including colonisation, which enhances their efficacy. They produce inhibitory constituents like organic acids, fatty acids, hydrogen peroxide, SCFAs, and bacteriocin-like inhibitory substances that prevent pathogens. (Tharmaraj and Shah, 2009). Bacteriocins refer to antimicrobial peptides or proteins that are produced by bacteria via ribosomal mechanisms. The aforementioned compounds demonstrate a diverse spectrum of antimicrobial efficacy against pathogenic bacteria, encompassing multiple strains of Staphylococcus, Listeria, Bacillus, Clostridium, and other bacterial species (Dai et al., 2021; Fathizadeh et al., 2022). The significance of bacteriocins synthesised by probiotic bacteria is ascribed to their secure utilisation in diverse domains, including the food industry, pharmaceuticals, veterinary medicine, and human healthcare (Barcenilla et al., 2021; Sharma et al., 2021; Somashekaraiah et al., 2021). The compounds cited in the scholarly articles (Sheoran and Tiwari, 2019; Sheoran and Tiwari, 2020; Chen et al., 2021) exhibit inhibitory effects on a wide range of bacteria. Their mechanism of action involves reducing cell viability, modifying bacterial cell metabolism, and inhibiting toxin production. Additional mechanisms of probiotic action include competitive inhibition on the surface of the intestinal epithelium, wherein the probiotics obstruct the binding sites of the intestinal epithelial surface, as well as a reduction in the interaction between the pathogen and the host. Similarly, the investigation of nutrient competition is also examined as a mechanism underlying probiotic activity. According to the suggestions of the World Health Organization (WHO), Food and Agriculture Organization (FAO), and the European Food Safety Authority (EFSA), in their selection process, probiotic strains must meet both safety and functionality criteria, as well as those related to their technological usefulness. Probiotic characteristics are not associated with the genus or species of a microorganism, but with few and specially selected strains of a particular species (Hill et al., 2014). FAO and WHO have jointly developed guidelines that propose a systematic approach for conducting comprehensive evaluations of probiotics in food products, to provide scientific evidence to support health claims and benefits. The essential criteria for an ideal probiotic are presented by Pandey et al. (2015). The application of the FAO/WHO guidelines on Probiotics has the potential to serve as a universal benchmark for evaluating probiotics in food products, thereby facilitating the validation of health-related assertions.

The guidelines stipulate that the following activities must be carried out:

The identification and categorisation of strains.

• A comprehensive elucidation of the strain(s) about their safety and probiotic attributes.

• Verification of the health benefits observed in human studies.

• Ensuring the accuracy and transparency of efficacy claims and product information throughout the entire duration of the product’s shelf life.

6.1 Probiotics in ulcerative colitis

The colon contains the highest concentration of microorganisms, suggesting treating colon microbiome abnormalities could benefit individuals with ulcerative colitis. Studies have shown potential benefits from various probiotic strains. (Basso et al., 2019). Previous studies have demonstrated that the use of the non-pathogenic strain of E. coli Nissle 1917 (EcN) exhibited similar efficacy and safety characteristics in the maintenance therapy of patients with mild or moderate ulcerative colitis when compared to treatment with salicylates (Basso et al., 2019; Bischoff et al., 2020). The potential therapeutic and prophylactic effects of the probiotic strain EcN against adherent-invasive Escherichia coli (AIEC) infection in zebrafish were investigated. In the zebrafish model, the researchers discovered that EcN effectively reduced AIEC colonisation, tissue damage, and pro-inflammatory responses. Furthermore, EcN reduced AIEC hyperinfection in zebrafish, especially when propionic acid was present. The effectiveness of EcN in combating AIEC infection in a zebrafish model is highlighted in this study. (Nag et al., 2022) The utilisation of various strains of lactic acid bacteria and bifidobacteria as supplementary treatment has been observed to significantly improve the progression of the disease and the sustained absence of symptoms in individuals diagnosed with ulcerative colitis (Basso et al., 2019). A highly regarded probiotic complex is VSL#3, which consists of four strains of Lactobacillus (L. casei, L. acidophilus, L. plantarum and L. delbrueckii subsp. bulgaricus), three strains of Bifidobacterium (B. longum, B. breve, and B. infantis), and one strain of Streptococcus (S. salivarius subsp. termophilus) (Basso et al., 2019). Studies conducted on mouse models have demonstrated that the administration of this probiotic mixture leads to the suppression of NF-B and TNF expression in the TLR4-NF-B signalling pathway. Consequently, the downregulation of pro-inflammatory cytokines and toll-like receptors (TLR) occurs while the upregulation of regulatory cytokines is observed (Jakubczyk et al., 2020; Silva et al., 2020). Previous studies have demonstrated the efficacy of VSL#3 in inducing and sustaining remission in individuals with mild to moderate UC when used as adjuvant therapy or as a standalone treatment (Basso et al., 2019).

In a systematic review and meta-analysis conducted by Derwa et al. (2017), it was found that the VSL#3 probiotic mixture demonstrated potential efficacy in inducing remission among individuals with UC. Furthermore, the study suggested that the use of VSL#3 probiotics may be comparable to 5-5-ASA in terms of preventing disease exacerbations. Under the recommendations put forth by the European Society for Clinical Nutrition and Metabolism (ESPEN), the investigation of probiotics such as VSL#3 and EcN in the treatment of mild to moderate UC is warranted, as they have shown potential for inducing remission in affected patients. According to Bischoff et al. (2020), the use of probiotics is contraindicated in cases of severe ulcerative UC. The use of probiotics as a supplementary treatment may have particular efficacy in the management of patients who experience intolerance to 5-ASA (Miele et al., 2018; Bischoff et al., 2020). Table 2 presents several clinical trial findings regarding the use of probiotics for the treatment of UC.

TABLE 2
www.frontiersin.org

Table 2 Some clinical trial data of probiotics for treating UC.

7 Prebiotics: nourishing beneficial bacteria

The term “prebiotics” is a relatively recent concept that is clearly defined as “nondigestible food ingredients that beneficially alter the host by selectively stimulating the growth and/or activity of a specific group of bacteria in the colon, thereby improving the overall health of the host” (Gibson and Roberfroid, 1995). Recently, the International Scientific Association for Probiotics and Prebiotics (ISAPP) convened a panel of experts to reassess the definition of prebiotics. The panel has expanded the definition to encompass a broader understanding, defining it as “a substrate that is selectively utilised by host microorganisms, resulting in a health benefit.” The definition presented by Gibson et al. (2017) expands upon the concept of prebiotics by considering the potential inclusion of non-carbohydrate substances, exploring their relevance to body sites beyond the gastrointestinal tract, and identifying distinct groups beyond those found in food. These substances are typically classified as short-chain carbohydrates that are resistant to digestion yet serve as substrates for the proliferation of probiotic microorganisms within the upper gastrointestinal tract. Cocoa-derived flavanols are compounds that are not classified as carbohydrates but have been proposed as potential prebiotics. According to Tzounis et al. (2011), experimental studies conducted both in vivo and in vitro demonstrate that flavanols can enhance the proliferation of lactic acid bacteria.

A significant increase in the Bifidobacteria population in faecal samples has been reported due to fructooligosaccharides (FOS) consumption. Prebiotics, consisting of glucose, fructose, galactose and/or xylose, undergo minimal hydrolysis in the intestinal tract and have caloric value due to their resistance to digestion and energy metabolism through fermentation (Roberfroid, 1993). Prebiotics like FOS, isomalto-oligosaccharides (IMO), and xylooligosaccharides (XOS) are studied for their potential benefits on stool volume, constipation relief, and faecal acidity. FOS, like inulin and neosugar, are dietary fibres that improve stool volume and faecal acidity. These prebiotics are easily metabolised by bifidobacteria and other microorganisms like Lactobacillus acidophilus, Bacteroides vulgatus, B. ovatus, B. thetaiotaomicron, B. fragilis, and Enterococcus faecium, E. faecalis (Guarino et al., 2020) An optimal prebiotic substance should possess several key characteristics, as illustrated in Figure 2. IMO can be identified in various fermented food products, including miso, soy sauce, and honey. These compounds are known to be metabolised by bifidobacteria and the Bacteroides groups. IMO facilitates the proliferation of Bifidobacterium and Lactobacillus species, leading to both local and systemic Th1-like immune responses and the regulation of immune function. The presence of positive outcomes has been identified in the clinical trials. XOS are a class of prebiotics that occur naturally in various sources such as fruits, bamboo shoots, vegetables, milk, honey, and others (Aachary and Prapulla, 2010). Previous research has indicated that Bifidobacterium adolescentis can utilise XOS, while, Lactobacillus rhamnosus, L. plantarum and Lactococcus lactis have been found to efficiently metabolise oat -glucooligosaccharides. According to Van Laere et al. (2000), the fermentation process of arabinose-XOS derived from wheat meal by bifidobacteria can be attributed to the existence of xylanolytic enzyme systems such as xylosidase and a limited number of arabinosidases. The presence of a -D-xylosidase derived from Bifidobacterium breve K-110, as well as arabinosidases originating from B. adolescentis DSM20083 (Van Laere et al., 2000) and B. breve (Shin et al., 2000), has been documented in the literature. Zeng et al. (2007) conducted a study on the species B. bifidum, B. adolescentis, and B. infantis, and reported that these species exclusively exhibited arabinosidase and xylosidase activity, while no activity of, glucuronidase, or acetyl xylan esterase and xylanase was observed. Gullon et al. (2008) studied the fermentation process of XOS from rice husks using probiotic bacteria. They found that Bifidobacterium adolescentis CECT 5781 showed significantly greater growth compared to Breve CECT 4839, Bifidobacterium longum CECT 4503 and Infantis CECT 4551 in the presence of XOS. This highlights the importance of industrial development in prebiotics for enhancing food quality and human health.

FIGURE 2
www.frontiersin.org

Figure 2 Characteristics of prebiotic.

A variety of functional foods containing prebiotics have been utilised in the production of biscuits, candies, tabletop sweeteners, frozen yoghurt, and other similar products (Davani-Davari et al., 2019). In Japan, the Foods for Specified Health Uses (FOSHU) programme has incorporated various prebiotics, such as FOS, IMO, XOS, lacto-sucrose, and lactulose oligosaccharides, into specific food products. Additional examples of prebiotic foods include yoghurts, cereals, cakes; cereal bars biscuits high in dietary fibre, powdered beverages, pasta, sauces, bread, infant formula products and various fruit juices (Desai, 2008). Prebiotics are commonly associated with specific types of dietary fibre, such as inulin and FOS. However, they can also be found in various food sources, including cultivated plants and wild plants. Below are several examples of food plants that are abundant in prebiotics; categorised accordingly as cultivated and wild plants.

Cultivated plants

• Bananas: Bananas contain FOS, which is a type of prebiotic that is fermented by beneficial bacteria in the gut. FOS has been shown to advance gut health, decrease inflammation, and improve the immune system. (Sabater-Molina et al., 2009)

• Garlic: Garlic contains inulin, a prebiotic fibre that nourishes gut bacteria. It is also known for its antimicrobial properties. (Davani-Davari et al., 2019)

• Legumes: Legumes, such as beans, lentils, and peas, are excellent sources of resistant starch, a type of prebiotic that is fermented by beneficial bacteria in the gut. Resistant starch has been shown to increase gut health, decrease the risk of chronic diseases, and benefit weight management. (Mirmiran, 2014)

• Oats: Oats are a great source of beta-glucans, a category of soluble fibre that is fermented by beneficial bacteria in the gut. Beta-glucans has been shown to improve gut health, reduce cholesterol levels, and help with weight management. (Sabater-Molina et al., 2009)

• Whole grains: Whole grains, such as brown rice, quinoa, and barley, are excellent sources of resistant starch and other prebiotics. (Sabater-Molina et al., 2009)

• Onion: Like garlic, onions are rich in inulin and can promote the growth of helpful gut bacteria. (Davani-Davari et al., 2019)

Asparagus: Asparagus is a good source of inulin, a type of soluble fibre that is fermented by beneficial bacteria in the gut. Inulin has been shown to improve gut health, boost immunity, and decrease the risk of chronic disorders. (Sabater-Molina et al., 2009)

Jerusalem artichoke: Jerusalem artichokes are high in inulin content, making them an excellent prebiotic food. (Sabater-Molina et al., 2009)

• Chicory Root: Chicory root is often used as a source of inulin in supplements, which is proven to increase the growth of Bifidobacterium. It can be consumed raw or roasted (Davani-Davari et al., 2019).

Wild Plants:

Taraxacum officinale (Dandelion greens): A very common plant, dandelion grows wild almost everywhere. Dandelion is a native of Europe. In India, it is found in the Himalayas. Nutritionally, the dandelion has remarkable value. Dandelion greens are wild plant that contains inulin and other prebiotic fibres, along with various nutrients (Wirngo et al., 2016).

Arctium lappa L (Burdock Root): Burdock root is a wild plant that contains inulin and has been traditionally used as a medicinal food with prebiotic properties. This species is native to the temperate regions of the Old World, from Scandinavia to the Mediterranean, and from the British Isles through Russia, and the Middle East to India, China, Taiwan and Japan. (Moro and Clerici, 2021).

Cichorium intybus L (Chicory Greens): Wild chicory greens are rich in inulin and can be consumed in salads or cooked as a side dish. The Chicory crop is cultivated in a few States, mostly Uttar Pradesh, and Gujarat. These two states account for 97% of the total production of Chicory in India (Nwafor et al., 2017).

Musa paradisiaca (Plantain): Plantain leaves are a common wild plant that contains prebiotic fibres, among other beneficial compounds. Plantain fruit is widely consumed in Nigeria, Africa and some other parts of the world (Ukwah et al., 2014).

Urtica Dioica (Nettles): Nettles are known for their numerous health benefits and can provide prebiotic effects due to their fibre content. It is most common in Europe, North America, North Africa, and parts of Asia (Behzad et al., 2018).

These are just a few examples, and there are many other cultivated and wild plants that can offer prebiotic benefits. Combining a type of these prebiotic-rich foods into your diet can aid in promoting a healthy gut microbiome (Crittenden and Playne, 2008; Davani-Davari et al., 2019). For a product (food or supplement) to be believed to be a prebiotic, it must meet the below-mentioned conditions (Gibson et al., 2017).

Prebiotic enhance the proliferation and metabolic function of selected bacterial strains that exhibit beneficial impacts on overall well-being in the following ways.

• Lower the pH of the substances presents in the intestines.

• To exhibit resistance to hydrolysis and the effects of gastrointestinal enzymes.

• To avoid being absorbed in the upper gastrointestinal tract.

• To maintain a suitable environment for one or more beneficial microorganisms within the colon.

• To maintain stability throughout the food processing procedure,

7.1 Prebiotics in UC

Prebiotics, mostly fermentable carbohydrates, promote local or systemic health (Pandey et al., 2015; Akram et al., 2019). Prebiotics can alter the intestinal microbiota, improve the intestinal barrier, and promote the growth of beneficial microbes in the digestive tract, which produce host-beneficial metabolites. (Aljuraiban et al., 2023) Like probiotics, clinical trials on prebiotics in specific diseases are difficult to prove. Thus, prebiotic data in UC patients is carefully regulated. Prebiotics may help treat UC by supplementing fermentable carbohydrate-containing fibre fractions. Promoting specific bacteria and/or their metabolites achieves this (Pandey et al., 2015; Rasmussen and Hamaker, 2017). Prebiotics may help maintain remission or low clinical disease activity. Prebiotics used in UC studies are oligosaccharides and inulin (Rasmussen and Hamaker, 2017). The mechanism for the ameliorative effect of prebiotics in UC studies is illustrated in Figure 3. Prebiotics encourage the growth of beneficial microorganisms, which compete with harmful species and produce beneficial fermentation substances like SCFAs, which have immunomodulatory properties and influence toll-like receptor-4 signalling and pro-inflammatory cytokines. (Van der Beek et al., 2017)

FIGURE 3
www.frontiersin.org

Figure 3 Mechanism for ameliorative effect of prebiotics in UC studies.

Casellas et al. (2007) studied the effects of mesalazine treatment, oligofructose-enriched inulin, and placebo on patients with mild to moderate UC. Oral oligofructose-enriched inulin was well-received and reduced faecal calprotectin levels. Faecal calprotectin has been employed as a reliable and quantifiable indicator of intestinal inflammation, with a significant correlation observed between its levels and the assessment of disease activity in UC, as determined through histologic and endoscopic methods (Konikoff and Denson, 2006).In Japan, germinated barley food (GBF) has been explored as a therapeutic intervention for UC. GBF, a dietary fibre and protein-rich in glutamine, has been shown to reduce clinical activity in individuals with UC and is an effective treatment option for maintaining remission. The safety profile of the treatment is noteworthy, as the study conducted by Hanai et al. (2004) found no incidence of side effects in the study group associated with the use of GBF. Nevertheless, further clinical trials are required to provide additional confirmation regarding the efficacy of dietary fibre as a prebiotic in the management of UC. Prebiotics has been extensively utilised in the management and treatment of UC. Table 3 presents a comprehensive compilation of various studies that have examined the advantageous effects of prebiotics in the management of UC.

TABLE 3
www.frontiersin.org

Table 3 Studies of prebiotics in the management of UC.

8 Synbiotics

Gibson’s proposition of prebiotics was accompanied by a rationale for the potential advancements that could be achieved through the combination of prebiotics with probiotics, leading to the establishment of what he referred to as Synbiotics (De Vrese and Schrezenmeir, 2008). The term “synbiotics” pertains to the amalgamation of prebiotics and probiotics to improve the well-being of humans or animals (Markowiak and Śliżewska, 2017). According to research conducted by Perrin et al. (2001) and Sharma and Shukla (2016), the probiotic bacteria found in synbiotic food products make selective use of prebiotics as a substrate for their growth. A group of specialists from the International Scientific Association for Probiotics and Prebiotics got together and rethought the concept of synbiotics. Synbiotic treatment aims to improve the endurance and metabolic function of beneficial probiotic strains in the gut microbiota. Common combinations include lactobacilli and bifidobacteria with oligosaccharides, inulin, or fibres as prebiotics. This approach reduces systemic inflammation by increasing the population of bacteria producing SCFAs and providing substrates for fermentation (Pandey et al., 2015).

According to their classification, synbiotics can be categorised into two distinct classes: complementary and synergistic. A complementary synbiotic is comprised of a probiotic and a prebiotic, which collectively provide one or more health benefits without requiring interdependent functions. According to Swanson et al. (2020), a synergistic synbiotic is composed of a substrate that is specifically utilised by microorganisms that are co-administered. These guidelines are expected to be efficacious in establishing expectations regarding the comprehension of the interplay between pre- and probiotics as well as the advancement of synbiotic products for the promotion of health and therapeutic interventions. Numerous reports suggest that the consumption of synbiotic foods has a beneficial impact on the health and nutritional status of the host. The study conducted by Yang et al. (2005) revealed that the administration of synbiotics had a dual effect on the faecal sample. Firstly, it was observed that synbiotics increased the abundance of probiotic bacteria, specifically Lactobacillus and Bifidobacterium, while simultaneously reducing the presence of coliform bacteria. Additionally, the test group exhibited an improvement in the levels of various digestive enzymes, including lactase, sucrase, lipase and isomaltase. Recently, there has been a report indicating that the consumption of synbiotics has significantly decreased various cardiovascular risk factors, the prevalence of metabolic syndrome, and markers of insulin resistance among elderly individuals (Cicero et al., 2021). The ability of bifidobacteria to metabolise prebiotics is dependent on the species, which is a valuable characteristic for modulating the gut microbiota through the use of specialised prebiotics (Bielecka et al., 2002; Biedrzycka and Bielecka, 2004). The β-fructofuranosidase enzyme derived from Bifidobacterium adolescentis G1 exhibits a preference for fructooligomers over inulin as its substrate. This preference is also observed in B. bifidum. In contrast, it has been observed that B. longum and B.animalis possess the ability to hydrolyze a diverse array of FOS and XOS, including those derived from inulin (Bruno et al., 2002). According to a study conducted by Bruno et al. (2002), food products contain the greatest concentration of viable bifidobacteria. Likewise, the efficacy of B. longum supplemented with FOS is greater in curd. According to Martinez-Villaluenga and Gomez (2007), the probiotic B. lactis possesses the enzymes β-glucosidase and β -fructofuranosidase, which enable it to metabolise oligosaccharides present in fermented milk. This metabolic activity promotes the growth and metabolism of the probiotic organism. The therapeutic potentials and health benefits of different probiotics, prebiotics, and synbiotics are presented in Table 4.

TABLE 4
www.frontiersin.org

Table 4 Therapeutic potential and health benefits of probiotics, prebiotics and synbiotics.

8.1 Synbiotics in ulcerative colitis

Synbiotics are products that combine probiotics and prebiotics, resulting in a synergistic interaction. This concept was introduced to address challenges in probiotic survival, especially during transit through the upper gastrointestinal tract. The use of a synbiotic enhances probiotic colonisation efficacy and facilitates the proliferation of probiotic strains (Pandey et al., 2015).

There is a limited body of research examining the impact of a synbiotic supplement on individuals diagnosed with UC. Table 5 presents a compilation of the studies that have been cited most frequently and illustrated in Figure 4. Wong et al. (2022) investigated the impact of prebiotic mixtures, probiotic mixtures, and synbiotics on colitis in a murine model. The findings of the study indicated that the administration of synbiotic treatment had a protective effect on the structure of the colon, as evidenced by the preservation of its integrity. Furthermore, there was an observed increase in the expression of occludin, a protein involved in maintaining the tight junctions between cells, which is indicative of improved barrier function. Additionally, the treatment was found to be effective in reducing the infiltration of cells into the colon and resulted in alterations to the gut microbiome, enhancements in colonic integrity, and the suppression of markers associated with inflammation. Although the existing studies have yielded promising results thus far, it is important to note that the methodologies employed often exhibit inconsistencies, lack proper description, and/or suffer from inadequate study design. Additionally, it is worth mentioning that the majority of cases have been characterised by a notably limited number of registered patients.

TABLE 5
www.frontiersin.org

Table 5 Summary of the most relevant studies involving synbiotics treatments in UC.

FIGURE 4
www.frontiersin.org

Figure 4 Use of synbiotics for ulcerative colitis treatment.

9 Safety and considerations in ulcerative colitis treatment

When it comes to treating ulcerative colitis, ensuring safety and considering various factors are of paramount importance. Here are some key safety considerations to keep in mind:

9.1 Medical supervision

Ulcerative colitis treatment should always be conducted under the guidance and supervision of a qualified healthcare professional. They will assess the severity of the condition, consider individual patient factors, and prescribe appropriate medications or therapies (Kerner et al., 2014).

9.2 Monitoring and follow-up

Regular monitoring is crucial to evaluating the effectiveness of the treatment and assessing any potential side effects or complications. Patients should attend follow-up appointments as scheduled and communicate any changes in symptoms or concerns to their healthcare provider (Click and Regueiro, 2019).

9.3 Medication safety

Patients need to follow the prescribed medication regimen diligently. They should be aware of potential side effects and know when to seek medical attention. Certain medications used in ulcerative colitis treatment, such as immunosuppressants or biologics, may require additional precautions and monitoring due to their impact on the immune system (Click and Regueiro, 2019).

9.4 Individualised approach

Each patient with ulcerative colitis is exceptional, and management plans should be designed to meet their special needs. Factors such as age, disease severity, comorbidities, and medication tolerance should be taken into consideration when developing a personalised treatment strategy (Kerner et al., 2014).

9.5 Lifestyle modifications

In addition to medication, lifestyle changes can play a major role in managing ulcerative colitis. These may include dietary changes, stress management techniques, regular exercise, and getting sufficient rest. Patients should work with healthcare professionals, such as dietitians and psychologists, to make appropriate lifestyle adjustments (Click and Regueiro, 2019).

9.6 Risk-benefit assessment

Every treatment decision involves a careful evaluation of the potential risks and benefits. Patients and healthcare providers should discuss the expected outcomes, potential side effects, and long-term implications of various treatment options. This shared decision-making process ensures that the chosen treatment aligns with the patient’s goals and preferences (Kerner et al., 2014).

9.7 Complementary and alternative therapies

If considering complementary or alternative therapies, patients should consult with their healthcare provider to ensure safety and assess potential interactions with conventional treatments. It is important to be cautious of unproven or unsupported therapies that may promise a cure without scientific evidence (Click and Regueiro, 2019).

9.8 Patient education and support

Patient education about their condition, treatment options, and self-care is essential. Ulcerative colitis patients can benefit from educational resources and support groups. In conclusion, ulcerative colitis treatment must prioritise safety and individual factors. Patients can manage their condition while minimising risks and maximising treatment benefits with medical supervision, regular monitoring, personalised approaches, and informed decision-making (Kerner et al., 2014).

10 Future perspectives and research

UC significantly impacts individuals’ health and quality of life, with treatment options including aminosalicylates, corticosteroids, immunomodulators, biological therapies, and surgical interventions. However, some patients have not responded well to these treatments, leading to prolonged suffering and reduced quality of life. Alternative therapies like experimental drugs are considered, but their effectiveness remains uncertain and comes with risks and complications. As we gain more understanding of the pathophysiology of UC and its relationship with the microbiome, there will be a rise in research trials to determine the efficacy of emerging treatments. Scholars are currently investigating “next-generation” probiotics, such as Clostridium clusters IV, XIVa, and XVIII, Bacteroides uniformis, Bacteroides fragilis, Akkermansia muciniphila, Eubacterium hallii, and Faecalibacterium prausnitzii as potential alternatives.

Collaborative engagement between patients and healthcare providers is crucial for determining the most suitable treatment approach. Emerging treatments like prebiotics, probiotics, and synbiotics have gained attention for their potential therapeutic benefits in improving gut health and treating various conditions. For patients with IBD, traditional medications may not always work for every patient, and their side effects can be severe. In clinical practice, incorporating probiotics, prebiotics, and synbiotics holds great promise for transforming the approach to managing ulcerative colitis, offering patients a safer and more focused alternative to conventional therapeutic interventions. This will facilitate our understanding of the interplay between human physiological processes and the microbiome.

11 Discussion

The search for a definitive solution for UC continues to be a persistent effort, with existing therapeutic interventions primarily focused on maintaining a state of remission. In the management of this condition, conventional therapeutic approaches encompass the utilisation of aminosalicylates, corticosteroids, immunomodulators, and biological therapies. These interventions are employed with the objectives of attaining remission, averting relapses, and facilitating the restoration of mucosal integrity. Current therapeutic approaches encompass the use of monoclonal antibodies that specifically target proinflammatory cytokines, adhesion molecules, and T-cell activation, as well as anti-inflammatory cytokines like IL10 and TGF-. However, the administration of these medications can lead to various adverse effects, both at the systemic and local levels. These effects may include headaches, nausea, abdominal pain, loss of appetite, vomiting, and the development of a rash. The use of corticosteroids for an extended period is discouraged due to the potential increase in susceptibility to infections (Mowat et al., 2011). In contemporary times, functional food is increasingly recognised for its dual capacity to ensure nutritional security and confer health benefits on the consumer (Mijan and Lim, 2018). Promising advancements have been observed in the utilisation of probiotics, prebiotics, and synbiotics for the management of UC. Bacterial strains, such as Lactobacillus and Bifidobacterium species, have been linked to beneficial outcomes concerning symptom management and the maintenance of remission in individuals with a diagnosis of UC. By encouraging the growth of advantageous microorganisms and producing anti-inflammatory fatty acids, prebiotics supports gut health and digestion. Non-digestible fibres in foods like whole grains, garlic, and bananas support gut flora, lowering the risk of gastrointestinal conditions like irritable bowel syndrome and enhancing immune function. Prebiotic-rich foods are an excellent way to support digestive health and uphold general wellbeing through diet. The synergistic effects of prebiotics and probiotics allow for the effective management of UC (Fujimori et al., 2009). Therefore, it is imperative to conduct a comprehensive examination of these interventions to effectively mitigate any potential negative consequences and develop strategies for long-term supervision. The management of UC can be tailored to the individual by combining conventional treatments with probiotics, prebiotics, and synbiotics. The evaluated combination therapies demonstrate encouraging synergistic effects through the simultaneous targeting of various aspects of disease pathogenesis, including inflammation, immune dysregulation, and gut microbiota imbalance. Nevertheless, it is of utmost importance to conduct a thorough assessment of treatment interactions, contraindications, and individual patient variables to guarantee the safety and effectiveness of the intervention. It is imperative to undertake additional research, employing meticulously crafted clinical trials, to ascertain the most advantageous strains, compositions, and therapeutic regimens for probiotics, prebiotics, and synbiotics within the particular framework of UC. The aforementioned advancements provide empirical support for the capacity to enhance therapeutic outcomes, alleviate adverse effects, and establish innovative and well-tolerated strategies for managing UC. The current literature on probiotics and prebiotics in IBD is influenced by significant heterogeneity, with varying study designs, doses, and outcomes. The study populations varied, with some focusing on active disease patients and others on remission maintenance. Most studies enrolled small numbers of patients, limiting statistical power. The exact mechanisms of action of probiotics, prebiotics, and synbiotics are not yet fully understood. Insufficient evidence on probiotic dosages and immunological mechanisms is needed to establish health claims. The interaction between microbiota, host, and prebiotic components is also limited. Clinical trials and validation studies with larger sample sizes require understanding of these interactions. Limited published literature in manufacturing processes and formulation further needs improvement. (Astó et al., 2019)

In conclusion, the utilisation of probiotics, prebiotics, and synbiotics in the management of UC showcases a promising advancement with potential implications. These interventions present innovative strategies for manipulating the gut microbiota, reducing inflammation, and improving overall health. Additional investigation is required to determine the most effective utilisation and potential incorporation of these interventions into individualised therapeutic approaches for managing ulcerative colitis. Through the adoption of innovative approaches in the treatment of UC and the consideration of individual patient requirements, we have the potential to advance towards a more optimistic future for individuals afflicted by this complex inflammatory bowel disease.

Author contributions

AJ: Conceptualization, Data curation, Writing – original draft. SJ: Conceptualization, Supervision, Validation, Writing – review & editing. SV: Writing – review & editing. AS: Writing – review & editing. BK: Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Acknowledgments

The authors are thankful to all the authorities of Bharati Vidyapeeth (Deemed to be a University) for their overall support and encouragement.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Aachary, A. A., Prapulla, S. G. (2010). Xylooligosaccharides (XOS) as an emerging prebiotic: Microbial synthesis, utilization, structural characterization, bioactive properties, and applications. Compr. Rev. Food Sci. Food Saf. 10, 2–16. doi: 10.1111/j.1541-4337.2010.00135.x

CrossRef Full Text | Google Scholar

Agraib, L. M., Yamani, M. I., Tayyem, R., Abu-Sneineh, A. T., Rayyan, Y. M. (2022). 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 51, 83–91. doi: 10.1016/j.clnesp.2022.08.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahmed, J., Reddy, B. S., Molbak, L., Leser, T. D., MacFie, J. (2013). Impact of probiotics on colonic microflora in patients with colitis: A prospective double blind randomised crossover study. Int. J. Surg. 11, 1131–1136. doi: 10.1016/j.ijsu.2013.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Akram, W., Garud, N., Joshi, R. (2019). Role of inulin as prebiotics on inflammatory bowel disease. Drug Discoveries Ther. 13, 1–8. doi: 10.5582/ddt.2019.01000

CrossRef Full Text | Google Scholar

Alatab, S., Sepanlou, S. G., Ikuta, K., Vahedi, H., Bisignano, C., Safiri, S., et al. (2020). The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 5, 17–30. doi: 10.1016/s2468-1253(19)30333-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Aljuraiban, G. S., Algabsani, S. S., Sabico, S., AlShammari, S., Aljazairy, E. A., AL-Musharaf, S. (2023). Types of fiber and gut microbiota composition and diversity among arab females. Saudi J. Biol. Sci. 30, 103767. doi: 10.1016/j.sjbs.2023.103767

PubMed Abstract | CrossRef Full Text | Google Scholar

Amiriani, T., Rajabli, N., Faghani, M., Besharat, S., Roshandel, G., Akhavan, T., et al. (2020). Effect of Lactocare® synbiotic on disease severity in ulcerative colitis: A randomized placebo-controlled double-blind clinical trial. Middle East J. Digestive Dis. 12, 27–33. doi: 10.15171/mejdd.2020.160

CrossRef Full Text | Google Scholar

Ananthakrishnan, A. N. (2015). Epidemiology and risk factors for IBD. Nat. Rev. Gastroenterol. Hepatol. 12, 205–217. doi: 10.1038/nrgastro.2015.34

PubMed Abstract | CrossRef Full Text | Google Scholar

Arribas, B., Rodriguez, C. M., Camuesco, D., Comalada, M., Bailon, E., Utrilla, P., et al. (2009). A probiotic strain of Escherichia coli, Nissle 1917, given orally exerts local and systemic anti-inflammatory effects in lipopolysaccharide-induced sepsis in mice. Br. J. Pharmacol. 157, 1024–1033. doi: 10.1111/j.1476-5381.2009.00270.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Astó, E., Méndez, I., Audivert, S., Farrán-Codina, A., Espadaler, J. (2019). The efficacy of probiotics, prebiotic inulin-type fructans, and synbiotics in human ulcerative colitis: A systematic review and meta-analysis. Nutrients (11), 293. doi: 10.3390/nu11020293

PubMed Abstract | CrossRef Full Text | Google Scholar

Bamola, V. D., Dubey, D., Samanta, P., Kedia, S., Madempudi, R. S., Neelamraju, J., et al. (2022). Effect of Bacillus coagulans unique is-2 in inflammatory bowel disease (IBD): A randomized controlled trial. medRxiv 78, 1–29. doi: 10.1101/2021.07.18.21260556

CrossRef Full Text | Google Scholar

Barcenilla, C., Ducic, M., Lopez, M., Prieto, M., Alvarez- Ordonez, A. (2021). Application of lactic acid bacteria for the biopreservation of meat products: A systematic review. Meat Sci. 183, 1–13.108661. doi: 10.1016/j.meatsci.2021.108661

CrossRef Full Text | Google Scholar

Barefoot, S. F., Klaenhammer, T. R. (1983). Detection and activity of lactacin B, a bacteriocin produced by Lactobacillus acidophilus. Appl. Environ. Microbiol. 45, 1808–1815. doi: 10.1128/aem.45.6.1808-1815.1983

PubMed Abstract | CrossRef Full Text | Google Scholar

Barnaba, V., Sinigaglia, F. (1997). Molecular mimicry and T cell–mediated autoimmune disease. J. Exp. Med. 185, 1529–1532. doi: 10.1084/jem.185.9.1529

PubMed Abstract | CrossRef Full Text | Google Scholar

Basso, P. J., Camara, N. O., Sales-Campos, H. (2019). Microbial-based therapies in the treatment of inflammatory bowel disease – an overview of human studies. Front. Pharmacol. 9. doi: 10.3389/fphar.2018.01571

CrossRef Full Text | Google Scholar

Behzad, K., Asghar, K., Mohammad, A. K., Rezvan, P. (2018). Probiotic yogurt formulated with nettle (Urtica dioica) extract, a compound with dual functionalities: Bifidobacterium growth promoter and Helicobacter pylori growth inhibitor. BioMed. J. Sci. Tech Res. 10, 7532–7538. doi: 10.26717/BJSTR.2018.10.001888

CrossRef Full Text | Google Scholar

Berrada, N., Lemeland, J.-F., Laroche, G., Thouvenot, P., Piaia, M. (1991). Bifidobacterium from fermented milks: Survival during Gastric transit. J. Dairy Sci. 74, 409–413. doi: 10.3168/jds.s0022-0302(91)78183-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Bibiloni, R., Fedorak, R. N., Tannock, G. W., Madsen, K. L., Gionchetti, P., Campieri, M., et al. (2005). VSL3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am. J. Gastroenterol. 100, 1539–1546. doi: 10.1111/j.1572-0241.2005.41794.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Biedrzycka, E., Bielecka, M. (2004). Prebiotic effectiveness of fructans of different degrees of polymerization. Trends Food Sci. Technol. 15, 170–175. doi: 10.1016/j.tifs.2003.09.014

CrossRef Full Text | Google Scholar

Bielecka, M., Biedrzycka, E., Majkowska, A. (2002). Selection of probiotics and prebiotics for synbiotics and confirmation of their in vivo effectiveness. Food Res. Int. 35, 125–131. doi: 10.1016/s0963-9969(01)00173-9

CrossRef Full Text | Google Scholar

Bischoff, S. C., Escher, J., Hebuterne, X., Klęk, S., Krznaric, Z., Schneider, S., et al. (2020). Espen practical guideline: Clinical nutrition in inflammatory bowel disease. Clin. Nutr. 39, 632–653. doi: 10.1016/j.clnu.2019.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Bjarnason, I., Sission, G., Hayee, B. (2019). A randomised, double-blind, placebo-controlled trial of a multi-strain probiotic in patients with asymptomatic ulcerative colitis and crohn’s disease. Inflammopharmacology 27, 465–473. doi: 10.1007/s10787-019-00595-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Britton, R. A., Hoffmann, D. E., Khoruts, A. (2021). Probiotics and the microbiome—how can we help patients make sense of probiotics? Gastroenterology 160, 614–623. doi: 10.1053/j.gastro.2020.11.047

PubMed Abstract | CrossRef Full Text | Google Scholar

Bruno, F. A., Lankaputhra, W. E. V., Shah, N. P. (2002). Growth, viability and activity of Bifidobacterium spp. in skim milk containing prebiotics. J. Food Sci. 67, 2740–2744. doi: 10.1111/j.1365-2621.2002.tb08807.x

CrossRef Full Text | Google Scholar

Buck, L. M., Gilliland, S. E. (1994). Comparisons of freshly isolated strains of Lactobacillus acidophilus of human intestinal origin for ability to assimilate cholesterol during growth. J. Dairy Sci. 77, 2925–2933. doi: 10.3168/jds.s0022-0302(94)77233-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Casellas, F., Borruel, N., Torrejon, A., Varela, E., Antolin, M., Guarner, F., et al. (2007). Oral oligofructose-enriched inulin supplementation in acute ulcerative colitis is well tolerated and associated with lowered faecal calprotectin. Alimentary Pharmacol. Ther. 25, 1061–1067. doi: 10.1111/j.1365-2036.2007.03288.x

CrossRef Full Text | Google Scholar

Çavdar, M., Çavdar, M. (2023). Alternative therapeutic applications used in the treatment of ulcerative colitis: probiotics, prebiotics, synbiotics and fecal microbiota transplantation. Prog. Nutr. doi: 10.23751/pn.v25i2.13775

CrossRef Full Text | Google Scholar

Chapman, C. M., Gibson, G. R. 0., Rowland, I. (2011). Health benefits of probiotics: Are mixtures more effective than single strains? Eur. J. Nutr. 50, 1–17. doi: 10.1007/s00394-010-0166-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Zhang, L., Hong, G., Huang, C., Qian, W., Bai, T., et al. (2020c). Probiotic mixtures with aerobic constituent promoted the recovery of multi-barriers in DSS-induced chronic colitis. Life Sciences 240, 117089. doi: 10.1016/j.lfs.2019.117089

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Jin, Y., Stanton, C., Ross, R. P., Wang, Z., Zhao, J., et al. (2020b). Dose-response efficacy and mechanisms of orally administered cla-producing Bifidobacterium breve CCFM683 on DSS-induced colitis in mice. J. Funct. Foods 75, 104245. doi: 10.1016/j.jff.2020.104245

CrossRef Full Text | Google Scholar

Chen, X., Liu, Y., Jin, J., Liu, H., Hao, Y., Zhang, H., et al. (2021). YbfA regulates the sensitivity of Escherichia coli K12 to Plantaricin BM-1 via the BasS/BasR two-component regulatory system. Front. Microbiol. 12. doi: 10.3389/fmicb.2021.659198

CrossRef Full Text | Google Scholar

Chen, P., Xu, H., Tang, H., Zhao, F., Yang, C., Kwok, L. Y., et al. (2020a). Modulation of gut mucosal microbiota as a mechanism of probiotics-based adjunctive therapy for ulcerative colitis. Microbial Biotechnol. 13, 2032–2043. doi: 10.1111/1751-7915.13661

CrossRef Full Text | Google Scholar

Cicero, A. F. G., Fogacci, F., Bove, M., Giovannini, M., Borghi, C. (2021). Impact of a short-term synbiotic supplementation on metabolic syndrome and systemic inflammation in elderly patients: a randomized placebo-controlled clinical trial. Eur. J. Nutr. 60, 655–663. doi: 10.1007/s00394-020-02271-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Click, B., Regueiro, M. (2019). A practical guide to the safety and monitoring of new IBD therapies. Inflammatory Bowel Dis. 25, 831–842. doi: 10.1093/ibd/izy313

CrossRef Full Text | Google Scholar

Cosovanu, C., Neumann, C. (2020). The many functions of Foxp3+ Regulatory T cells in the intestine. Front. Immunol. 11. doi: 10.3389/fimmu.2020.600973

PubMed Abstract | CrossRef Full Text | Google Scholar

Crittenden, R., Playne, M. J. (2008). “Nutrition News. Facts and functions of prebiotics, probiotics and synbiotics,” in Handbook of Probiotics and Prebiotics. Eds. Lee, Y. K., Salminen, S. (Hoboken, NJ, USA; Manhattan, KS, USA: Wiley-Interscience, Kansas State University), 2008 535–582.

Google Scholar

Cui, H. H., Chen, C. L., Wang, J. D., Yang, Y. J., Cun, Y., Wu, J. B., et al. (2004). Effects of probiotic on intestinal mucosa of patients with ulcerative colitis. World journal of gastroenterology 10, 1521–1525. doi: 10.3748/wjg.v10.i10.1521

PubMed Abstract | CrossRef Full Text | Google Scholar

Cui, L., Guan, X., Ding, W., Luo, Y., Wang, W., Bu, W., et al. (2021). Scutellaria baicalensis Georgi polysaccharide ameliorates DSS-induced ulcerative colitis by improving intestinal barrier function and modulating gut microbiota. Int. J. Biol. macromolecules 166, 1035–1045. doi: 10.1016/j.ijbiomac.2020.10.259

CrossRef Full Text | Google Scholar

Dai, M., Li, Y., Xu, L., Wu, D., Zhou, Q., Li, P., et al. (2021). A novel bacteriocin from Lactobacillus pentosus ZFM94 and its antibacterial mode of action. Front. Nutr. 8. doi: 10.3389/fnut.2021.710862

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalile, B., Van Oudenhove, L., Vervliet, B., Verbeke, K. (2019). The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol. 16, 461–478. doi: 10.1038/s41575-019-0157-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Davani-Davari, D., Negahdaripour, M., Karimzadeh, I., Seifan, M., Mohkam, M., Masoumi, S. J., et al. (2019). Prebiotics:definition, types, sources, mechanisms, and clinical applications. Foods (Basel Switzerland) 8, 92. doi: 10.3390/foods8030092

PubMed Abstract | CrossRef Full Text | Google Scholar

Derikx, L., Dieleman, L. A., Hoentjen, F. (2016). Probiotics and prebiotics in ulcerative colitis. Best Pract. Res. Clin. Gastroentrology 30, 55–71. doi: 10.1016/j.bpg.2016.02.005

CrossRef Full Text | Google Scholar

Derwa, Y., Gracie, D. J., Hamlin, P. J., Ford, A. C. (2017). Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease. Alimentary Pharmacol. Ther. 46, 389–400. doi: 10.1111/apt.14203

CrossRef Full Text | Google Scholar

Desai, A. R. (2008). Strain identification, viability and probiotics properties of Lactobacillus casei. Ph.D. thesis, School of Biomedical and Health Sciences (Werribee Campus Victoria, Australia: Victoria University).

Google Scholar

De Vrese, M., Schrezenmeir, J. (2008). Probiotics, prebiotics, and synbiotics. Food Biotechnol. 111, 1–66. doi: 10.1007/10_2008_097

CrossRef Full Text | Google Scholar

D’Incà, R., Barollo, M., Scarpa, M., Grillo, A. R., Brun, P., Vettorato, M. G., et al. (2010). Rectal administration of Lactobacillus casei DG modifies flora composition and Toll-Like Receptor expression in colonic mucosa of patients with mild ulcerative colitis. Digestive Dis. Sci. 56, 1178–1187. doi: 10.1007/s10620-010-1384-1

CrossRef Full Text | Google Scholar

Dominguez-Bello, M. G., Costello, E. K., Contreras, M., Magris, M., Hidalgo, G., Fierer, N., et al. (2010). Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. 107, 11971–11975. doi: 10.1073/pnas.1002601107

CrossRef Full Text | Google Scholar

Dos Santos Cruz, B. C., da Silva Duarte, V., Giacomini, A., Corich, V., Oliveira de Paula, S., da Silva, F. L., et al. (2020). Synbiotic VSL3 and Yacon-based product modulate the intestinal microbiota and prevent the development of pre-neoplastic lesions in a colorectal carcinogenesis model. Appl. Microbiol. Biotechnol. 104, 8837–8857. doi: 10.1007/s00253-020-10863-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Eslamparast, T., Zamani, F., Hekmatdoost, A., Sharafkhah, M., Eghtesad, S., Malekzadeh, R., et al. (2014). Effects of synbiotic supplementation on insulin resistance in subjects with the metabolic syndrome: a randomised, double-blind, placebo-controlled pilot study. Br. J. Nutr. 112, 438–445. doi: 10.1017/S0007114514000919

PubMed Abstract | CrossRef Full Text | Google Scholar

Fan, H., Du, J., Liu, X., Zheng, W. W., Zhuang, Z. H., Wang, C. D., et al. (2019). Effects of pentasa-combined probiotics on the microflora structure and prognosis of patients with inflammatory bowel disease. Turkish J. Gastroenterol. 30, 680–685. doi: 10.5152/tjg.2019.18426

CrossRef Full Text | Google Scholar

Fathizadeh, H., Pakdel, F., Saffari, M., Esmaeili, D., Heravi, M. M., Dao, S., et al. (2022). Bacteriocins: recent advances in its application as an antimicrobial alternative. Curr. Pharm. Biotechnol. 23, 1028–1040. doi: 10.2174/1389201022666210907121254

PubMed Abstract | CrossRef Full Text | Google Scholar

Federico, A., Tuccillo, C., Grossi, E., Abbiati, R., Garbagna, N., Romano, M., et al. (2009). The effect of a new symbiotic formulation on plasma levels and peripheral blood mononuclear cell expression of some pro-inflammatory cytokines in patients with ulcerative colitis: a pilot study. Eur. Rev. Med. Pharmacol. Sci. 13, 285–293. doi: 10.1200/JCO.2008.21.3991

PubMed Abstract | CrossRef Full Text | Google Scholar

Fei, Y., Zhang, S., Han, S., Qiu, B., Lu, Y., Huang, W., et al. (2022). The role of dihydroresveratrol in enhancing the synergistic effect of Ligilactobacillus salivarius Li01 and resveratrol in ameliorating colitis in mice. Res. (Washington D.C.) 2022, 9863845. doi: 10.34133/2022/9863845

CrossRef Full Text | Google Scholar

Fernandez-Banares, F. (1999). Randomized clinical trial of Plantago ovata seeds (dietary fiber) as compared with mesalamine in maintaining remission in ulcerative colitis. Am. J. Gastroenterol. 94, 427–433. doi: 10.1016/s0002-9270(98)00753-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Fujimori, S., Gudis, K., Mitsui, K., Seo, T., Yonezawa, M., Tanaka, S., et al. (2009). A randomized controlled trial on the efficacy of synbiotic versus probiotic or prebiotic treatment to improve the quality of life in patients with ulcerative colitis. Nutr. (Burbank Los Angeles County Calif.) 25, 520–525. doi: 10.1016/j.nut.2008.11.017

CrossRef Full Text | Google Scholar

Fuller, R. (1989). Probiotics in man and animals. J. Appl. Bacteriol. 66, 365–378.

PubMed Abstract | Google Scholar

Furrie, E., Macfarlane, S., Kennedy, A., Cummings, J. H., Walsh, S. V., O'neil, D. A., et al. (2005). Synbiotic therapy (Bifidobacterium Longum/synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: A randomized controlled pilot trial. Gut 54, 242–249. doi: 10.1136/gut.2004.044834

PubMed Abstract | CrossRef Full Text | Google Scholar

Fuss, I. J., Strober, W. (2008). The role of IL-13 and NK T cells in experimental and human ulcerative colitis. Mucosal Immunol. 1, S31–S33. doi: 10.1038/mi.2008.40

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibson, G. R., Beatty, E. R., Wang, X., Cummings, J. H. (1995). Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin. Gastroenterology 108, 975–982. doi: 10.1016/0016-5085(95)90192-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibson, G. R., Hutkins, R., Sanders, M. E., Prescott, S. L., Reimer, R. A., Salminen, S. J., et al. (2017). Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 14, 491–502. doi: 10.1038/nrgastro.2017.75

PubMed Abstract | CrossRef Full Text | Google Scholar

Gibson, G. R., Roberfroid, M. B. (1995). Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 125, 1401–1412. doi: 10.1093/jn/125.6.1401

PubMed Abstract | CrossRef Full Text | Google Scholar

Gill, H. (1998). Stimulation of the immune system by lactic cultures. Int. Dairy J. 8, 535–544. doi: 10.1016/s0958-6946(98)00074-0

CrossRef Full Text | Google Scholar

Gilliland, S. E., Speck, M. L. (1977). Deconjugation of bile acids by intestinal lactobacilli. Appl. Environ. Microbiol. 33, 15–18. doi: 10.1128/aem.33.1.15-18.1977

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldin, B. R., Gualtieri, L. J., Moore, R. P. (1996). The effect of Lactobacillus GG on the initiation and promotion of DMH induced intestinal tumors in the rat. Nutr. Cancer 25, 197–204. doi: 10.1080/01635589609514442

PubMed Abstract | CrossRef Full Text | Google Scholar

Guarino, M., Altomare, A., Emerenziani, S., Di Rosa, C., Ribolsi, M., Balestrieri, P., et al. (2020). Mechanisms of action of prebiotics and their effects on gastro-intestinal disorders in adults. Nutrients 12, 1037. doi: 10.3390/nu12041037

PubMed Abstract | CrossRef Full Text | Google Scholar

Gullon, P., Moura, P., Esteves, M. P., Girio, F. M., Dominguez, H., Parajo, J. C. (2008). Assessment on the fermentability of xylooligosaccharides from rice husks by probiotic bacteria. J. Agric. Food Chem. 56, 7482–7487. doi: 10.1021/jf800715b

PubMed Abstract | CrossRef Full Text | Google Scholar

Guslandi, M., Giollo, P., Testoni, P. A. (2003). A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur. J. Gastroenterol. Hepatol. 15, 697–698. doi: 10.1097/00042737-200306000-00017

PubMed Abstract | CrossRef Full Text | Google Scholar

Hanai, H., Kanauchi, O., Mitsuyama, K., Andoh, A., Takeuchi, K., Takayuki, I., et al. (2004). Germinated barley foodstuff prolongs remission in patients with ulcerative colitis. Int. J. Mol. Med. 3, 643–647. doi: 10.3892/ijmm.13.5.643

CrossRef Full Text | Google Scholar

Hart, A. L., Al-Hassi, H. O., Rigby, R. J., Bell, S. J., Emmanuel, A. V., Knight, S. C., et al. (2005). Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 129, 50–65. doi: 10.1053/j.gastro.2005.05.013

PubMed Abstract | CrossRef Full Text | Google Scholar

He, Q., He, L., Zhang, F., Jian, Z., Sun, J., Chen, J., et al. (2020). Stachyose modulates gut microbiota and alleviates dextran sulfate sodium-induced acute colitis in mice. Saudi J. Gastroenterol. 26, 153. doi: 10.4103/sjg.sjg_580_19

PubMed Abstract | CrossRef Full Text | Google Scholar

Heller, F., Florian, P., Bojarski, C., Richter, J., Christ, M., Hillenbrand, B., et al. (2005). Interleukin-13 the key effector Th2 cytokine in ulcerative colitis that affects epithelial tight junctions, apoptosis, and cell restitution. Gastroenterology 129, 550–564. doi: 10.1016/j.gastro.2005.05.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Heller, F., Fromm, A., Gitter, A. H., Mankertz, J., Schulzke, J. D. (2008). Epithelial apoptosis is a prominent feature of the epithelial barrier disturbance in intestinal inflammation: effect of pro-inflammatory interleukin-13 on epithelial cell function. Mucosal Immunol. 1, s58–s61. doi: 10.1038/mi.2008.46

PubMed Abstract | CrossRef Full Text | Google Scholar

Henker, J., Laass, M., Baretton, G., Fischer, R., Aust, D. (2008). Fallstricke bei der Zöliakiediagnostik. Z. Für Gastroenterologie 46, 675–680. doi: 10.1055/s-2008-1027409

CrossRef Full Text | Google Scholar

Higuchi, L. M., Khalili, H., Chan, A. T., Richter, J. M., Bousvaros, A., Fuchs, C. S. (2012). A prospective study of cigarette smoking and the risk of inflammatory bowel disease in women. Am. J. Gastroenterol. 107, 1399–1406. doi: 10.1038/ajg.2012.196

PubMed Abstract | CrossRef Full Text | Google Scholar

Hill, C., Guarner, F., Reid, G., Gibson, G., Merenstein, D., Pot, B., et al. (2014). The international scientific association for probiotics and prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11, 506–514. doi: 10.1038/nrgastro.2014.66

PubMed Abstract | CrossRef Full Text | Google Scholar

Hilton, E., Kolakowski, P., Singer, C., Smith, M. (1997). Efficacy of Lactobacillus GG as a diarrheal preventive in travelers. J. Travel Med. 4, 41–43. doi: 10.1111/j.1708-8305.1997.tb00772.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, K. A., Gubatan, J. (2023). Gut microbiome–based therapeutics in inflammatory bowel disease. Clin. Trans. Discovery 3, 1-9. doi: 10.1002/ctd2.182

CrossRef Full Text | Google Scholar

Huang, M., Chen, Z., Lang, C., Chen, J., Yang, B., Xue, L., et al. (2018). Efficacy of mesalazine in combination with bifid triple viable capsules on ulcerative colitis and the resultant effect on the inflammatory factors. Pakistan J. Pharm. Sci. 31, 2891–2895.

Google Scholar

Huda-Faujan, N., Abdulamir, A., Fatimah, A., Anas, O. M., Shuhaimi, M., Yazid, A., et al. (2010). The impact of the level of the intestinal short chain fatty acids in inflammatory bowel disease patients versus healthy subjects. Open Biochem. J. 4, 53–58. doi: 10.2174/1874091x01004010053

PubMed Abstract | CrossRef Full Text | Google Scholar

Hughes, D. B., Hoover, D. G. (1995). Viability and enzymatic activity of bifidobacteria in milk. J. Dairy Sci. 78, 268–276. doi: 10.3168/jds.s0022-0302(95)76634-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Im, J. P., Ye, B. D., Kim, Y. S., Kim, J. S. (2018). Changing treatment paradigms for the management of inflammatory bowel disease. Korean J. Internal Med. 33, 28–35. doi: 10.3904/kjim.2017.400

CrossRef Full Text | Google Scholar

Ipar, N., Aydogdu, S. D., Yildirim, G. K., Inal, M., Gies, I., Vandenplas, Y., et al. (2015). Effects of synbiotic on anthropometry, lipid profile and oxidative stress in obese children. Beneficial Microbes 6, 775–781. doi: 10.3920/bm2015.0011

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishikawa, H., Akedo, I., Umesaki, Y., Tanaka, R., Imaoka, A., Otani, T. (2003). Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis. J. Am. Coll. Nutr. 22, 56–63. doi: 10.1080/07315724.2003.10719276

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishikawa, H., Matsumoto, S., Ohashi, Y., Imaoka, A., Setoyama, H., Umesaki, Y., et al. (2011). Beneficial effects of probiotic Bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: A randomized controlled study. Digestion 84, 128–133. doi: 10.1159/000322977

PubMed Abstract | CrossRef Full Text | Google Scholar

Ivanovska, T. P., Mladenovska, K., Zhivikj, Z., Pavlova, M. J., Gjurovski, I., Ristoski, T., et al. (2017). Synbiotic loaded chitosan-Ca-alginate microparticles reduces inflammation in the TNBS model of rat colitis. Int. J. Pharmaceutics 527, 126–134. doi: 10.1016/j.ijpharm.2017.05.049

CrossRef Full Text | Google Scholar

Jakubczyk, D., Leszczynska, K., Gorska, S. (2020). The effectiveness of probiotics in the treatment of inflammatory bowel disease (IBD)—A critical review. Nutrients 12, 1973. doi: 10.3390/nu12071973

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, W., Ai, H., Huang, Q., Li, C., He, X., Jin, Z., et al. (2023). Preclinical evidence of probiotics in ulcerative colitis: a systematic review and network meta-analysis. Front. Pharmacol. 14. doi: 10.3389/fphar.2023.1187911

CrossRef Full Text | Google Scholar

Judge, T. A., Lichtenstein, G. R. (2001). Refractory inflammatory bowel disease. Curr. Treat Options Gastroenterol. 4, 267–273. doi: 10.1007/s11938-001-0039-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaila, M., Isolauri, E., Soppi, E., Virtanen, E., Laine, S., Arvilommi, H. (1992). Enhancement of the circulating antibody secreting cell response in human diarrhea by a human Lactobacillus strain. Pediatr. Res. 32, 141–144. doi: 10.1203/00006450-199208000-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

Kamarli, A. H., Akal, Y. E., Akin, M. (2019). Effects of synbiotic therapy in mild-to-moderately active ulcerative colitis: A randomized placebo-controlled study. Turkish J. Gastroenterol. 30, 313–320. doi: 10.5152/tjg.2019.18356

CrossRef Full Text | Google Scholar

Kanwal, S., Joseph, T. P., Aliya, S., Song, S., Saleem, M. Z., Nisar, M. A., et al. (2020). Attenuation of DSS induced colitis by Dictyophora indusiata polysaccharide (DIP) via modulation of gut microbiota and inflammatory related signaling pathways. J. Funct. Foods 64, 103641. doi: 10.1016/j.jff.2019.103641

CrossRef Full Text | Google Scholar

Kato, K., Mizuno, S., Umesaki, Y., Ishii, Y., Sugitani, M., Imaoka, A., et al. (2004). Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis. Alimentary Pharmacol. Ther. 20, 1133–1141. doi: 10.1111/j.1365-2036.2004.02268.x

CrossRef Full Text | Google Scholar

K-da, S., Peerakietkhajorn, S., Siringoringo, B., Muangnil, P., Wichienchot, S., Khuituan, P. (2020). Oligosaccharides from Gracilaria fisheri ameliorate gastrointestinal dysmotility and gut dysbiosis in colitis mice. J. Funct. Foods 71, 104021. doi: 10.1016/j.jff.2020.104021

CrossRef Full Text | Google Scholar

Kerner, C., Lewis, J. D., Osterman, M. T. (2014). “Safety considerations in the medical therapy of ulcerative colitis,” in Medical Therapy of Ulcerative Colitis. Ed. Lichtenstein, G. (New York, NY: Springer).

Google Scholar

Khan, K. J., Ullman, T. A., Ford, A. C., Abreu, M. T., Abadir, A., Marshall, J. K., et al. (2011). Antibiotic therapy in inflammatory bowel disease: A systematic review and meta-analysis. Am. J. Gastroenterology. 106, 661–673. doi: 10.1038/ajg.2011.72

CrossRef Full Text | Google Scholar

Khan, I., Wei, J., Li, A., Liu, Z., Yang, P., Jing, Y., et al. (2022). Lactobacillus plantarum strains attenuated DSS-induced colitis in mice by modulating the gut microbiota and immune response. Int. Microbiol. 25, 587–603. doi: 10.1007/s10123-022-00243-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Kilara, A., Shahani, K. (1976). Lactase activity of cultured and acidified dairy products. J. Dairy Sci. 59, 2031–2035. doi: 10.3168/jds.s0022-0302(76)84484-0

CrossRef Full Text | Google Scholar

Kim, D. H., Kim, S., Ahn, J. B., Kim, J. H., Ma, H. W., Seo, D. H., et al. (2020). Lactobacillus plantarum CBT LP3 ameliorates colitis via modulating T cells in mice. Int. J. Med. Microbiol. 310, 151391. doi: 10.1016/j.ijmm.2020.151391

PubMed Abstract | CrossRef Full Text | Google Scholar

Kmiec, Z., Cyman, M., Ślebioda, T. J. (2017). Cells of the innate and adaptive immunity and their interactions in inflammatory bowel disease. Adv. Med. Sci. 62, 1–16. doi: 10.1016/j.advms.2016.09.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Kohmoto, T., Fukui, F., Takaku, H., Machida, Y., Arai, M., Mitsuoka, T. (1988). Effect of isomalto-oligosaccharides on human Fecal flora. bifidobacteria Microflora 7, 61–69. doi: 10.12938/bifidus1982.7.2_61

CrossRef Full Text | Google Scholar

Komaki, S., Haque, A., Miyazaki, H., Matsumoto, T., Nakamura, S. (2020). Unexpected effect of probiotics by Lactococcus lactis subsp. lactis against colitis induced by dextran sulfate sodium in mice. J. Infection Chemotherapy 26, 549–553. doi: 10.1016/j.jiac.2020.01.006

CrossRef Full Text | Google Scholar

Konikoff, M. R., Denson, L. A. (2006). Role of fecal calprotectin as a biomarker of intestinal inflammation in inflammatory bowel disease. Inflammatory Bowel Dis. 12, 524–534. doi: 10.1097/00054725-200606000-00013

CrossRef Full Text | Google Scholar

Kruis, W., Fric, P., Pokrotnieks, J., Lukás, M., Fixa, B., Kascák, M., et al. (2004). Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 53, 1617–1623. doi: 10.1136/gut.2003.037747

PubMed Abstract | CrossRef Full Text | Google Scholar

Kruis, W., Schütz, E., Fric, P., Fixa, B., Judmaier, G., Stolte, M. (1997). Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. Alimentary Pharmacol. Ther. 11, 853–858. doi: 10.1046/j.1365-2036.1997.00225.x

CrossRef Full Text | Google Scholar

Kuhn, F., Klar, E. (2015). Surgical principles in the treatment of ulcerative colitis. Visceral Med. 31, 246–250. doi: 10.1159/000438894

CrossRef Full Text | Google Scholar

Lambert, J., Hull, R. (1996). Upper gastrointestinal tract disease and probiotics. Asia Pacific J. Clin. Nutr. 5, 31–35.

Google Scholar

Lankaputhra, W. E. V., Shah, N. P., Britz, M. L. (1996). Evaluation of media for selective enumeration of Lactobacillus acidophilus and Bifidobacterium spp. Food Aust. 48, 113–118.

Google Scholar

Laroia, S., Martin, J. (1990). bifidobacteria as possible dietary adjuncts in cultured dairy products-a review. Cult Dairy Prod J. 25, 18–22.

Google Scholar

Lavelle, A., Sokol, H. (2020). Gut microbiota-derived metabolites as key actors in inflammatory bowel disease. Nat. Rev. Gastroenterol. Hepatol. 17, 223–237. doi: 10.1038/s41575-019-0258-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, P., Xiao, N., Zeng, L., Xiao, J., Huang, J., Xu, Y., et al. (2020). Structural characteristics of a mannoglucan isolated from Chinese yam and its treatment effects against gut microbiota dysbiosis and DSS-induced colitis in mice. Carbohydr. Polymers 250, 116958. doi: 10.1016/j.carbpol.2020.116958

CrossRef Full Text | Google Scholar

Lidbeck, A., Allinger, U. G., Orrhage, K. M., Ottova, L., Brismar, B., Gustafsson, J., et al. (1991). Impact of Lactobacillus acidophilus supplements on the faecal microflora and soluble faecal bile acids in colon cancer patients. Microbial Ecol. Health Dis. 4, 81–88. doi: 10.3109/08910609109140267

CrossRef Full Text | Google Scholar

Linares, D. M., Gomez, C., Renes, E., Fresno, J. M., Tornadijo, M. E., Ross, R. P., et al. (2017). Lactic acid bacteria and bifidobacteria with potential to design natural biofunctional health-promoting dairy foods. Front. Microbiol. 8. doi: 10.3389/fmicb.2017.00846

CrossRef Full Text | Google Scholar

Liu, Y., Fang, H., Liu, H., Cheng, H., Pan, L., Hu, M., et al. (2021). Goji berry juice fermented by probiotics attenuates dextran sodium sulfate-induced ulcerative colitis in mice. J. Funct. Foods 83, 104491. doi: 10.1016/j.jff.2021.104491

CrossRef Full Text | Google Scholar

Liu, R., Li, Y., Zhang, B. (2016). The effects of konjac oligosaccharide on TNBS-induced colitis in rats. Int. Immunopharmacol. 40, 385–391. doi: 10.1016/j.intimp.2016.08.040

PubMed Abstract | CrossRef Full Text | Google Scholar

Machiels, K., Joossens, M., Sabino, J., De Preter, V., Arijs, I., Eeckhaut, V., et al. (2013). A decrease of the butyrate-producing species Roseburia hominis and Faecali bacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut 63, 1275–1283. doi: 10.1136/gutjnl-2013-304833

PubMed Abstract | CrossRef Full Text | Google Scholar

Mackowiak, P. A. (2013). Recycling Metchnikoff: Probiotics, the intestinal microbiome and the quest for long life. Front. Public Health 1. doi: 10.3389/fpubh.2013.00052

CrossRef Full Text | Google Scholar

Majamaa, H., Isolauri, E. (1997). Probiotics: A novel approach in the management of food allergy. J. Allergy Clin. Immunol. 99, 179–185. doi: 10.1016/s0091-6749(97)70093-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Malathi, K., Nandini, R., Dhanasekar, K. R., Shilpa, B. N. (2019). A randomized open label study to evaluate the efficacy and tolerability of synbiotic in the treatment of ulcerative colitis. J. Vaccine Clin. trials 5, 1–4. doi: 10.35248/2475-3181.5.165

CrossRef Full Text | Google Scholar

Manichanh, C., Borruel, N., Casellas, F., Guarner, F. (2012). The gut microbiota in IBD. Nat. Rev. Gastroenterol. Hepatol. 9, 599–608. doi: 10.1038/nrgastro.2012.152

PubMed Abstract | CrossRef Full Text | Google Scholar

Mariat, D., Firmesse, O., Levenez, F., Guimaraes, V. D., Sokol, H., Dore, J., et al. (2009). The Firmicutes/bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 9, 1–6. doi: 10.1186/1471-2180-9-123

PubMed Abstract | CrossRef Full Text | Google Scholar

Markowiak, P., Śliżewska, K. (2017). Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients 9, 1021. doi: 10.3390/nu9091021

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinez-Villaluenga, C., Gomez, R. (2007). Characterization of bifidobacteria as starters in fermented milk containing raffinose family of oligosaccharides from lupin as prebiotic. Int. Dairy J. 17, 116–122. doi: 10.1016/j.idairyj.2006.02.003

CrossRef Full Text | Google Scholar

Matsuoka, K., Uemura, Y., Kanai, T., Kunisaki, R., Suzuki, Y., Yokoyama, K., et al. (2018). Efficacy of Bifidobacterium breve fermented milk in maintaining remission of ulcerative colitis. Digestive Dis. Sci. 63, 1910–1919. doi: 10.1007/s10620-018-4946-2

CrossRef Full Text | Google Scholar

Matthes, H., Krummenerl, T., Giensch, M., Wolff, C., Schulze, J. (2010). Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complementary Altern. Med. 10, 1–8. doi: 10.1186/1472-6882-10-13

CrossRef Full Text | Google Scholar

McIntosh, G. H. (1996). Probiotics and colon cancer prevention. Asia Pacific J. Clin. Nutr. 5, 48–52.

Google Scholar

Merson, M. H., Morris, G. K., Sack, D. A., Wells, J. G., Feeley, J. C., Sack, R. B., et al. (1976). Travelers’ Diarrhea in Mexico. New Engl. J. Med. 294, 1299–1305. doi: 10.1056/nejm197606102942401

CrossRef Full Text | Google Scholar

Miele, E., Pascarella, F., Giannetti, E., Quaglietta, L., Baldassano, R. N., Staiano, A. (2009). Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am. J. Gastroenterol. 104, 437–443. doi: 10.1038/ajg.2008.118

PubMed Abstract | CrossRef Full Text | Google Scholar

Miele, E., Shamir, R., Aloi, M., Assa, A., Braegger, C., Bronsky, J., et al. (2018). Nutrition in pediatric inflammatory bowel disease: A position paper on behalf of the porto inflammatory bowel disease group of the European Society of Pediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 66, 687–708. doi: 10.1097/mpg.0000000000001896

PubMed Abstract | CrossRef Full Text | Google Scholar

Mijan, M. A., Lim, B. O. (2018). Diets, functional foods, and nutraceuticals as alternative therapies for inflammatory bowel disease: Present status and future trends. World J. Gastroenterol. 24, 2673–2685. doi: 10.3748/wjg.v24.i25.2673

PubMed Abstract | CrossRef Full Text | Google Scholar

Mirmiran, P. (2014). Functional foods-based diet as a novel dietary approach for management of type 2 diabetes and its complications: A review. World J. Diabetes 5, 267–281. doi: 10.4239/wjd.v5.i3.26

PubMed Abstract | CrossRef Full Text | Google Scholar

Mishra, C., Lambert, J. (1996). Production of anti-microbial substances by probiotics. Asia Pacific J. Clin. Nutr. 5, 20–24.

Google Scholar

Mitsuyama, K., Saiki, T., Kanauchi, O., Iwanaga, T., Tomiyasu, N., Nishiyama, T., et al. (1998). Treatment of ulcerative colitis with germinated barley foodstuff feeding: a pilot study. Alimentary Pharmacol. Ther. 12, 1225–1230. doi: 10.1046/j.1365-2036.1998.00432.x

CrossRef Full Text | Google Scholar

Molodecky, N. A., Soon, I. S., Rabi, D. M., Ghali, W. A., Ferris, M., Chernoff, G., et al. (2012). Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 142, 46–54.e42. doi: 10.1053/j.gastro.2011.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Moro, T. M. A., Clerici, T. P. S. (2021). Burdock (Arctium lappa L) roots as a source of inulin-type fructans and other bioactive compounds: Current knowledge and future perspectives for food and non-food applications. Food Res. Int. (Ottawa Ont.) 141, 109889. doi: 10.1016/j.foodres.2020.109889

CrossRef Full Text | Google Scholar

Mowat, C., Cole, A., Windsor, A., Ahmad, T., Arnott, I., Driscoll, R., et al. (2011). Guidelines for the management of inflammatory bowel disease in adults. Gut 60, 571–607. doi: 10.1136/gut.2010.224154

PubMed Abstract | CrossRef Full Text | Google Scholar

Nag, D., Farr, D., Raychaudhuri, S., Withey, J. H. (2022). An adult zebrafish model for adherent-invasive Escherichia coli indicates protection from AIEC infection by probiotic E. coli Nissle. iScience 25, 104572. doi: 10.1016/j.isci.2022.104572

PubMed Abstract | CrossRef Full Text | Google Scholar

Ng, S. C., Plamondon, S., Kamm, M. A., Hart, A. L., Al-Hassi, H. O., Guenther, T., et al. (2010). Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis. Inflammatory Bowel Dis. 16, 1286–1298. doi: 10.1002/ibd.21222

CrossRef Full Text | Google Scholar

Ng, S. C., Shi, H. Y., Hamidi, N., Underwood, F. E., Tang, W., Benchimol, E. I., et al. (2017). Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 390, 2769–2778. doi: 10.1016/s0140-6736(17)32448-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Nwafor, I. C., Shale, K., Achilonu, M. C. (2017). Chemical composition and nutritive benefits of chicory (Cichorium intybus) as an ideal complementary and/or alternative livestock feed supplement. Sci. World J. 2017, 7343928. doi: 10.1155/2017/7343928

CrossRef Full Text | Google Scholar

Okazaki, M., Fujikawa, S., Matsumoto, N. (1990). Effect of xylooligosaccharide on the growth of bifidobacteria. Bifidobacteria Microflora 9, 77–86. doi: 10.12938/bifidus1982.9.2_77

CrossRef Full Text | Google Scholar

Oksanen, P. J., Salminen, S., Saxelin, M., Hamalainen, P., Ihantola-Vormisto, A., Muurasniemi-Isoviita, L., et al. (1990). Prevention of travellers diarrhoea by Lactobacillus GG. Ann. Med. 22, 53–56. doi: 10.3109/07853899009147242

PubMed Abstract | CrossRef Full Text | Google Scholar

Oliva, S., Di Nardo, G., Ferrari, F., Mallardo, S., Rossi, P., Patrizi, G., et al. (2011). Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis. Alimentary Pharmacol. Ther. 35, 327–334. doi: 10.1111/j.1365-2036.2011.04939.x

CrossRef Full Text | Google Scholar

Palumbo, V. D., Romeo, M., Gammazza, A. M., Carini, F., Damiani, P., Damiano, G., et al. (2016). The long-term effects of probiotics in the therapy of ulcerative colitis: A clinical study. Biomed. Papers 160, 372–377. doi: 10.5507/bp.2016.044

CrossRef Full Text | Google Scholar

Pandey, K. R., Naik, S. R., Vakil, B. V. (2015). Probiotics, prebiotics and synbiotics- a review. J. Food Sci. Technol. 52, 7577–7587. doi: 10.1007/s13197-015-1921-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Panigrahi, P., Parida, S., Nanda, N. C., Satpathy, R., Pradhan, L., Chandel, D. S., et al. (2017). A randomized synbiotic trial to prevent sepsis among infants in rural India. Nature 548, 407–412. doi: 10.1038/nature23480

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, K. T., Ehrlich, O. G., Allen, J. I., Meadows, P., Szigethy, E. M., Henrichsen, K., et al. (2020). Corrigendum to the cost of Inflammatory Bowel Disease: An initiative from the Crohn’s & Colitis Foundation. Inflammatory Bowel Dis. 26, 1118–1118. doi: 10.1093/ibd/izaa081

CrossRef Full Text | Google Scholar

Pathmakanthan, S., Walsh, M., Bengmark, S. (2002). Efficacy and tolerability treating acute distal ulcerative colitis with synbiotic enemas: a pilot trial (abstract) (Geneva: United European Gastroenterology Week), 190.

Google Scholar

Perez-Conesa, D., Lopez, G., Abellan, P., Ros, G. (2006). Bioavailability of calcium, magnesium and phosphorus in rats fed probiotic, prebiotic and synbiotic powder follow-up infant formulas and their effect on physiological and nutritional parameters. J. Sci. Food Agric. 86, 2327–2336. doi: 10.1002/jsfa.2618

CrossRef Full Text | Google Scholar

Perrin, S., Warchol, M., Grill, J., Schneider, F. (2001). Fermentations of fructooligosaccharides and their components by Bifidobacterium infantis ATCC 15697 on batch culture in semi-synthetic medium. J. Appl. Microbiol. 90, 859–865. doi: 10.1046/j.1365-2672.2001.01317.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Petersen, A. M., Mirsepasi, H., Halkjær, S. I., Mortensen, E. M., Nordgaard-Lassen, I., Krogfelt, K. A. (2014). Ciprofloxacin and probiotic Escherichia coli Nissle add-on treatment in active ulcerative colitis: A double-blind randomized placebo-controlled clinical trial. J. Crohn’s Colitis 8, 1498–1505. doi: 10.1016/j.crohns.2014.06.001

CrossRef Full Text | Google Scholar

Poggi, A., Benelli, R., Vene, R., Costa, D., Ferrari, N., Tosetti, F., et al. (2019). Human gut-associated natural killer cells in health and disease. Front. Immunol. 10. doi: 10.3389/fimmu.2019.00961

PubMed Abstract | CrossRef Full Text | Google Scholar

Qian, X., Jiang, H., Wu, Y., Shao, M. H., He, J. W., Bao, X., et al. (2022). Fecal microbiota transplantation combined with prebiotics ameliorates ulcerative colitis in mice. Res. Square 2022, 1–20. doi: 10.21203/rs.3.rs-1742266/v1

CrossRef Full Text | Google Scholar

Rasmussen, H. E., Hamaker, B. R. (2017). Prebiotics and inflammatory bowel disease. Gastroenterol. Clinics North America 46, 783–795. doi: 10.1016/j.gtc.2017.08.004

CrossRef Full Text | Google Scholar

Reddy, B. S., Rivenson, A. (1993). Inhibitory effect of Bifidobacterium longum on colon, mammary, and liver carcinogenesis induced by 2-amino-3-methylimidazo [4,5-f] quinoline, a food mutagen. Cancer Res. 53, 3914–3918.

PubMed Abstract | Google Scholar

Rembacken, B., Snelling, A., Hawkey, P., Chalmers, D., Axon, A. (1999). Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet 354, 635–639. doi: 10.1016/s0140-6736(98)06343-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Roberfroid, M. B. (1993). Dietary fiber, inulin, and oligofructose: a review comparing their physiological effects. Crit. Rev. Food Sci. Nutr. 33, 103–104. doi: 10.1080/10408399309527616

PubMed Abstract | CrossRef Full Text | Google Scholar

Roy, S., Dhaneshwar, S. (2023). Role of prebiotics, probiotics, and synbiotics in management of inflammatory bowel disease: Current perspectives. World J. Gastroenterol. 29, 2078–2100. doi: 10.3748/wjg.v29.i14.2078

PubMed Abstract | CrossRef Full Text | Google Scholar

Sabater-Molina, M., Larqué, E., Torrella, F., Zamora, S. (2009). Dietary fructooligosaccharides and potential benefits on health. J. Physiol. Biochem. 65, 315–328. doi: 10.1007/BF03180584

PubMed Abstract | CrossRef Full Text | Google Scholar

Safavi, M., Farajian, S., Kelishadi, R., Mirlohi, M., Hashemipour, M. (2013). The effects of synbiotic supplementation on some cardio-metabolic risk factors in overweight and obese children: a randomized triple-masked controlled trial. Int. J. Food Sci. Nutr. 64, 687–693. doi: 10.3109/09637486.2013.775224

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez, M., Darimont, C., Drapeau, V., Emady-Azar, S., Lepage, M., Rezzonico, E., et al. (2014). Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women. Br. J. Nutr. 111, 1507–1519. doi: 10.1017/s0007114513003875

PubMed Abstract | CrossRef Full Text | Google Scholar

Schiffrin, E., Rochat, F., Link-Amster, H., Aeschlimann, J., Donnet-Hughes, A. (1995). Immunomodulation of human blood cells following the ingestion of Lactic acid bacteria. J. Dairy Sci. 78, 491–497. doi: 10.3168/jds.s0022-0302(95)76659-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Seo, G. S., Chae, S. C. (2014). Biological therapy for ulcerative colitis: an update. World J. Gastroenterol. 20, 13234–13238. doi: 10.3748/wjg.v20.i37.13234

PubMed Abstract | CrossRef Full Text | Google Scholar

Seong, G., Lee, S., Min, Y. W., Jang, Y. S., Park, S. Y., Kim, C. H., et al. (2020). Effect of a synbiotic containing Lactobacillus paracasei and Opuntia humifusa on a murine model of irritable bowel syndrome. Nutrients 12, 3205. doi: 10.3390/nu12103205

PubMed Abstract | CrossRef Full Text | Google Scholar

Shahani, K. M., Chandan, R. C. (1979). Nutritional and healthful aspects of cultured and culture-containing dairy foods. J. Dairy Sci. 62, 1685–1694. doi: 10.3168/jds.s0022-0302(79)83481-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, P., Kaur, S., Chadha, B. S., Kaur, R., Kaur, M., Kaur, S. (2021). Anticancer and antimicrobial potential of enterocin 12a from Enterococcus faecium. BMC Microbiol. 21, 39. doi: 10.1186/s12866-021-02086-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, M., Shukla, G. (2016). Metabiotics: One Step ahead of probiotics; an insight into mechanisms involved in anticancerous effect in colorectal cancer. Front. Microbiol. 7. doi: 10.3389/fmicb.2016.01940

CrossRef Full Text | Google Scholar

Sheoran, P., Tiwari, S. K. (2019). Enterocin LD3 from Enterococcus hirae LD3 causing efflux of intracellular ions and UV absorbing materials in gram negative bacteria. J. Appl. Microbiol. 126, 1059–1069. doi: 10.1111/jam.14203

PubMed Abstract | CrossRef Full Text | Google Scholar

Sheoran, P., Tiwari, S. K. (2020). Synergistically-acting Enterocin LD3 and Plantaricin LD4 against gram-positive and gram-negative pathogenic bacteria. Probiotics Antimicrobial Proteins 13, 542–554. doi: 10.1007/s12602-020-09708-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Shin, H. S., Lee, J. H., Pestka, J., Ustunol, Z. (2000). Growth and viability of commercial Bifidobacterium spp in skim milk containing oligosaccharides and inulin. J. Food Sci. 65, 884–887. doi: 10.1111/j.1365-2621.2000.tb13605.x

CrossRef Full Text | Google Scholar

Siitonen, S., Vapaatalo, H., Salminen, S., Gordin, A., Saxelin, M., Wikberg, R., et al. (1990). Effect of Lactobacillus GG yoghurt in prevention of antibiotic associated diarrhoea. Ann. Med. 22, 57–59. doi: 10.3109/07853899009147243

PubMed Abstract | CrossRef Full Text | Google Scholar

Silva, N. O. E., Brito, B. B. D., Silva, F. A. F. D., Santos, M. L. C., Melo, F. F. D. (2020). Probiotics in inflammatory bowel disease: Does it work? World J. Meta-Analysis 8, 54–66. doi: 10.13105/wjma.v8.i2.54

CrossRef Full Text | Google Scholar

Silveira, D. S. C., Veronez, L. C., Lopes-Junior, L. C., Anatriello, E., Brunaldi, M. O., Pereira-da-Silva, G. (2020). Lactobacillus bulgaricus inhibits colitis-associated cancer via a negative regulation of intestinal inflammation in azoxymethane/dextran sodium sulfate model. World J. Gastroenterol. 26, 6782–6794. doi: 10.3748/wjg.v26.i43.6782

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, A., Mahajan, R., Kedia, S., Dutta, A. K., Anand, A., Bernstein, C. N., et al. (2022). Use of thiopurines in inflammatory bowel disease: an update. Intestinal Res. 20, 11–30. doi: 10.5217/ir.2020.00155

CrossRef Full Text | Google Scholar

Smid, A., Strnisa, L., Bajc, K., Vujic-Podlipec, D., Bogovic Matijasic, B., Rogelj, I. (2016). Randomized clinical trial: The effect of fermented milk with the probiotic cultures Lactobacillus acidophilus La-5® and Bifidobacterium BB-12® and Beneo dietary fibres on health-related quality of life and the symptoms of irritable bowel syndrome in adults. J. Funct. Foods 24, 549–557. doi: 10.1016/j.jff.2016.04.031

CrossRef Full Text | Google Scholar

Somashekaraiah, R., Mottawea, W., Gunduraj, A., Joshi, U., Hammami, R., Sreenivasa, M. Y. (2021). Probiotic and antifungal attributes of Levilactobacillus brevis MYSN105, isolated from an Indian traditional fermented food pozha. Front. Microbiol. 12. doi: 10.3389/fmicb.2021.696267

PubMed Abstract | CrossRef Full Text | Google Scholar

Son, S., Koh, J., Park, M., Ryu, S., Lee, W., Yun, B., et al. (2019). Effect of the Lactobacillus rhamnosus strain GG and tagatose as a synbiotic combination in a dextran sulfate sodium-induced colitis murine model. J. Dairy Sci. 102, 2844–2853. doi: 10.3168/jds.2018-15013

PubMed Abstract | CrossRef Full Text | Google Scholar

Sood, A., Mahajan, R., Singh, A., Midha, V., Mehta, V., Narang, V., et al. (2019). Role of faecal microbiota transplantation for maintenance of remission in patients with ulcerative colitis: A pilot study. J. Crohn’s Colitis 13, 1311–1317. doi: 10.1093/ecco-jcc/jjz060

CrossRef Full Text | Google Scholar

Sood, A., Midha, V., Makharia, G. K., Ahuja, V., Singal, D., Goswami, P., et al. (2009). The probiotic preparation, VSL3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin. Gastroenterol. Hepatol. 7, 1202–1209.e1. doi: 10.1016/j.cgh.2009.07.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Steinhart, A., Feagan, B. G., Wong, C. J., Vandervoort, M., Mikolainis, S., Croitoru, K., et al. (2002). Combined budesonide and antibiotic therapy for active Crohn’s disease: A randomized controlled trial. Gastroenterology 123, 33–40. doi: 10.1053/gast.2002.34225

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, H., Kang, Q., Wang, H., Yin, H., Duan, L., Liu, Y., et al. (2018). Effects of glucocorticoids combined with probiotics in treating crohn’s disease on inflammatory factors and intestinal microflora. Exp. Ther. Med. 16, 2999–3003. doi: 10.3892/etm.2018.6559

PubMed Abstract | CrossRef Full Text | Google Scholar

Sutas, Y., Hurme, M., Isolauri, E. (1996). Down regulation of anti-CD3 antibody induced IL-4 production by bovine caseins hydrolysed with Lactobacillus GG derived enzymes. Scandinavian J. Immunol. 43, 687–689. doi: 10.1046/j.1365-3083.1996.d01-258.x

CrossRef Full Text | Google Scholar

Swanson, K. S., Gibson, G. R., Hutkins, R., Reimer, R. A., Reid, G., Verbeke, K., et al. (2020). The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of synbiotics. Nat. Rev. Gastroenterol. Hepatol. 17, 687–701. doi: 10.1038/s41575-020-0344-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Tajadadi-Ebrahimi, M., Bahmani, F., Shakeri, H., Hadaegh, H., Hijijafari, M., Abedi, F., et al. (2014). Effects of daily consumption of synbiotic bread on insulin metabolism and serum high-sensitivity C-reactive protein among diabetic patients: A double-blind, randomized, controlled clinical trial. Ann. Nutr. Metab. 65, 34–41. doi: 10.1159/000365153

PubMed Abstract | CrossRef Full Text | Google Scholar

Tamaki, H., Nakase, H., Inoue, S., Kawanami, C., Itani, T., Ohana, M., et al. (2015). Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: A randomized, double blinded, placebo controlled multicenter trial. Digestive Endoscopy 28, 67–74. doi: 10.1111/den.12553

PubMed Abstract | CrossRef Full Text | Google Scholar

Tharmaraj, N., Shah, N. P. (2009). Antimicrobial effects of probiotic bacteria against selected species of yeasts and moulds in cheese-based dips. Int. J. Food Sci. Technol. 44, 1916–1926. doi: 10.1111/j.1365-2621.2009.01986.x

CrossRef Full Text | Google Scholar

Tojo, M., Oikawa, T., Morikawa, Y., Yamashita, N., Iwata, S., Satoh, Y., et al. (1987). The effects of Bifidobacterium breve administration on Campylobacter enteritis. Pediatr. Int. 29, 160–167. doi: 10.1111/j.1442-200x.1987.tb00024.x

CrossRef Full Text | Google Scholar

Tolonen, A. C., Beauchemin, N., Bayne, C., Li, L., Tan, J., Lee, J., et al. (2022). Synthetic glycans control gut microbiome structure and mitigate colitis in mice. Nat. Commun. 13, 1244. doi: 10.1038/s41467-022-28856-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Turner, D., Ricciuto, A., Lewis, A., D’Amico, F., Dhaliwal, J., Griffiths, A. M., et al. (2021). STRIDE-II: An update on the selecting therapeutic targets in inflammatory bowel disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): Determining therapeutic goals for treat-to-target strategies in IBD. Gastroenterology 160, 1570–1583. doi: 10.1053/j.gastro.2020.12.031

PubMed Abstract | CrossRef Full Text | Google Scholar

Tursi, A., Brandimarte, G., Papa, A., Giglio, A., Elisei, W., Giorgetti, G. M., et al. (2010). Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL3 as adjunctive to a standard pharmaceutical treatment: A double-blind, randomized, placebo-controlled Study. Am. J. Gastroenterol. 105, 2218–2227. doi: 10.1038/ajg.2010.218

PubMed Abstract | CrossRef Full Text | Google Scholar

Tzounis, X., Rodriguez-Mateos, A., Vulevic, J., Gibson, G. R., Kwik-Uribe, C., Spencer, J. P. (2011). Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 93, 62–72. doi: 10.3945/ajcn.110.000075

PubMed Abstract | CrossRef Full Text | Google Scholar

Ukwah, B. N., Ezeonu, I. M., Uguru, J. (2014). Assessment of prebiotic roles of Musa paradisiaca (Plantain) peels for controlling spread of Salmonella and other infectious disease agents by broilers. J. Biol. Chem. Res. 31, 901–911.

Google Scholar

Ungaro, R., Mehandru, S., Allen, P. B., Peyrin-Biroulet, L., Colombel, J. F. (2017). Ulcerative colitis. Lancet 389, 1756–1770. doi: 10.1016/s0140-6736(16)32126-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Valcheva, R., Koleva, P., Martinez, I., Walter, J., Ganzle, M. G., Dieleman, L. A. (2018). Inulin-type fructans improve active ulcerative colitis associated with microbiota changes and increased short-chain fatty acids levels. Gut Microbes 10, 334–357. doi: 10.1080/19490976.2018.1526583

PubMed Abstract | CrossRef Full Text | Google Scholar

Valcheva, R., Armstrong, H., Kovic, O., Bording-Jorgensen, M., Veniamin, S., Pérez-Muñoz, M. E., et al (2022). Double blind placebo-controlled trial for the prevention of ulcerative colitis relapses by β-fructan prebiotics: efficacy and metabolomic analysis. Preprint. Available from: medRxiv . doi: 10.1101/2022.01.16.22269376

CrossRef Full Text | Google Scholar

Van der Beek, C. M., Dejong, C. H. C., Troost, F. J., Masclee, A. A. M., Lenaerts, K. (2017). Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr. Rev. 75, 286–305. doi: 10.1093/nutrit/nuw067

PubMed Abstract | CrossRef Full Text | Google Scholar

Van der Meer, R., Bovee-Oudenhoven, I. (1998). Dietary modulation of intestinal bacterial infections. Int. Dairy J. 8, 481–486. doi: 10.1016/s0958-6946(98)00072-7

CrossRef Full Text | Google Scholar

Van Laere, K. M. J., Hartemink, R., Bosveld, M., Schols, H. A., Voragen, A. G. J. (2000). Fermentation of plant cell wall derived polysaccharides and their corresponding oligosaccharides by intestinal bacteria. J. Agric. Food Chem. 48, 1644–1652. doi: 10.1021/jf990519i

PubMed Abstract | CrossRef Full Text | Google Scholar

Venturi, A., Gionchetti, P., Rizzello, F., Johansson, R., Zucconi, E., Brigidi, P., et al. (1999). Impact on the composition of the faecal flora by a new probiotic preparation: preliminary data on maintenance treatment of patients with ulcerative colitis. Alimentary Pharmacol. Ther. 13, 1103–1108. doi: 10.1046/j.1365-2036.1999.00560.x

CrossRef Full Text | Google Scholar

Vernero, M., De Blasio, F., Ribaldone, D. G., Bugianesi, E., Pellicano, R., Saracco, G. M., et al. (2020). The usefulness of microencapsulated sodium butyrate add-on therapy in maintaining remission in patients with ulcerative colitis: A prospective observational study. J. Clin. Med. 9, 3941. doi: 10.3390/jcm9123941

PubMed Abstract | CrossRef Full Text | Google Scholar

Vyas, U., Ranganathan, N. (2012). Probiotics, prebiotics, and synbiotics: Gut and beyond. Gastroenterol. Res. Pract. 2012, 1–16. doi: 10.1155/2012/872716

CrossRef Full Text | Google Scholar

Waljee, A. K., Wiitala, W. L., Govani, S., Stidham, R., Saini, S., Hou, J., et al. (2016). Corticosteroid use and complications in a US inflammatory bowel disease cohort. PloS One 11, e0158017. doi: 10.1371/journal.pone.0158017

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, W., Chen, L., Zhou, R., Wang, X., Song, L., Huang, S. (2014). Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease. J. Clin. Microbiol. 52, 398–406. doi: 10.1128/jcm.01500-13

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, Y. N., Meng, X. C., Dong, Y. F., Zhao, X. H., Qian, J. M., Wang, H. Y., et al. (2019). Effects of probiotics and prebiotics on intestinal microbiota in mice with acute colitis based on 16S rRNA gene sequencing. Chin. Med. J. 132, 1833–1842. doi: 10.1097/cm9.0000000000000308

CrossRef Full Text | Google Scholar

Wild, G. E., Waschke, K. A., Bitton, A., Thomson, A. B. (2003). The mechanisms of prednisone inhibition of inflammation in crohn’s disease involve changes in intestinal permeability, mucosal TNFalpha production and nuclear factor kappa B expression. Aliment Pharmacol. Ther. 18, 309–317. doi: 10.1046/j.1365-2036.2003.01611.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wildt, S., Nordgaard, I., Hansen, U., Brockmann, E., Rumessen, J. J. (2011). A randomized 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. Crohn’s Colitis 5, 115–121. doi: 10.1016/j.crohns.2010.11.004

CrossRef Full Text | Google Scholar

Wirngo, F. E., Lambert, M. N., Jeppesen, P. B. (2016). The physiological effects of dandelion (Taraxacum officinale) in type 2 diabetes. Rev. Diabetic studies: RDS. 13, 113–131. doi: 10.1900/RDS.2016.13.113

CrossRef Full Text | Google Scholar

Wong, W.-Y., Chan, B. D., Leung, T.-W., Chen, M., Tai, W. C.-S. (2022). Beneficial and anti-inflammatory effects of formulated prebiotics, probiotics, and synbiotics in normal and acute colitis mice. J. Funct. Foods 88, 104871. doi: 10.1016/j.jff.2021.104871

CrossRef Full Text | Google Scholar

Wong, V. W. S., Wong, G. L. H., Chim, A. M. L., Chu, W. C. W., Yeung, D. K. W., Li, K. C. T., et al. (2013). Treatment of nonalcoholic steatohepatitis with probiotics. A proof-of-concept study. Ann. Hepatol. 12, 256–262. doi: 10.1016/s1665-2681(19)31364-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Xue, Z., Li, R., Liu, J., Zhou, J., Zhang, X., Zhang, T., et al. (2023). Preventive and synbiotic effects of the soluble dietary fiber obtained from Lentinula edodes byproducts and Lactobacillus plantarum LP90 against dextran sulfate sodium induced colitis in mice. J. Sci. Food Agric. 103, 616–626. doi: 10.1002/jsfa.12173

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, S. C., Chen, J. Y., Shang, H. F., Cheng, T. Y., Tsou, S. C., Chen, J. R. (2005). Effect of synbiotics on intestinal microflora and digestive enzyme activities in rats. World J. Gastroenterol. 11, 7413. doi: 10.3748/wjg.v11.i47.7413

PubMed Abstract | CrossRef Full Text | Google Scholar

Yilmaz, I., Dolar, M. E., Ozpinar, H. (2019). Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turkish J. Gastroenterol. 30, 242–253. doi: 10.5152/tjg.2018.18227

CrossRef Full Text | Google Scholar

Yoo, J. W., Shin, Y. J., Ma, X., Son, Y. H., Jang, H. M., Lee, C. K., et al. (2022). The alleviation of gut microbiota-induced depression and colitis in mice by anti-inflammatory probiotics NK151, NK173, and NK175. Nutrients 14, 2080. doi: 10.3390/nu14102080

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshimatsu, Y., Yamada, A., Furukawa, R., Sono, K., Osamura, A., Nakamura, K., et al. (2015). Effectiveness of probiotic therapy for the prevention of relapse in patients with inactive ulcerative colitis. World J. Gastroenterol. 21, 5985–5994. doi: 10.3748/wjg.v21.i19.5985

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, H., Xue, Y., Peng, T., Shao, W. (2007). Properties of xylanolytic enzyme system in bifidobacteria and their effects on the utilization of xylooligosaccharides. Food Chem. 101, 1172–1177. doi: 10.1016/j.foodchem.2006.03.019

CrossRef Full Text | Google Scholar

Zocco, M. A., Dal Verme, L. Z., Cremonini, F., Piscaglia, A. C., Nista, E. C., Candelli, M., et al. (2006). Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Alimentary Pharmacol. Ther. 23, 1567–1574. doi: 10.1111/j.1365-2036.2006.02927.x

CrossRef Full Text | Google Scholar

Keywords: inflammatory bowel diseases, ulcerative colitis, probiotics, prebiotics, synbiotics, therapeutic uses

Citation: Jadhav A, Jagtap S, Vyavahare S, Sharbidre A and Kunchiraman B (2023) Reviewing the potential of probiotics, prebiotics and synbiotics: advancements in treatment of ulcerative colitis. Front. Cell. Infect. Microbiol. 13:1268041. doi: 10.3389/fcimb.2023.1268041

Received: 04 August 2023; Accepted: 22 November 2023;
Published: 08 December 2023.

Edited by:

Venkatakrishna Rao Jala, University of Louisville, United States

Reviewed by:

Robert Fultz, Brightseed, United States
Alberto Finamore, Council for Agricultural and Economics Research (CREA), Italy
Dhrubajyoti Nag, City College of New York (CUNY), United States

Copyright © 2023 Jadhav, Jagtap, Vyavahare, Sharbidre and Kunchiraman. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Suresh Jagtap, chiritatml@rediffmail.com

Download