Next Article in Journal
Novel Glutamate–Putrescine Ligase Activity in Haloferax mediterranei: A New Function for glnA-2 Gene
Next Article in Special Issue
Probiotics and Trained Immunity
Previous Article in Journal
Identification of Biomolecules Involved in the Adaptation to the Environment of Cold-Loving Microorganisms and Metabolic Pathways for Their Production
Previous Article in Special Issue
Probiotics, Prebiotics and Postbiotics on Mitigation of Depression Symptoms: Modulation of the Brain–Gut–Microbiome Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotics, Prebiotics, and Synbiotics in the Irritable Bowel Syndrome Treatment: A Review

by
Agnieszka Chlebicz-Wójcik
* and
Katarzyna Śliżewska
*
Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Lodz University of Technology, Wólczańska 171/173, 90-530 Łódź, Poland
*
Authors to whom correspondence should be addressed.
Biomolecules 2021, 11(8), 1154; https://doi.org/10.3390/biom11081154
Submission received: 3 July 2021 / Revised: 2 August 2021 / Accepted: 2 August 2021 / Published: 4 August 2021
(This article belongs to the Special Issue Prebiotics and Probiotics in Health and Disease)

Abstract

:
Irritable bowel syndrome is not a life-threatening disease, yet it significantly affects the quality of life and contributes to economic loss. It is estimated that even up to 45% of the world’s population can suffer from the disease. The first attempts to diagnose irritable bowel syndrome were made at the end of the 19th century; however, establishing appropriate diagnostic criteria and treatment methods is still ongoing. To date, little is known about the etiology of irritable bowel syndrome; however, growing attention is drawn to the intestinal microbiota as a factor in the disease development. For this reason, researchers have conducted many studies on therapies that modulate the microbiota, among which probiotics, prebiotics, and synbiotics are widely studied. To date, most studies have examined probiotics; however, there are also several studies demonstrating the efficacy of prebiotics and synbiotics. The aim of this review was to summarize findings on the usefulness of probiotics, prebiotics, and synbiotics in the treatment of irritable bowel syndrome.

1. Introduction

Irritable bowel syndrome (IBS) is an intestinal functional disorder that is classified as a non-life-threatening disease. It causes a decline in life quality and abates an ability to function in society, as well as attributes to economic losses [1,2]. The cost of IBS is estimated to be up to EUR 8 billion in Europe, nearly USD 2 billion in China, and up to USD 10 billion in the United States of America [1]. It is predicted that up to 10% of the worldwide population suffers from this disease. Some sources estimate an even higher prevalence reaching 45% [1,3]. Women are 1.5–3 times more likely to develop IBS than men, with a two times higher possibility of constipation-associated symptoms, whereas men exhibit a diarrheal form of the condition [4].
Based on the manifestation of the disease, IBS is divided into four subtypes, namely, forms with predominant diarrhea (IBS-D), with prevailing constipation (IBS-C subtype), or mixed defecation types (IBS-M), as well as IBS that cannot be subdivided [5,6]. Besides altered bowel habits, IBS patients may also suffer from abdominal pain or discomfort, flatulence, nausea, dyspepsia, or reflux [7].
The etiology of IBS is still unknown. However, many factors could be responsible for the development of the disease. Besides psychological disturbances, the altered intestinal motility, food hypersensitivity, genetics, abnormalities of the intestinal microbiota, and impairment of the bidirectional communication pathways between the gut, its microbiota, and the central nervous system, called the gut–brain axis (GBA), could trigger the disease [8,9]. Moreover, bacterial overgrowth or post-infectious (PI) changes in the gastrointestinal tract (GIT) and inflammation are acknowledged as IBS initiators [10]. Physical or sexual abuse in childhood, a short period of breastfeeding, food allergies, obesity, or surgical interventions might also lead to the evolvement of the disease [6]. IBS patients are more prone to exhibit psychological disorders, such as anxiety or depression. This makes them less receptive to IBS treatment when it is introduced alone [11]. Therefore, psychological therapy is recommended for these individuals either as an alternative to medication or to support it [12].
Antispasmodic, antidiarrheal drugs, and laxatives are the most commonly used pharmaceuticals to treat IBS, but long-term use can lead to severe side effects [13]. Additionally, acupuncture and massage, along with traditional Chinese medicine (TCM; e.g., Huoxiang Zhengqi Soft Capsule, Guchang Zhixie Pill, Shugan Decoction, and Sishen Pill), which comprise natural plant-based ingredients, are applied as remedies for IBS [13,14]. Another promising approach to alleviate IBS is targeting the gut microbiota, for which probiotics, prebiotics, and synbiotics can be used [15]. Probiotics are defined as live microorganisms that confer a health benefit on the host, while prebiotics are substrates for the favorable microorganisms inhabiting the host’s GIT [16,17,18]. Synbiotics combine probiotics and prebiotics to improve the health effects on the host compared to using these components separately. There are two types of synbiotics: synergistic, in which prebiotic serves as a substrate for administered probiotic microorganisms, or complementary, which influence the host’s indigenous microbiota [19].
This review aimed to recapitulate the literature in the field of IBS since its early diagnosis and the first treatment attempts, as well as compile the information on the alteration of GIT microbiota among IBS individuals. Above all, the following paper intended to summarize research on the use of probiotics, prebiotics, and synbiotics in the treatment of IBS and to assess the limitations of the studies cited.

2. Method

The literature search was conducted until June 2021 in the PubMed Central database. Terms: “IBS”, “Irritable Bowel Syndrome”, “Irritable Bowel”, “IBS AND Microbiota”, “Irritable Bowel AND Microbiota”, “IBS AND Probiotics”, “Irritable Bowel AND Probiotics”, “IBS AND Prebiotics”, “Irritable Bowel AND Prebiotics”, “IBS AND Synbiotics”, “Irritable Bowel AND Synbiotics” were used in the field of article’s title, abstract, as well as keywords. We included studies of all types and did not restrict the search by publication date or IBS diagnosis criteria.

3. History of the Illness Recognition

The history of the IBS diagnosis began in 1871 when da Costa described a condition called membranous enteritis in which patients suffered from intestinal pain accompanied by excretion of mucus [20,21]. Doctor Hale-White, who studied patients with various conditions of organic origin (e.g., colon cancer, ulcerative colitis, or appendiceal abscess), mentioned the disease at the beginning of the 20th century [22]. On the other hand, when doctor Herbert P. Hawkins (1906) analyzed reasons for the misdiagnosis of appendicitis, he concluded that there are several symptoms associated with intestines functionality, which do not have an origin in any pathological changes. The scientist speculated that constipation, diarrhea, intestinal spasms, and abdominal pain can have a nervous etiology [23]. Doctor John R. Ryle investigated a similar problem, which he termed spastic colon, as he also observed a number of unnecessary abdominal surgeries that failed to provide relief to patients suffering from chronic abdominal pain. He emphasized the vital importance of a detailed diagnosis, focusing on continuous pain, not typical for acute enteritis or bowel obstruction, as well as unusual palpability of patients’ colon caused by muscle spasm [24]. In 1937, doctor Earle P. Scarlett agreed that Ryle’s term “spastic colon” was more appropriate, rather than “colitis”, which should be used only in cases of clearly demonstrated inflammation of the colon. Additionally, the “irritable colon” appellation, first introduced by doctor Sippy, was mentioned as the equally appropriate term for this spectrum of symptoms. Although, doctor Scarlett pointed out that the disturbance of functionality might not only affect the colon but the whole intestines [25]. In the following years, IBS was found to be related to the overstimulated autonomic nervous system and patients’ personality, which, together with X-ray examinations and observations of symptomatology, led to correct diagnostics [26]. Furthermore, Misiewicz, Wallet, and Eissner (1966) observed the impact of elevated levels of 5-hydroxytryptamine (5-HT, serotonin), an amine produced in the alimentary tract, on intestinal motility resulting in diarrhea. It marked the beginning of extensive studies on the role of serotonin in the etiology of IBS [27]. Moreover, the role of the gut microbiota in IBS etiology has been extensively analyzed since the 1980s [28]. It was not until the late 1990s that gastroenteritis was found to be a contributing factor to the development of IBS [29,30]. However, the differences between the fecal microbiota of healthy individuals and IBS patients were not described until the early 2000s [31,32].
In 1962, Chaudhary and Truelove, who studied IBS as a spectrum including both mucus colitis and spastic colon, divided 130 patients into those with painless diarrhea and ones with abdominal pain, among whom Ritchie and Tuckey (1969) observed similarities in colon motor activity, although bowel function ranged from normal to diarrhea or constipation [21,22]. Meanwhile, Connell (1968) described diagnostic criteria based on the combination of symptoms such as abdominal pain and bowel function abnormalities without pathological changes [33]. Ritchie (1970) added the sensitivity of the colon while being pressured, as well as the profuse excretion and transition of the mucus to the list of typical symptoms [21]. Both researchers stressed the significance of excluding other gastroenterological diseases that might present with similar manifestations [21,33]. Moreover, Manousos et al. (1967) analyzed intestinal content transition time in 75 individuals with irritable colon syndrome, 43 patients with diverticulosis, and 88 subjects who had no abnormalities in bowel functions. The results showed that people suffering from both irritable colon and diverticulosis had a shortened transition time of food through the digestive tract. The researchers believed it might be related to a disturbance in colonic muscle function [34]. However, Cann et al. (1983) noted changed food transition time not only in the colon but also in the small intestine. It proved that IBS should be considered a disease affecting the whole intestine. The researchers also described that individuals with constipation had a prolonged transition time of intestinal content, whereas diarrheic ones shortened [35].
Since 1978, researchers and physicians have referred to the criteria described by Manning et al. (1978). They included flatulence, pain relief after intestinal movement, frequent and looser stools, the presence of mucus, or the impression of incomplete defecation as a highly possible differentiation of IBS from organic diseases [36]. A few years later, Kruis drew attention to the duration of symptoms as an essential diagnostic criteria, which was not universally accepted [37]. Subsequently, the Rome criteria for IBS were first presented at the 13th Rome Congress in 1988 and published as a result of Thomson, Drossman, Heaton, Dotteval, and Kruis collaboration [38]. The Rome Committee continued its work on the proper definition and diagnosis of IBS, which was amended in 1992, 1999, and 2006. The latest version was presented in 2016 as the Rome IV criteria. It states that IBS must manifest with abdominal pain relapsing at least one day per week for the past three months, began at least six months before diagnosis, and correlate with no less than two of the following criteria: being linked to defecation, be associated with shifts in stool regularity and/or its form [37]. Besides the typical symptoms of IBS, nausea, vomiting, heartburn, or even anxiety, insomnia, or depression have been observed in some patients suffering from the disease [39].
As more became known about IBS, more attention was paid to the treatment methods. In 1966, doctor MacDougall recommended a psychological and physical approach to managing the disease. This included reducing bowel stimulation through a diet adapted to the observed symptoms, the use of pharmaceuticals (e.g., codeine phosphate, diphenoxylate, propantheline), and reducing stress [40]. Later, in 1977, Diamant published a review on irritable colon syndrome in which he analyzed the latest reports on the treatments of the disease. The researcher drew attention to the importance of psychological factors since almost 50% of patients responded positively to placebo in various trials. However, Dotevall and Groll (1974), in their studies on mepiprazole, a type of tranquilizer, found that the placebo was ineffective after some time compared to the analyzed substance. In addition to tranquilizers, antispasmodic drugs such as anticholinergics or mebeverine, as well as agents to increase stool mass, were also prescribed by physicians for patients with IBS, either individually or as a combination of treatments [41]. Diamant (1977) also suggested that a low-fiber diet may be responsible for the development of the syndrome. The researcher proposed the adjustment of the dosage of dietary fibers to the observed symptoms of patients as an approach to treat the disease [42]. Nowadays, the European Food Safety Authority (EFSA) defines dietary fibers as non-digestible carbohydrates, such as pectins, cellulose, resistant starch, and non-starch polysaccharides, or fructooligosaccharides, as well as lignin [43]. At the end of the 20th century, psychological treatments (e.g., psychotherapy, behavioral or group therapy, and hypnotherapy) and the exclusion of certain foods, were also acknowledged as an additional or alternative approach to the medical treatment of IBS [44]. Simultaneously, preliminary studies of agents modifying 5-HT receptors’ functionality had begun [45]. To date, the greatest interest in the pharmacological management of IBS patients is focused on receptor subtypes 5-HT3 and 5-HT4 due to their impact on the functionality of GIT [46]. In the early 2000s, it was hypothesized that 5-HT3 receptor antagonists such as ondansetron and granisetron might be effective in IBS treatment, while cilansetron was already approved for use [47]. Moreover, studies have been conducted on 5-HT4 receptor agonists, namely, tegaserod or prucalopride [48]. Currently, new 5-HT3 receptor antagonists and 5-HT4 receptor agonists are still being sought [49,50].
Since the late 1970s, researchers have searched for natural ways of symptomatic treatments of IBS because of the side effects associated with prolonged pharmaceutical treatment. One of the alternatives was peppermint oil, whose antispasmodic properties were observed both in vitro and in vivo by Rees et al. (1979) [51]. However, it was not until the turn of the 20th to the 21st century that probiotics began to be investigated as a means of treating IBS [28]. Furthermore, Hunter et al. (1999) initiated studies on prebiotics as management of IBS [52]. Nowadays, synbiotics are also a subject of interest in IBS management analysis. However, even after 2010, there was not much data available on synbiotics’ impact on gastrointestinal disorders [53,54]. To date, research groups have focused on probiotics, prebiotics, and synbiotics as means to help IBS patients.

4. Intestinal Microbiota in Patients with Irritable Bowel Syndrome

Human GIT is colonized by up to 1014 organisms belonging to about 1000 species, among which prokaryotes (bacteria and unicellular microbes) dominate. However, fungi, archaea, parasites, and viruses are also inhabitants of the gut [55,56]. Bacteria residing in the GIT are mostly classified into four phyla, namely, Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria [57]. Differences in the GIT microbiota of individuals can be attributed to the type of delivery and feeding of the infant, age, sex, diet, sanitary and living conditions, health issues, and administrated pharmaceuticals, as well as geographical regions [58]. Nevertheless, dysbiosis of the GIT microbiota can trigger immune system responses that result in inflammation in the gut and disruption of the GBA [59]. The imbalance of the gut microbiota, which may be associated with overgrowth or lack of certain microorganisms, or genetic abnormalities, can lead to a variety of cardiovascular, neurological, or intestinal diseases, one of which is IBS [19,57].
Pittayanon et al. (2019) conducted a systematic review of 24 studies on the microbiota of IBS patients and observed significant differences in the results. These researchers found that in some studies, the number of potentially harmful bacteria from Enterobacteriaceae family and Bacteroides genus was increased. Simultaneously, a decrease in the prevalence of representatives of the beneficial microbiota, namely, genus Bifidobacterium and Faecalibacterium, was observed in individuals with IBS compared to the healthy ones [60]. Chong et al. (2019) made analog observations in their review. Additionally, the article mentioned the increased number of bacteria belonging to Firmicutes phyla, including Lactobacillus and Ruminococcus genus, as well as a decreased abundance of Erysipelotrichaceae and methanogens in IBS patients [61]. Mei et al. (2021), who analyzed only IBS-D patients in China, also observed elevated abundance of bacteria belonging to Enterobacteriaceae family, as well as Proteobacteria phyla, and decreased prevalence of Firmicutes, Fusobacteria phyla, and Alloprevotella, Fusobacterium genus in contrast to the healthy population [62]. Tap et al. (2017) observed the correlation between the severity of IBS and the number of Methanobacteriales capable of producing methane, which is linked to the occurrence of constipation [63]. In addition, the research group noted the reduced number of bacteria from Prevotella genus in IBS patients compared to healthy subjects. However, Su et al. (2018) and Barandouzi et al. (2021) found increased abundance of this genus in IBS individuals [63,64,65]. What is more, Shukla et al. (2015) found a lower abundance of Bifidobacterium and Lactobacillus genus and increased prevalence of Veillonella genus, Clostridium coccoides, Bacteroides thetaiotamicron, Ruminococcus productus, and P. aeruginosa [66]. Although several studies have described an increased ratio of Firmicutes to Bacteroidetes in patients with IBS compared to healthy individuals, Barandouzi et al. (2021) observed a similar abundance of bacteria belonging to these two phyla, that constitute about 90% of the total microbiota of GIT [65,67]. This research group observed, among IBS patients, a higher prevalence of bacteria belonging to Verrucomicrobia phyla, as well as from Blautia genus, which is acknowledged as a marker of microbiota imbalance [65]. Similarly, Lee et al. (2021) found no significant differences in phyla levels in fecal samples of IBS patients and healthy population. However, these researchers observed an increase in pathogenic bacteria from Desulfovibrionaceae family, and a simultaneous decrease in the beneficial Lachnospiraceae family [68]. On the contrary, Ahluwalia et al. (2021) did not notice any differences between the fecal microbiota of IBS and healthy individuals. They concluded the variations were likely related to microbiota functionality rather than composition [69]. Moreover, Dlugosz et al. (2015) reported no significant differences in the small intestine microbiota of IBS and healthy subjects [70].
Despite the efforts of researchers to find the dysbiosis patterns in the microbiota of people suffering from IBS, there are still many inconsistencies in the obtained results. It could be due to different GIT parts from which samples were gathered, different analytical methods, or even population disparity [71].

5. Probiotics, Prebiotics, and Synbiotics in IBS Treatment

Growing evidence of GIT microbial population disturbances and gastroenteritis being factors of IBS development resulted in the search for therapies based on microbiota manipulations. For this purpose, probiotics, prebiotics, and synbiotics could be used [72,73]. To date, researchers have carried out multiple studies on the impact of various probiotic strains, prebiotics, and their mixtures on people suffering from IBS. A few studies were conducted on animal models which can be applied if the etiology of the induced disease is as similar as possible as it is in humans. These models play an important role in pre-clinical research on the treatment or mechanisms of functional gastrointestinal disorders, including IBS [74]. Since psychological pressure might cause the development of IBS or provoke its symptoms in humans, stress is an inducing factor for most animal models. Nevertheless, IBS might emerge in individuals after infections; therefore, the post-infectious animal models are also used, which are caused by pathogenic bacteria or parasites. Chemical or mechanical stimulation might also trigger IBS symptoms in rodents [75].

5.1. Probiotics

The probiotic effect is attributed to the strain and even two different strains of the same species might impact the patient to various extension [76,77]. Therefore, the influence of one probiotic cannot be extrapolated to another one from the same species or even to a different strain [78]. Moreover, probiotics can act differently in various populations, as well as stages and types of diseases [77,78].
In 2002, Sen et al. published a study on the influence of Lactobacillus plantarum 299V on IBS patients. The strain did not exhibit any beneficial effect on the disease symptoms [79]. On the contrary, Ducrotté et al. (2012) described the potentially beneficial impact of the strain on the IBS symptoms of studied subjects [80]. Nevertheless, the research conducted by Sen et al. (2002) was not the only one leading to the conclusion that tested probiotic might not be effective in IBS treatment [79]. Pedersen et al. (2010), Simrén et al. (2010), Søndergaard et al. (2011), Amirimani et al. (2013), Roberts et al. (2013), Lorenzo-Zúñiga et al. (2014), and Cremon et al. (2018) obtained analog results [81,82,83,84,85,86,87]. Both Simrén et al. (2010), and Søndergaard et al. (2011) analyzed probiotic Cultura, which was milk fermented with Lactobacillus bulgaricus, as well as Streptococcus thermophilus, and containing three probiotic strains, namely, Lactobacillus paracasei F19, Lactobacillus acidophilus La5, and Bifidobacterium animalis subsp. lactis Bb12 [82,83]. Pedersen et al. (2010), who studied the same probiotic fermented milk product, observed that acidified milk itself caused the improvement in IBS symptoms [81]. Robert et al. (2013) also studied dairy product, which was yogurt with starter cultures of S. thermophilus CNCM I-1630, Lb. bulgaricus CNCM I-1632 and Lb. bulgaricus CNCM I-1519, as well as probiotic strain B. lactis DN-173 010 [85]. On the other hand, Amirimani et al. (2013) analyzed the influence of probiotic tablets Biogaia® containing Lactobacillus reuteri on IBS patients [84]. Lorenzo-Zúñiga et al. (2014) conducted research on encapsulated multi-strain probiotic, including Lb. plantarum CECT7484, Lb. plantarum CECT7485, and Pediococcus acidilactici CECT7483, whereas Cremon et al. (2018) analyzed the influence of capsules containing Lb. paracasei CNCM I-1572 [86,87]. Furthermore, Ligaarden et al. (2010) observed an unfavorable effect of encapsulated Lb. plantarum MF1298 on IBS subjects [88].
However, there are several trials, which proved the beneficial influence of probiotics on IBS symptoms and their severity. Among them, there were probiotic capsules with Bifidobacterium infantis 35,624, or Bifidobacterium bifidum MIMBb75, or Lactobacillus brevis KB290 studied by Whorwell et al. (2006), Guglielmetti et al. (2011), and Murakami et al. (2012), respectively [89,90,91]. Additionally, Martoni et al. (2020) performed the analysis involving two separate probiotics Lb. acidophilus DDS®-1, and B. lactis UABla-12™, in a form of capsules. Besides improvement in overall IBS symptoms, Martoni et al. (2020) described the positive influence of both probiotic strains on stool consistency and the severity of abdominal pain. Additionally, Lb. acidophilus DDS®-1 contributed to the reduction of stress levels of IBS individuals [92]. Caviglia et al. (2020) and Zhou et al. (2020) used Bifidobacterium langum as a probiotic in studies conducted on humans and rats, respectively [93,94]. Caviglia et al. (2020), who analyzed strain B. langum ES1, observed improvement in overall IBS symptoms and enhanced intestinal barrier integrity, as well as immune-inflammatory state of IBS-D patients [93]. Similarly, Zhou et al. (2020) noted the improvement of intestinal permeability, along with positive changes in GIT microbiota in the rat WAS (water avoidance stress) model after B. langum treatment [94]. However, contrary to Caviglia et al. (2020), the research team did not establish any impact of B. langum on serum cytokines levels in rats [93,94]. Lewis et al. (2020) conducted a trial involving two separate probiotics. Besides B. longum HA-196 Lb. paracasei R0175 was administrated to IBS individuals. Both probiotics ameliorated the social quality of participants’ lives. Although, only B. langum HA-196 contributed to the increase of Bifidobacterium genus abundance in fecal samples, which was not observed for Lb. paracasei R0175 and corresponding Lactobacillus spp. [95]. What is more, Lb. gasseri BNR17 has been described by Kim et al. (2018) as suitable for use as IBS treatment since it improved its symptom [96]. Shin et al. (2018) analyzed the same strain and noted its impact on intestinal microbiota, as well as bowel habits of trial participants [97]. Lb. casei is yet another species from Lactobacillus genus, which researchers studied as probiotics in IBS treatment [98,99]. Compare et al. (2017), who performed ex vivo analysis on ileal and colonic mucosa culture tissue model harvested from PI-IBS-D patients, established that Lb. casei DG is able to diminish mucosal inflammation [98]. On the other hand, Seong et al. (2021) conducted a study on a rat IBS model induced with chronic restrain stress. Those researchers also observed the capability of Lb. casei DKGF7 to decrease inflammatory cytokines in colonic tissue, as well as serum corticosterone levels, along with amelioration of IBS symptoms and increased expression of tight junction proteins [99]. Furthermore, Dapoigny et al. (2012) described the trial in which participants received Lb. rhamnosus. This probiotic improved symptoms of IBS only among individuals suffering from the IBS-D subtype [100]. Other effective probiotics in improving IBS symptoms are strains of Bacillus coagulans [101,102,103,104]. Majeed et al. (2018) observed that B. coagulans MTCC 5856 could reduce sleeplessness and depression in IBS-D patients [102]. Gupta et al. (2012) noted the beneficial impact of B. coagulans LBSC on GIT microbiota of IBS individuals [104]. Likewise, Sun et al. (2018) used Clostridium butyricum as IBS treatment in the trial, in which probiotic exhibited the ability to ameliorate symptoms of the disease. However, no significant impact on the intestinal microbiota of tested subjects was observed [105]. Zhao et al. (2019) introduced C. butyricum probiotic strain to mice PI-IBS model induced with 2,4,6-trinitrobenzenesulfonic acid (TNBS). They described a decrease in intestinal visceral hypersensitivity, along with reduced low-grade mucosal inflammation [106]. Interestingly, only Kruis et al. (2012) used Escherichia coli Nissle 1917 (MUTAFLOR®) in their trial on IBS-C and IBS-D patients and concluded that it poses therapeutic potential for PI-IBS, as well as post-antibiotic IBS individuals [107]. Other rarely studied probiotics are Saccharomyces spp. yeast, among which Saccharomyces cerevisiae CNCM I-3856 strain was used as probiotic in de Chambrun et al. (2015) and Spiller et al. (2016) studies. De Chambrun et al. (2015) observed reduced severity of abdominal pain and discomfort, whereas Spiller et al. (2016) noted overall improvement of IBS symptoms but only among IBS-C subjects [108,109]. Moreover, Abbas et al. (2014) described the potential of Saccharomyces boulardii to reduce inflammation in the GIT of IBS-D individuals [110]. Probiotic S. boulardii yeast were also used as a component of multi-strain preparations studied by Hong et al. (2019), as well as Leventogiannis et al. (2019) [111,112]. Hong et al. (2019), who used the PI-IBS mice model, established that studied probiotic preparation composed of S. boulardii, Lb. acidophilus LA-5, and B. lactis BB-12, contributed to the reduction of serum levels of pro-inflammatory cytokines, and visceral hypersensitivity [111]. Leventogiannis et al. (2019) conducted human studies and noted that Lactolevure® probiotic, including S. boulardii, B. lactis BB-12, Lb. acidophilus LA-5, and Lb. plantarum, can be effective in attenuation of bloating and abdominal pain severity, especially in patients with IBS combined with small intestinal bacterial overgrowth (SIBO) [112]. Reduction of SIBO was also observed by Barret et al. (2008), who studied Yakult® dairy product containing Lactobacillus casei Shirota. This probiotic also decreased the early rise in breath hydrogen after lactulose (ERBHAL), and, simultaneously, abdominal pain [113]. Lee et al. (2018) described the comparable observation concerning the influence of probiotic on SIBO. Their trial involved only IBS-D patients treated with a multi-strain probiotic (Ther-Biotic® Complete) containing S. thermophilus, along with strains belonging to Lactobacillus spp. (7) and Bifidobacterium spp. (4) [114].
Multi-strain probiotic preparations are widely studied as the mode of IBS management [81,82,83,85,86,111,112,114,115,116,117,118,119,120,121,122,123,124,125,126]. They can be effective in attenuation of IBS symptoms, such as, ones including Lb. acidophilus SDC 2012, along with Lb. acidophilus SDC 2013 tested by Sinn et al. (2008), as well as a bifid triple viable capsule containing B. longum, and Lb. acidophilus used in Cui and Hu (2012) trial, or Bio-Kult® composed of five Bifidobacterium spp. and six Lactobacillus spp. strains, along with S. thermophilus PXN 66 analyzed by Ishaque et al. (2018) [115,118,123]. Sisson et al. (2014) described another multi-strain probiotic Symprove, comprising Lb. rhamnosus NCIMB 30174, Lb. plantarum NCIMB 30173, Lb. acidophilus NCIMB 30175, and Enterococcus faecium NCIMB 30176, which exhibited a beneficial impact on IBS symptoms [119]. Oh et al. (2019), also described the amelioration of IBS symptoms among participants of the trial, which excluded individuals suffering from IBS-C, after treatment with Foodies Lactobacillus (Lb. paracasei, Lb. salivarius, Lb. plantarum) [124]. The results of this study indicate the ability of the preparation to reduce the severity of abdominal pain, which was similar to results obtained by Hong et al. (2009), Zhang et al. (2019), and Skrzydło-Radomańska et al. (2021) [116,124,125,126]. Hong et al. (2009) used the preparation including B. bifidum BGN4, B. lactis AD011, Lb. acidophilus AD031, and Lb. casei IBS041 [116]. Bifico® (B. longum, Lb. acidophilus, E. faecalis) administrated to IBS-D individuals in the trial conducted by Zhang et al. (2019) not only reduced abdominal pain but also exhibited beneficial impact on GIT microbiota and short-chain fatty acids (SCFAs) concentrations, as well as reduced plasma levels of cytokines [125]. Skrzydło-Radomańska et al. (2021) observed additional improvement in quality of life after treatment of IBS-D patients with NordBiotic™ including S. thermophilus ST250, as well as Lactobacillus spp. (5), and Bifidobacterium spp. (3) strains [126]. Michail and Kenche (2011), as well as Hod et al. (2018), focused their research on the influence of multi-strain probiotics on IBS-D patients’ microbiota [117,122]. Michail and Kenche (2011) did not observe any impact of VSL#3 probiotic (S. thermophilus, B. breve, B. longum, B. infantis, Lb acidophilus, Lb plantarum, Lb paracasei, Lb. bulgaricus) on fecal microbiota, and, additionally, no significant changes in participants’ body mass index (BMI) [117]. Additionally, Hod et al. (2018), who analyzed BIO-25, composed of S. thermophilus ST3, Lactococcus lactis SL6, along with Lactobacillus spp. (6), and Bifidobacterium spp. (4) strains, in management of IBS-D, noted that the preparation did not affect microbial diversity in participants’ feces. Nevertheless, they observed increased abundance of Lactobacillus spp. and Lactococcus spp. in stool samples of subjects whose abdominal pain and bloating were reduced, and a simultaneous decrease in Bilophila genus prevalence among individuals with decreased abdominal pain and improved stool consistency [122]. Furthermore, Yoon et al. (2015), established that LacClean Gold-S® (B. bifidum KCTC 12199BP, B. lactis KCTC11904BP, B. longum KCTC 12200BP, Lb. acidophilus KCTC 11906BP, Lb. rhamnosus KCTC 12202BP, S. thermophilus KCTC 11870BP) improve symptoms only among patients with IBS-D, whereas probiotic strain included in the preparation were present in fecal samples of all trial participants [120]. Comparable results, on two different products containing Lb. acidophilus DSM 24936, and Lb. reuteri DSM 25175, or Lb. rhamnosus DSM 25568, Lb. plantarum DSM 24937, and B. lactis DSM 25566, were obtained by Mezzasalma et al. (2016) among individuals suffering from IBS-C [121].
Most of the cited research was carried out in double-blind mode; however, there are still some trial designs conducted without placebo, or control group [93,112,113,114,125], as well as research conducted in full awareness of investigators, and/or participants of the administrated product (treatment/control) [81,93,112,113,114,125]. Moreover, the number of studies including the dose-related effect of probiotics on IBS subjects is very limited [86,89,96]. Animal or ex vivo culture tissue models are also rarely used for the analysis of active agents’ potential impact on IBS individuals [94,98,99,111]. Most of the above-described studies are focused on all IBS subtypes [80,81,82,87,88,89,95,100,108,109,112,113,115,116,118,119,120]. Although, several studies did not differentiate the form of the disease among participants [79,83,84,85,90,91,92,96,101,103,104,127]. Moreover, Oh et al. (2019) performed the analysis of probiotic impact on IBS patients with the exclusion of subjects suffering from IBS-C [124]. This subtype was chosen as the inclusion criteria only by Kruis et al. (2012) and Mezzasalma et al. (2016) [107,121]. However, Kruis et al. (2012) included also people with IBS-D, which was the subtype on which Michail and Kenche (2011), Abbas et al. (2014), Lorenzo-Zúñiga et al. (2014), Majeed et al. (2016), Compare et al. (2017), Hod et al. (2018), Ishaque et al. (2018), Lee et al. (2018), Shin et al. (2018), Sun et al. (2018), Zhang et al. (2019), Caviglia et al. (2020), and Skrzydło-Radomańska et al. (2021) focused [86,93,97,98,102,105,107,110,114,117,122,123,125,126]. Details of the above-mentioned trials are listed in Table 1.

5.2. Prebiotics

At present, only a few pieces of research on the impact of prebiotics on the health improvement of people suffering from IBS have been conducted. Therefore, this area of study is still in need of investigation.
The first study on the influence of prebiotics on IBS management was published in 1999 by Hunter et al., who analyzed the oligofructose, the effect of which was marginal and only in patients with IBS-C [52]. Similarly, Olesen et al. (2000), who studied fructooligosaccharides (FOS), did not conclude whether use of prebiotic helped to improve the condition of IBS patients or not [128]. Short-chain FOS (scFOS) was also studied by Azpiroz et al. (2016), who described the influence of the prebiotic on the anxiety level of IBS individuals and Bifidobacterium spp. count in their stool [129]. Silk et al. (2009) tested the effectiveness of another prebiotic, namely, the trans-galactooligosaccharide (GOS) mixture produced by Bifidobacterium bifidum NCIMB 41171 from lactose. They observed that it not only relieved symptoms such as flatulence, abdominal pain, and discomfort, as well as stool patterns but also increased the number of Bifidobacterium spp. in fecal samples [130]. Both GOS and FOS, along with inulin and anthocyanins, were included in the preparation used in the trial conducted by Chen et al. (2017) in the IBS mice model. The product exhibited the ability to diminish inflammation and improve the intestinal barrier. Additionally, if used prior to infection, it could help to establish PI microbial homeostasis in the GIT. Nonetheless, the blend of prebiotics needs to be further investigated in humans [131]. Niv et al. (2016) proved the effectiveness of partially hydrolyzed guar gum (PHGG) for IBS patients suffering mostly from gasses and bloating. The use of this compound did not cause any side effects. However, it did not exhibit any influence on the rest of the possible IBS symptoms [132].
All of the cited research was placebo-controlled [52,128,129,130,132], except from animal studies described by Chen et al. (2017) [131]. Among mentioned human trials, only ones conducted by Olesen et al. (2000) [128], and Silk et al. (2009) [130], were not carried out in a double-blind system. However, the study design by Silk et al. (2009) was the only one including the influence of a dose on prebiotic effectiveness in IBS therapy [130]. Research carried out by Hunter et al. (1999) and Olesen et al. (2000) did not differentiate IBS subtypes [52,128], whereas ones conducted by Silk et al. (2009), Azpiroz et al. (2016), and Niv et al. (2016) included patients exhibiting all forms of the disease [129,130,132]. Details of the above-mentioned research are presented in Table 2.

5.3. Synbiotics

Despite the concept of synbiotics being introduced in 1995, and the first attempts to recognize IBS were made at the end of the 19th century, the idea of using these preparations to treat the disease appeared in the last decade [133].
In 2013, Cappello et al. performed the first analysis of Probinul® and its impact on IBS individuals. The synbiotic included inulin, tapioca-resistant starch, and Lactobacillus spp. (6) and Bifidobacterium spp. (2) strains, as well as Streptococcus thermophilus. Probinul® did not diminish flatulence and bloating to the satisfying level according to participants of the trial [134]. Shavakhi et al. (2014), who studied the impact of Balance®, which included FOS, and probiotic strains from Lactobacillus (4) and Bifidobacterium (3) genus, as well as S. thermophilus, did not observe any influence of the preparation on IBS patients [135]. Similar results were obtained by Bogovič Matijašic et al. (2016), who studied synbiotic fermented milk product containing Lb. acidophilus La-5, B. lactis BB-12, and 2% dietary fiber (Beneo Orafti Synergy1; 90% inulin, 10% oligofructose) [136]. On the contrary, Bucci et al. (2014) noted the attenuation of flatulence in IBS subjects after 4 weeks of the disease treatment with Probinul®, which was sustained during a 6 months period of therapy [137]. Another synbiotic, namely, Lactol®, including B. coagulans and FOS, was tested by Rogha et al. (2014). The research team noted relief of abdominal pain, discomfort, and diarrhea in IBS individuals; however, no improvement in constipation-related symptoms was observed. Nonetheless, the study revealed some side effects of the preparation; therefore, its safety has to be further evaluated [54]. Although, Asgarshirazi et al. (2015), who analyzed the effectiveness of Lactol® in treating functional abdominal pain in children, did not notice any side effects of the preparation [138]. Moser et al. (2019) conducted a study on the impact of yet another synbiotic preparation named OMNi-BiOTiC® Stress Repair on IBS-D patients. The preparation comprised prebiotics, namely, corn starch, maltodextrin, inulin, and FOS, along with Lactococcus lactis W19, Lactobacillus spp. (5), and Bifidobacterium spp. (3) strains. Results showed a positive influence of the mixture on mucosal microbiota diversity and concentrations of acetate and butyrate in fecal samples [139]. Lee et al. (2019) studied the impact of another synbiotic preparation, containing inulin, FOS, and probiotic strains from Lactobacillus (6) and Bifidobacterium (2) genus on IBS patients. The research team observed improvement in bloating, fatigue, and abdominal discomfort in trial participants [140]. On the other hand, Min et al. (2012) and Bahrudin et al. (2020) analyzed the influence of synbiotic dairy products, such as yogurt with the addition of Bifidobacterium animalis subsp. lactis Bb-12 and acacia dietary fiber, and drink containing Lb. helveticus and polydextrose as a prebiotic, respectively, on IBS subjects [141,142]. Min et al. (2012) described that the studied product could attenuate symptoms of the disease in both IBS-C and IBS-D patients [141]. Bahrudin et al. (2020) observed the beneficial effect of synbiotic only in studied IBS-C individuals. Nevertheless, the researchers concluded that probiotic strain alone is the active agent [142]. The effective mixture of pro- and prebiotics for IBS-D patients, which reduce the feeling of incomplete intestinal movements, release abdominal pain, and help to regulate stool patterns, have been described by Skrzydło-Radomańska et al. (2021). The synbiotic comprised probiotics, namely, Lb. rhamnosus FloraActive 19070-2, Lb. acidophilus DSMZ 32418, B. lactis DSMZ 32269, B. longum DSMZ 32946, B. bifidum DSMZ 32403 strains, and scFOS as a prebiotic compound [143]. Last but not least, in 2020, Seong et al. described a synbiotic, containing Lb. paracasei DKGF and Opuntia humifusa extract as a prebiotic, effective in IBS murine model. Nevertheless, its functionality must be further assessed in humans [144].
Most of the cited research [54,134,135,136,137,140,142,143] was conducted as double-blind, placebo-controlled, except from Moser et al. (2019) [139], as well as Seong et al. (2020) [144], who performed the animal study. Two studies were conducted on people suffering from the IBS-C subtype [136,142], and the other two on subjects with IBS-D [139,143]. Four studies did not differentiate subtypes of the disease [54,134,135,137], whereas the rest of mentioned research focused on all subtypes [140,141]. Furthermore, only one trial involved dose-related dependencies [140]. Detailed descriptions of the aforementioned studies are listed in Table 3.

6. Summary

In conclusion, the use of probiotics, prebiotics, and synbiotics in IBS treatment is still in need of investigation and standardization. Researchers focused mostly on probiotics, especially multi-strain ones, creating a demand for new research on prebiotics, and synbiotics in the management of IBS.
The vast majority of trials are double-blind and placebo-controlled; however, there are still studies conducted in an open-label system, as well as with no control or placebo group, which could bring unreliable results. Additionally, analyzed preparations differ in probiotic strains, their number, and density, as well as amount and type of prebiotic or their combination, in the case of synbiotics. Moreover, the dosage of studied preparations varied among research, and its impact is rarely analyzed. Most of the trial designs would benefit from a longer treatment period and additional follow-up phase since this aspect is mostly described by researchers as a study limitation. Another obstruction of research might be the number of participants that rarely exceeded 100. However, this obstacle is hard to avoid because individuals are entering the trials voluntarily. Lastly, not every probiotic, prebiotic, and their combination would be an appropriate mode of treatment for each IBS subtype, which also needs to be studied in more detail.
Nevertheless, the last two decades of research on probiotics, prebiotics, and synbiotics bring satisfying results and they are acknowledged as effective and safe in IBS therapy. These preparations can be introduced as an alternative to drugs that might carry a risk of side effects, especially in long-term use. Among the field of microbiota-manipulation-based therapies, probiotics, prebiotics, and synbiotics are a promising direction of alleviation of symptoms for people suffering from IBS.

Author Contributions

Conceptualization, A.C.-W.; formal analysis, A.C.-W.; investigation, A.C.-W., and K.Ś.; writing—A.C.-W. and K.Ś.; writing—review and editing, A.C.-W.; visualization, A.C.-W.; supervision, K.Ś. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ford, A.C.; Sperber, A.D.; Corsetti, M.; Camilleri, M. Irritable Bowel Syndrome. Lancet 2020, 396, 1675–1688. [Google Scholar] [CrossRef]
  2. Üçüncü, M.Z.; Çoruh Akyol, B.; Toprak, D. The Early Diagnosis of Fibromyalgia in Irritable Bowel Syndrome Patients. Med. Hypotheses 2020, 143, 110119. [Google Scholar] [CrossRef] [PubMed]
  3. Hadjivasilis, A.; Tsioutis, C.; Michalinos, A.; Ntourakis, D.; Christodoulou, D.K.; Agouridis, A.P. New Insights into Irritable Bowel Syndrome: From Pathophysiology to Treatment. Ann. Gastroenterol. 2019, 32, 1–11. [Google Scholar] [CrossRef]
  4. Mosińska, P.; Sałaga, M. Clinical Features of Irritable Bowel Syndrome. In Introduction to Gastrointestinal Diseases; Fichna, J., Ed.; Springer: Cham, Switzerland, 2017; Volume 1, pp. 23–27. [Google Scholar]
  5. Ida, M.; Nishida, A.; Akiho, H.; Nakashima, Y.; Matsueda, K.; Fukudo, S. Evaluation of the Irritable Bowel Syndrome Severity Index in Japanese Male Patients with Irritable Bowel Syndrome with Diarrhea. BioPsychoSoc. Med. 2017, 11, 7. [Google Scholar] [CrossRef] [Green Version]
  6. Mokha, J.S.; Hyams, J.S. Irritable Bowel Syndrome. In Pediatric Neurogastroenterology Gastrointestinal Motility and Functional Disorders in Children; Faure, C., Thapar, N., di Lorenzo, C., Eds.; Springer International Publishing: Cham, Switzerland, 2017. [Google Scholar]
  7. Israel, E.J.; Kaplan, J.L.; Fiechtner, L. Gastroenterology and Nutrition: Healthy Eating in Adolescence and Nutritional Supplements; Irritable Bowel Syndrome; Inflammatory Bowel Disease. In The MassGeneral Hospital for Children Adolescent Medicine Handbook; Goldstein, M.A., Ed.; Springer International Publishing: New York, NY, USA, 2017. [Google Scholar]
  8. Van Malderen, K.; De Winter, B.Y.; De Man, J.G.; De Schepper, H.U.; Lamote, K. Volatomics in Inflammatory Bowel Disease and Irritable Bowel Syndrome. EBioMedicine 2020, 54, 102725. [Google Scholar] [CrossRef]
  9. Rutsch, A.; Kantsjö, J.B.; Ronchi, F. The Gut-Brain Axis: How Microbiota and Host Inflammasome Influence Brain Physiology and Pathology. Front. Immunol. 2020, 11, 604179. [Google Scholar] [CrossRef]
  10. Pazhouh, H.K.; Hosseini, S.M.; Taghipour, A.; Hamedi, S.; Noras, M. Anti-Irritable Bowel Syndrome Syrup Improves Constipation-Predominant Irritable Bowel Syndrome: A Randomized, Placebo-Controlled Trial. Chin. J. Integr. Med. 2020, 26, 729–735. [Google Scholar] [CrossRef]
  11. Staudacher, H.M.; Mikocka-Walus, A.; Ford, A.C. Common Mental Disorders in Irritable Bowel Syndrome: Pathophysiology, Management, and Considerations for Future Randomised Controlled Trials. Lancet Gastroenterol. Hepatol. 2021, 6, 401–410. [Google Scholar] [CrossRef]
  12. Shah, K.; Ramos-Garcia, M.; Bhavsar, J.; Lehrer, P. Mind-Body Treatments of Irritable Bowel Syndrome Symptoms: An Updated Meta-Analysis. Behav. Res. Ther. 2020, 128, 103462. [Google Scholar] [CrossRef]
  13. Su, X.-T.; Wang, L.-Q.; Zhang, N.; Li, J.-L.; Qi, L.-Y.; Wang, Y.; Yang, J.-W.; Shi, G.-X.; Liu, C.-Z. Standardizing and Optimizing Acupuncture Treatment for Irritable Bowel Syndrome: A Delphi Expert Consensus Study. Integr. Med. Res. 2021, 10, 100728. [Google Scholar] [CrossRef]
  14. Tang, Z.P. Traditional Chinese Medicine Clinical Experience of the Treatment for Irritable Bowel Syndrome. Chin. J. Integr. Med. 2009, 15, 93–94. [Google Scholar] [CrossRef]
  15. Herndon, C.C.; Wang, Y.P.; Lu, C.L. Targeting the Gut Microbiota for the Treatment of Irritable Bowel Syndrome. Kaohsiung J. Med. Sci. 2020, 36, 160–170. [Google Scholar] [CrossRef]
  16. FAO; WHO. Guidelines for the Evaluation of Probiotics in Food; Food and Agriculture Organization: Rome, Italy; World Health Organization: Geneva, Switzerland, 2002. [Google Scholar] [CrossRef]
  17. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert Consensus Document: The International Scientific Association for Probiotics and Prebiotics Consensus Statement on the Scope and Appropriate Use of the Term Probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [Green Version]
  18. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. 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. 2017, 14, 491–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Núñez-Sánchez, M.A.; Herisson, F.M.; Cluzel, G.L.; Caplice, N.M. Metabolic Syndrome and Synbiotic Targeting of the Gut Microbiome. Curr. Opin. Food Sci. 2021, 41, 60–69. [Google Scholar] [CrossRef]
  20. Schofield, P.F.; Haboubi, N.Y.; Martin, D.F. Irritable Bowel Syndrome. In Highlights in Coloproctology; Schofield, P.F., Haboubi, N.Y., Martin, D.F., Eds.; Springer: London, UK, 1993; pp. 112–115. [Google Scholar]
  21. Ritchie, J.A. The Transport of Colonic Contents in the Irritable Colon Syndrome. Gut 1970, 11, 668–672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Chaudhary, N.A.; Truelove, S.C. The Irritable Colon Syndrome: A Study of the Clinical Features, Predisposing Causes, and Prognosis in 130 Cases. QJM Int. J. Med. 1962, 31, 307–322. [Google Scholar] [CrossRef]
  23. Hawkins, H.P. The Reality of Enterospasm and Its Mimicry of Appendicitis. Br. Med. J. 1906, 1, 65–69. [Google Scholar] [CrossRef] [Green Version]
  24. Ryle, J.A. Chronic Spasmodic Affections of The Colon and The Disease Which They Simulate. Lancet 1928, 212, 1115–1119. [Google Scholar] [CrossRef]
  25. Scarlett, E.P. Functional Disturbances of the Colon: The Irritable (Spastic) Colon. Can. Med. Assoc. J. 1937, 36, 484–489. [Google Scholar]
  26. Gallagher, D.M. Evaluation of Colon Dysfunction. Calif. Med. 1961, 95, 156–158. [Google Scholar]
  27. Misiewicz, J.J.; Waller, S.L.; Eisner, M. Motor Responses of Human Gastrointestinal Tract to 5-Hydroxytryptamine in Vivo and in Vitro. Gut 1966, 7, 208–216. [Google Scholar] [CrossRef] [Green Version]
  28. Madden, J.A.J.; Hunter, J.O. A Review of the Role of the Gut Microflora in Irritable Bowel Syndrome and the Effects of Probiotics. Br. J. Nutr. 2002, 88, s67–s72. [Google Scholar] [CrossRef]
  29. Neal, K.R.; Hebden, J.; Spiller, R. Prevalence of Gastrointestinal Symptoms Six Months after Bacterial Gastroenteritis and Risk Factors for Development of the Irritable Bowel Syndrome: Postal Survey of Patients. Br. Med. J. 1997, 314, 779–782. [Google Scholar] [CrossRef] [Green Version]
  30. García Rodríguez, L.A.; Ruigómez, A. Increased Risk of Irritable Bowel Syndrome after Bacterial Gastroenteritis: Cohort Study. Br. Med. J. 1999, 318, 565–566. [Google Scholar] [CrossRef] [Green Version]
  31. Malinen, E.; Rinttilä, T.; Kajander, K.; Mättö, J.; Kassinen, A.; Krogius, L.; Saarela, M.; Korpela, R.; Palva, A. Analysis of the Fecal Microbiota of Irritable Bowel Syndrome Patients and Healthy Controls with Real-Time PCR. Am. J. Gastroenterol. 2005, 100, 373–382. [Google Scholar] [CrossRef]
  32. Kassinen, A.; Krogius-Kurikka, L.; Mäkivuokko, H.; Rinttilä, T.; Paulin, L.; Corander, J.; Malinen, E.; Apajalahti, J.; Palva, A. The Fecal Microbiota of Irritable Bowel Syndrome Patients Differs Significantly from That of Healthy Subjects. Gastroenterology 2007, 133, 24–33. [Google Scholar] [CrossRef] [PubMed]
  33. Connell, A.M. The Irritable Colon Syndrome. Postgrad. Med. J. 1968, 44, 668–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Manousos, O.N.; Truelove, S.C.; Lumsden, K. Transit Times of Food in Patients with Diverticulosis or Irritable Colon Syndrome and Normal Subjects. Br. Med. J. 1967, 3, 760–762. [Google Scholar] [CrossRef] [Green Version]
  35. Cann, P.A.; Read, N.W.; Brown, C.; Hobson, N.; Holdsworth, C.D. Irritable Bowel Syndrome: Relationship of Disorders in the Transit of a Single Solid Meal to Symptom Patterns. Gut 1983, 24, 405–411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Manning, A.P.; Heaton, K.W.; Thompson, W.G.; Morris, A.F. Towards Positive Diagnosis of the Irritable Bowel. Br. Med. J. 1978, 2, 653–654. [Google Scholar] [CrossRef] [Green Version]
  37. Lacy, B.; Patel, N. Rome Criteria and a Diagnostic Approach to Irritable Bowel Syndrome. J. Clin. Med. 2017, 6, 99. [Google Scholar] [CrossRef]
  38. Thompson, W.G. The Road to Rome. Gastroenterology 2006, 130, 1552–1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Dotevall, G.; Groll, E. Controlled Clinical Trial of Mepiprazole in Irritable Bowel Syndrome. Br. Med. J. 1974, 4, 16–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. MacDougall, I.P. Irritable Colon. J. R. Soc. Med. 1966, 59, 12–14. [Google Scholar] [CrossRef] [Green Version]
  41. Ritchie, J.A.; Truelove, S.C. Treatment of Irritable Bowel Syndrome with Lorazepam, Hyoscine Butylbromide, and Ispaghula Husk. Br. Med. J. 1979, 1, 376–378. [Google Scholar] [CrossRef] [Green Version]
  42. Diamant, N.E. Diarrhea in the Irritable Colon Syndrome. Can. Med. Assoc. J. 1977, 116, 745–746. [Google Scholar]
  43. Barber, T.M.; Kabisch, S.; Pfeiffer, A.F.H.; Weickert, M.O. The Health Benefits of Dietary Fibre. Nutrients 2020, 12, 3209. [Google Scholar] [CrossRef]
  44. Hall, M.J.; Barry, R.E. Current Views on the Aetiology and Management of the Irritable Bowel Syndrome. Postgrad. Med. J. 1991, 67, 785–789. [Google Scholar] [CrossRef] [Green Version]
  45. Francis, C.Y.; Whorwell, P.J. The Irritable Bowel Syndrome. Postgrad. Med. J. 1997, 73, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Madia, V.N.; Messore, A.; Saccoliti, F.; Tudino, V.; De Leo, A.; De Vita, D.; Bortolami, M.; Scipione, L.; Pindinello, I.; Costi, R.; et al. Tegaserod for the Treatment of Irritable Bowel Syndrome. Anti Inflamm. Anti Allergy Agents Med. Chem. 2020, 19, 342–369. [Google Scholar] [CrossRef] [PubMed]
  47. Crowell, M.D. Role of Serotonin in the Pathophysiology of the Irritable Bowel Syndrome. Br. J. Pharmacol. 2004, 141, 1285–1293. [Google Scholar] [CrossRef] [PubMed]
  48. Gunn, M.C.; Cavin, A.A.; Mansfield, J.C. Management of Irritable Bowel Syndrome. Postgrad. Med. J. 2003, 79, 154–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Zheng, Y.; Yu, T.; Tang, Y.; Xiong, W.; Shen, X.; Jiang, L.; Lin, L. Efficacy and Safety of 5-Hydroxytryptamine 3 Receptor Antagonists in Irritable Bowel Syndrome: A Systematic Review and Metaanalysis of Randomized Controlled Trials. PLoS ONE 2017, 12, e0172846. [Google Scholar] [CrossRef] [Green Version]
  50. Fukudo, S.; Nakamura, M.; Hamatani, T.; Kazumori, K.; Miwa, H. Efficacy and Safety of 5-HT4 Receptor Agonist Minesapride for Irritable Bowel Syndrome with Constipation in a Randomized Controlled Trial. Clin. Gastroenterol. Hepatol. 2021, 19, 538–546. [Google Scholar] [CrossRef]
  51. Rees, W.D.W.; Evans, B.K.; Rhodes, J. Treating Irritable Bowel Syndrome with Peppermint Oil. Br. Med. J. 1979, 2, 835–836. [Google Scholar] [CrossRef] [Green Version]
  52. Hunter, J.O.; Tuffnell, Q.; Lee, A.J. Controlled Trial of Oligofructose in the Management of Irritable Bowel Syndrome. J. Nutr. 1999, 129, 1451–1453. [Google Scholar] [CrossRef]
  53. Ghoshal, U.C.; Shukla, R.; Ghoshal, U.; Gwee, K.A.; Ng, S.C.; Quigley, E.M.M. The Gut Microbiota and Irritable Bowel Syndrome: Friend or Foe? Int. J. Inflamm. 2012, 2012, 151085. [Google Scholar] [CrossRef]
  54. Rogha, M.; Esfahani, M.Z.; Zargarzadeh, A.H. The Efficacy of a Synbiotic Containing Bacillus coagulans in Treatment of Irritable Bowel Syndrome: A Randomized Placebo-Controlled Trial. Gastroenterol. Hepatol. Bed Bench 2014, 7, 156–163. [Google Scholar] [CrossRef]
  55. Al Bander, Z.; Nitert, M.D.; Mousa, A.; Naderpoor, N. The Gut Microbiota and Inflammation: An Overview. Int. J. Environ. Res. Public Health 2020, 17, 7618. [Google Scholar] [CrossRef]
  56. Matsumoto, N.; Park, J.; Tomizawa, R.; Kawashima, H.; Hosomi, K.; Mizuguchi, K.; Honda, C.; Ozaki, R.; Iwatani, Y.; Watanabe, M.; et al. Relationship between Nutrient Intake and Human Gut Microbiota in Monozygotic Twins. Medicina 2021, 57, 275. [Google Scholar] [CrossRef] [PubMed]
  57. Mari, A.; Abu Baker, F.; Mahamid, M.; Sbeit, W.; Khoury, T. The Evolving Role of Gut Microbiota in the Management of Irritable Bowel Syndrome: An Overview of the Current Knowledge. J. Clin. Med. 2020, 9, 685. [Google Scholar] [CrossRef] [Green Version]
  58. Zhuang, X.; Xiong, L.; Li, L.; Li, M.; Chen, M. Alterations of Gut Microbiota in Patients with Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis. J. Gastroenterol. Hepatol. (Aust.) 2017, 32, 28–38. [Google Scholar] [CrossRef] [PubMed]
  59. Ooi, S.L.; Correa, D.; Pak, S.C. Probiotics, Prebiotics, and Low FODMAP Diet for Irritable Bowel Syndrome—What Is the Current Evidence? Complementary Ther. Med. 2019, 43, 73–80. [Google Scholar] [CrossRef] [PubMed]
  60. Pittayanon, R.; Lau, J.T.; Yuan, Y.; Leontiadis, G.I.; Tse, F.; Surette, M.; Moayyedi, P. Gut Microbiota in Patients With Irritable Bowel Syndrome—A Systematic Review. Gastroenterology 2019, 157, 97–108. [Google Scholar] [CrossRef] [Green Version]
  61. Chong, P.P.; Chin, V.K.; Looi, C.Y.; Wong, W.F.; Madhavan, P.; Yong, V.C. The Microbiome and Irritable Bowel Syndrome—A Review on the Pathophysiology, Current Research and Future Therapy. Front. Microbiol. 2019, 10, 1136. [Google Scholar] [CrossRef] [Green Version]
  62. Mei, L.; Zhou, J.; Su, Y.; Mao, K.; Wu, J.; Zhu, C.; He, L.; Cui, Y. Gut Microbiota Composition and Functional Prediction in Diarrhea-Predominant Irritable Bowel Syndrome. BMC Gastroenterol. 2021, 21, 105. [Google Scholar] [CrossRef]
  63. Tap, J.; Derrien, M.; Törnblom, H.; Brazeilles, R.; Cools-Portier, S.; Doré, J.; Störsrud, S.; le Nevé, B.; Öhman, L.; Simrén, M. Identification of an Intestinal Microbiota Signature Associated with Severity of Irritable Bowel Syndrome. Gastroenterology 2017, 152, 111–123. [Google Scholar] [CrossRef] [Green Version]
  64. Su, T.; Liu, R.; Lee, A.; Long, Y.; Du, L.; Lai, S.; Chen, X.; Wang, L.; Si, J.; Owyang, C.; et al. Altered Intestinal Microbiota with Increased Abundance of Prevotella Is Associated with High Risk of Diarrhea-Predominant Irritable Bowel Syndrome. Gastroenterol. Res. Pract. 2018, 2018, 6961783. [Google Scholar] [CrossRef] [Green Version]
  65. Barandouzi, Z.A.; Lee, J.; Maas, K.; Starkweather, A.R.; Cong, X.S. Altered Gut Microbiota in Irritable Bowel Syndrome and Its Association with Food Components. J. Pers. Med. 2021, 11, 35. [Google Scholar] [CrossRef]
  66. Shukla, R.; Ghoshal, U.; Dhole, T.N.; Ghoshal, U.C. Fecal Microbiota in Patients with Irritable Bowel Syndrome Compared with Healthy Controls Using Real-Time Polymerase Chain Reaction: An Evidence of Dysbiosis. Dig. Dis. Sci. 2015, 60, 2953–2962. [Google Scholar] [CrossRef]
  67. König, J.; Brummer, R.J. Modulation of the gut ecosystem in irritable bowel syndrome. In Pharma Nutrition: An Overview (AAPS Advances in the Pharmaceutical Sciences Series); Springer: Cham, Switzerland, 2014; Volume 12, pp. 55–73. [Google Scholar]
  68. Lee, S.M.; Kim, N.; Yoon, H.; Kim, Y.S.; Choi, S.I.; Park, J.H.; Lee, D.H. Compositional and Functional Changes in the Gut Microbiota in Irritable Bowel Syndrome Patients. Gut Liver 2021, 15, 253. [Google Scholar] [CrossRef]
  69. Ahluwalia, B.; Iribarren, C.; Magnusson, M.K.; Sundin, J.; Clevers, E.; Savolainen, O.; Ross, A.B.; Törnblom, H.; Simrén, M.; Öhman, L. A Distinct Faecal Microbiota and Metabolite Profile Linked to Bowel Habits in Patients with Irritable Bowel Syndrome. Cells 2021, 10, 1459. [Google Scholar] [CrossRef] [PubMed]
  70. Dlugosz, A.; Winckler, B.; Lundin, E.; Zakikhany, K.; Sandström, G.; Ye, W.; Engstrand, L.; Lindberg, G. No Difference in Small Bowel Microbiota between Patients with Irritable Bowel Syndrome and Healthy Controls. Sci. Rep. 2014, 5, 8508. [Google Scholar] [CrossRef] [PubMed]
  71. Andrews, C.N.; Sidani, S.; Marshall, J.K. Clinical Management of the Microbiome in Irritable Bowel Syndrome. J. Can. Assoc. Gastroenterol. 2021, 4, 36–43. [Google Scholar] [CrossRef] [Green Version]
  72. Rodiño-Janeiro, B.K.; Vicario, M.; Alonso-Cotoner, C.; Pascua-García, R.; Santos, J. A Review of Microbiota and Irritable Bowel Syndrome: Future in Therapies. Adv. Ther. 2018, 35, 289–310. [Google Scholar] [CrossRef] [Green Version]
  73. Asha, M.Z.; Khalil, S.F.H. Efficacy and Safety of Probiotics, Prebiotics and Synbiotics in the Treatment of Irritable Bowel Syndrome a Systematic Review and Meta-Analysis. Sultan Qaboos Univ. Med. J. 2020, 20, e13. [Google Scholar] [CrossRef] [Green Version]
  74. Accarie, A.; Vanuytsel, T. Animal Models for Functional Gastrointestinal Disorders. Front. Psychiatry 2020, 11, 509681. [Google Scholar] [CrossRef]
  75. Wang, Y.; Bi, Z.; Wang, E.; Sun, B.; Zheng, Y.; Zhong, L.; Yuan, J. Rodent Model of Irritable Bowel Syndrome. Int. J. Gastroenterol. Disord. Ther. 2017, 4, 131. [Google Scholar] [CrossRef]
  76. Fijan, S. Microorganisms with Claimed Probiotic Properties: An Overview of Recent Literature. Int. J. Environ. Res. Public Health 2014, 11, 4745–4767. [Google Scholar] [CrossRef]
  77. McFarland, L.V.; Evans, C.T.; Goldstein, J.C. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review and Meta-Analysis. Front. Med. 2018, 5, 124. [Google Scholar] [CrossRef] [PubMed]
  78. Boyle, R.J.; Robins-Browne, R.M.; Tang, M.L. Probiotic use in clinical practice: What are the risks? Am. J. Clin. Nutr. 2006, 83, 1256–1264. [Google Scholar] [CrossRef]
  79. Sen, S.; Mullan, M.M.; Parker, T.J.; Woolner, J.T.; Tarry, S.A.; Hunter, J.O. Effect of Lactobacillus plantarum 299V on Colonic Fermentation and Symptoms of Irritable Bowel Syndrome. Dig. Dis. Sci. 2002, 47, 2615–2620. [Google Scholar] [CrossRef]
  80. Ducrotté, P.; Sawant, P.; Jayanthi, V. Clinical Trial: Lactobacillus plantarum 299V (DSM 9843) Improves Symptoms of Irritable Bowel Syndrome. World J. Gastroenterol. 2012, 18, 4012. [Google Scholar] [CrossRef]
  81. Pedersen, S.M.M.; Nielsen, N.C.; Andersen, H.J.; Olsson, J.; Simrén, M.; Öhman, L.; Svensson, U.; Malmendal, A.; Berthgam, H.C. The Serum Metabolite Response to Diet Intervention with Probiotic Acidified Milk in Irritable Bowel Syndrome Patients Is Indistinguishable from That of Non-Probiotic Acidified Milk by 1H NMR-Based Metabonomic Analysis. Nutrients 2010, 2, 1141–1155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Simrén, M.; Öhman, L.; Olsson, J.; Svensson, U.; Ohlson, K.; Posserud, I.; Strid, H. Clinical Trial: The Effects of a Fermented Milk Containing Three Probiotic Bacteria in Patients with Irritable Bowel Syndrome—A Randomized, Double-Blind, Controlled Study. Aliment. Pharmacol. Ther. 2010, 31, 218–227. [Google Scholar] [CrossRef]
  83. Søndergaard, B.; Olsson, J.; Ohlson, K.; Svensson, U.; Bytzer, P.; Ekesbo, R. Effects of Probiotic Fermented Milk on Symptoms and Intestinal Flora in Patients with Irritable Bowel Syndrome: A Randomized, Placebo-Controlled Trial. Scand. J. Gastroenterol. 2011, 46, 663–672. [Google Scholar] [CrossRef] [PubMed]
  84. Amirimani, B.; Nikfam, S.; Albaji, M.; Vahedi, S.; Nasseri-Moghaddam, S.; Sharafkhah, M.; Ansari, R.; Vahedi, H. Probiotic vs. Placebo in Irritable Bowel Syndrome: A Randomized Controlled Trial. Middle East J. Dig. Dis. 2013, 5, 98. [Google Scholar] [CrossRef]
  85. Roberts, L.M.; McCahon, D.; Holder, R.; Wilson, S.; Hobbs, F.R. A Randomised Controlled Trial of a Probiotic “functional Food” in the Management of Irritable Bowel Syndrome. BMC Gastroenterol. 2013, 13, 45. [Google Scholar] [CrossRef] [Green Version]
  86. Lorenzo-Zúñiga, V.; Llop, E.; Suárez, C.; Álvarez, B.; Abreu, L.; Espadaler, J.; Serra, J. I.31, a New Combination of Probiotics, Improves Irritable Bowel Syndrome-Related Quality of Life. World J. Gastroenterol. 2014, 20, 8709–8716. [Google Scholar] [CrossRef]
  87. Cremon, C.; Guglielmetti, S.; Gargari, G.; Taverniti, V.; Castellazzi, A.M.; Valsecchi, C.; Tagliacarne, C.; Fiore, W.; Bellini, M.; Bertani, L.; et al. Effect of Lactobacillus paracasei CNCM I-1572 on Symptoms, Gut Microbiota, Short Chain Fatty Acids, and Immune Activation in Patients with Irritable Bowel Syndrome: A Pilot Randomized Clinical Trial. United Eur. Gastroenterol. J. 2018, 6, 604–613. [Google Scholar] [CrossRef] [Green Version]
  88. Ligaarden, S.C.; Axelsson, L.; Naterstad, K.; Lydersen, S.; Farup, P.G. A Candidate Probiotic with Unfavourable Effects in Subjects with Irritable Bowel Syndrome: A Randomised Controlled Trial. BMC Gastroenterol. 2010, 10, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Whorwell, P.J.; Altringer, L.; Morel, J.; Bond, Y.; Charbonneau, D.; O’Mahony, L.; Kiely, B.; Shanahan, F.; Quigley, E.M.M. Efficacy of an Encapsulated Probiotic Bifidobacterium infantis 35,624 in Women with Irritable Bowel Syndrome. Am. J. Gastroenterol. 2006, 101, 1581–1590. [Google Scholar] [CrossRef] [PubMed]
  90. Guglielmetti, S.; Mora, D.; Gschwender, M.; Popp, K. Randomised Clinical Trial: Bifidobacterium bifidum MIMBb75 Significantly Alleviates Irritable Bowel Syndrome and Improves Quality of Life—A Double-Blind, Placebo-Controlled Study. Aliment. Pharmacol. Ther. 2011, 33, 1123–1132. [Google Scholar] [CrossRef]
  91. Murakami, K.; Habukawa, C.; Nobuta, Y.; Moriguchi, N.; Takemura, T. The Effect of Lactobacillus brevis KB290 against Irritable Bowel Syndrome: A Placebo-Controlled Double-Blind Crossover Trial. BioPsychoSoc. Med. 2012, 6, 16. [Google Scholar] [CrossRef] [Green Version]
  92. Martoni, C.J.; Srivastava, S.; Leyer, G.J. Lactobacillus acidophilus DDS-1 and Bifidobacterium lactis UABla-12 Improve Abdominal Pain Severity and Symptomology in Irritable Bowel Syndrome: Randomized Controlled Trial. Nutrients 2020, 12, 363. [Google Scholar] [CrossRef] [Green Version]
  93. Caviglia, G.P.; Tucci, A.; Pellicano, R.; Fagoonee, S.; Rosso, C.; Abate, M.L.; Olivero, A.; Armandi, A.; Vanni, E.; Saracco, G.M.; et al. Clinical Response and Changes of Cytokines and Zonulin Levels in Patients with Diarrhoea-Predominant Irritable Bowel Syndrome Treated with Bifidobacterium longum ES1 for 8 or 12 Weeks: A Preliminary Report. J. Clin. Med. 2020, 9, 2353. [Google Scholar] [CrossRef]
  94. Zhou, C.; Fang, X.; Xu, J.; Gao, J.; Zhang, L.; Zhao, J.; Meng, Y.; Zhou, W.; Han, X.; Bai, Y.; et al. Bifidobacterium longum Alleviates Irritable Bowel Syndrome-Related Visceral Hypersensitivity and Microbiota Dysbiosis via Paneth Cell Regulation. Gut Microbes 2020, 12, 1782156. [Google Scholar] [CrossRef] [PubMed]
  95. Lewis, E.D.; Antony, J.M.; Crowley, D.C.; Piano, A.; Bhardwaj, R.; Tompkins, T.A.; Evans, M. Efficacy of Lactobacillus paracasei Ha-196 and Bifidobacterium lngum R0175 in Alleviating Symptoms of Irritable Bowel Syndrome (IBS): A Randomized, Placebo-Controlled Study. Nutrients 2020, 12, 1159. [Google Scholar] [CrossRef] [Green Version]
  96. Kim, J.Y.; Park, Y.J.; Lee, H.J.; Park, M.Y.; Kwon, O. Effect of Lactobacillus gasseri BNR17 on Irritable Bowel Syndrome: A Randomized, Double-Blind, Placebo-Controlled, Dose-Finding Trial. Food Sci. Biotechnol. 2018, 27, 853–857. [Google Scholar] [CrossRef] [PubMed]
  97. Shin, S.P.; Choi, Y.M.; Kim, W.H.; Hong, S.P.; Park, J.M.; Kim, J.; Kwon, O.; Lee, E.H.; Hahm, K.B. A Double Blind, Placebo-Controlled, Randomized Clinical Trial That Breast Milk Derived-Lactobacillus gasseri BNR17 Mitigated Diarrhea-Dominant Irritable Bowel Syndrome. J. Clin. Biochem. Nutr. 2018, 62, 179–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Compare, D.; Rocco, A.; Coccoli, P.; Angrisani, D.; Sgamato, C.; Iovine, B.; Salvatore, U.; Nardone, G. Lactobacillus casei DG and Its Postbiotic Reduce the Inflammatory Mucosal Response: An Ex-Vivo Organ Culture Model of Post-Infectious Irritable Bowel Syndrome. BMC Gastroenterol. 2017, 17, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Seong, G.; Lee, S.; Min, Y.W.; Jang, Y.S.; Kim, H.S.; Kim, E.J.; Park, S.Y.; Kim, C.H.; Chang, D.K. Effect of Heat-Killed Lactobacillus casei DKGF7 on a Rat Model of Irritable Bowel Syndrome. Nutrients 2021, 13, 568. [Google Scholar] [CrossRef]
  100. Dapoigny, M.; Piche, T.; Ducrotte, P.; Lunaud, B.; Cardot, J.M.; Bernalier-Donadille, A. Efficacy and Safety Profile of LCR35 Complete Freeze-Dried Culture in Irritable Bowel Syndrome: A Randomized, Double-Blind Study. World J. Gastroenterol. 2012, 18, 2067. [Google Scholar] [CrossRef]
  101. Urgesi, R.; Casale, C.; Pistelli, R.; Rapaccini, G.L.; de Vitis, I. A Randomized Double-Blind Placebo-Controlled Clinical Trial on Efficacy and Safety of Association of Simethicone and Bacillus coagulans (Colinox®) in Patients with Irritable Bowel Syndrome. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 1344–1353. [Google Scholar] [PubMed]
  102. Majeed, M.; Nagabhushanam, K.; Natarajan, S.; Sivakumar, A.; Ali, F.; Pande, A.; Majeed, S.; Karri, S.K. Bacillus coagulans MTCC 5856 Supplementation in the Management of Diarrhea Predominant Irritable Bowel Syndrome: A Double Blind Randomized Placebo Controlled Pilot Clinical Study. Nutr. J. 2016, 15, 21. [Google Scholar] [CrossRef] [Green Version]
  103. Madempudi, R.S.; Ahire, J.J.; Neelamraju, J.; Tripathi, A.; Nanal, S. Randomized Clinical Trial: The Effect of Probiotic Bacillus coagulans Unique IS2 vs. Placebo on the Symptoms Management of Irritable Bowel Syndrome in Adults. Sci. Rep. 2019, 9, 12210. [Google Scholar] [CrossRef] [PubMed]
  104. Gupta, A.K.; Maity, C. Efficacy and Safety of Bacillus coagulans LBSC in Irritable Bowel Syndrome. Medicine 2021, 100, e23641. [Google Scholar] [CrossRef]
  105. Sun, Y.Y.; Li, M.; Li, Y.Y.; Li, L.X.; Zhai, W.Z.; Wang, P.; Yang, X.X.; Gu, X.; Song, L.J.; Li, Z.; et al. The Effect of Clostridium butyricum on Symptoms and Fecal Microbiota in Diarrhea-Dominant Irritable Bowel Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Sci. Rep. 2018, 8, 2964. [Google Scholar] [CrossRef] [Green Version]
  106. Zhao, Q.; Yang, W.R.; Wang, X.H.; Li, G.Q.; Xu, L.Q.; Cui, X.; Liu, Y.; Zuo, X.L. Clostridium butyricum Alleviates Intestinal Low-Grade Inflamm TNBS-Induced Irritable Bowel Syndrome in Mice by Regulating Functional Status of Lamina Propria Dendritic Cells. World J. Gastroenterol. 2019, 25, 5469. [Google Scholar] [CrossRef]
  107. Kruis, W.; Chrubasik, S.; Boehm, S.; Stange, C.; Schulze, J. A Double-Blind Placebo-Controlled Trial to Study Therapeutic Effects of Probiotic Escherichia coli Nissle 1917 in Subgroups of Patients with Irritable Bowel Syndrome. Int. J. Colorectal Dis. 2012, 27, 467–474. [Google Scholar] [CrossRef] [Green Version]
  108. De Chambrun, G.P.; Neut, C.; Chau, A.; Cazaubiel, M.; Pelerin, F.; Justen, P.; Desreumaux, P. A Randomized Clinical Trial of Saccharomyces cerevisiae versus Placebo in the Irritable Bowel Syndrome. Dig. Liver Dis. 2015, 47, 119–124. [Google Scholar] [CrossRef] [Green Version]
  109. Spiller, R.; Pélerin, F.; Cayzeele Decherf, A.; Maudet, C.; Housez, B.; Cazaubiel, M.; Jüsten, P. Randomized Double Blind Placebo-Controlled Trial of Saccharomyces cerevisiae CNCM I-3856 in Irritable Bowel Syndrome: Improvement in Abdominal Pain and Bloating in Those with Predominant Constipation. United Eur. Gastroenterol. J. 2016, 4, 353–362. [Google Scholar] [CrossRef] [Green Version]
  110. Abbas, Z.; Yakoob, J.; Jafri, W.; Ahmad, Z.; Azam, Z.; Usman, M.W.; Shamim, S.; Islam, M. Cytokine and Clinical Response to Saccharomyces boulardii Therapy in Diarrhea-Dominant Irritable Bowel Syndrome: A Randomized Trial. Eur. J. Gastroenterol. Hepatol. 2014, 26, 630–639. [Google Scholar] [CrossRef]
  111. Hong, K.B.; Seo, H.; Lee, J.S.; Park, Y. Effects of Probiotic Supplementation on Post-Infectious Irritable Bowel Syndrome in Rodent Model. BMC Complement. Altern. Med. 2019, 19, 195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Leventogiannis, K.; Gkolfakis, P.; Spithakis, G.; Tsatali, A.; Pistiki, A.; Sioulas, A.; Giamarellos-Bourboulis, E.J.; Triantafyllou, K. Effect of a Preparation of Four Probiotics on Symptoms of Patients with Irritable Bowel Syndrome: Association with Intestinal Bacterial Overgrowth. Probiotics Antimicrob. Proteins 2019, 11, 627–634. [Google Scholar] [CrossRef] [Green Version]
  113. Barrett, J.S.; Canale, K.E.K.; Gearry, R.B.; Irving, P.M.; Gibson, P.R. Probiotic Effects on Intestinal Fermentation Patterns in Patients with Irritable Bowel Syndrome. World J. Gastroenterol. 2008, 14, 5020. [Google Scholar] [CrossRef] [PubMed]
  114. Lee, S.H.; Joo, N.S.; Kim, K.M.; Kim, K.N. The Therapeutic Effect of a Multistrain Probiotic on Diarrhea-Predominant Irritable Bowel Syndrome: A Pilot Study. Gastroenterol. Res. Pract. 2018, 2018, 8791916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Sinn, D.H.; Song, J.H.; Kim, H.J.; Lee, J.H.; Son, H.J.; Chang, D.K.; Kim, Y.H.; Kim, J.J.; Rhee, J.C.; Rhee, P.L. Therapeutic Effect of Lactobacillus acidophilus-SDC 2012, 2013 in Patients with Irritable Bowel Syndrome. Dig. Dis. Sci. 2008, 53, 2714–2718. [Google Scholar] [CrossRef]
  116. Hong, K.S.; Kang, H.W.; Im, J.P.; Ji, G.E.; Kim, S.G.; Jung, H.C. Effect of Probiotics on Symptoms in Korean Adults with Irritable Bowel Syndrome. Gut Liver 2009, 3, 101. [Google Scholar] [CrossRef]
  117. Michail, S.; Kenche, H. Gut Microbiota Is Not Modified by Randomized, Double-Blind, Placebo-Controlled Trial of VSL#3 in Diarrhea-Predominant Irritable Bowel Syndrome. Probiotics Antimicrob. Proteins 2011, 3, 1–7. [Google Scholar] [CrossRef] [Green Version]
  118. Cui, S.; Hu, Y. Multistrain Probiotic Preparation Significantly Reduces Symptoms of Irritable Bowel Syndrome in a Double-Blind Placebo-Controlled Study. Int. J. Clin. Exp. Med. 2012, 5, 238. [Google Scholar]
  119. Sisson, G.; Ayis, S.; Sherwood, R.A.; Bjarnason, I. Randomised Clinical Trial: A Liquid Multi-Strain Probiotic vs. Placebo in the Irritable Bowel Syndrome—A 12 Week Double-Blind Study. Aliment. Pharmacol. Ther. 2014, 40, 51–62. [Google Scholar] [CrossRef] [Green Version]
  120. Yoon, H.; Park, Y.S.; Lee, D.H.; Seo, J.G.; Shin, C.M.; Kim, N. Effect of Administering a Multi-Species Probitic Mixture on the Change in Facial Microbiota and Symptoms of Irritable Bowel Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Clin. Biochem. Nutr. 2015, 57, 129–134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Mezzasalma, V.; Manfrini, E.; Ferri, E.; Sandionigi, A.; la Ferla, B.; Schiano, I.; Michelotti, A.; Nobile, V.; Labra, M.; di Gennaro, P. A Randomized, Double-Blind, Placebo-Controlled Trial: The Efficacy of Multispecies Probiotic Supplementation in Alleviating Symptoms of Irritable Bowel Syndrome Associated with Constipation. BioMed Res. Int. 2016, 2016, 4740907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Hod, K.; Dekel, R.; Aviv Cohen, N.; Sperber, A.; Ron, Y.; Boaz, M.; Berliner, S.; Maharshak, N. The Effect of a Multispecies Probiotic on Microbiota Composition in a Clinical Trial of Patients with Diarrhea-Predominant Irritable Bowel Syndrome. Neurogastroenterol. Motil. 2018, 30, e13456. [Google Scholar] [CrossRef] [Green Version]
  123. Ishaque, S.M.; Khosruzzaman, S.M.; Ahmed, D.S.; Sah, M.P. A Randomized Placebo-Controlled Clinical Trial of a Multi-Strain Probiotic Formulation (Bio-Kult®) in the Management of Diarrhea-Predominant Irritable Bowel Syndrome. BMC Gastroenterol. 2018, 18, 71. [Google Scholar] [CrossRef] [Green Version]
  124. Oh, J.H.; Jang, Y.S.; Kang, D.; Chang, D.K.; Min, Y.W. Efficacy and Safety of New Lactobacilli Probiotics for Unconstipated Irritable Bowel Syndrome: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2019, 11, 2887. [Google Scholar] [CrossRef] [Green Version]
  125. Zhang, L.; Liu, Y.X.; Wang, Z.; Wang, X.Q.; Zhang, J.J.; Jiang, R.H.; Wang, X.Q.; Zhu, S.W.; Wang, K.; Liu, Z.J.; et al. Clinical Characteristic and Fecal Microbiota Responses to Probiotic or Antidepressant in Patients with Diarrhea-Predominant Irritable Bowel Syndrome with Depression Comorbidity: A Pilot Study. Chin. Med. J. 2019, 132, 346. [Google Scholar] [CrossRef]
  126. Skrzydło-Radomańska, B.; Prozorow-Król, B.; Cichoż-Lach, H.; Majsiak, E.; Bierła, J.B.; Kanarek, E.; Sowińska, A.; Cukrowska, B. The Effectiveness and Safety of Multi-Strain Probiotic Preparation in Patients with Diarrhea-Predominant Irritable Bowel Syndrome: A Randomized Controlled Study. Nutrients 2021, 13, 756. [Google Scholar] [CrossRef]
  127. Majeed, M.; Nagabhushanam, K.; Arumugam, S.; Majeed, S.; Ali, F. Bacillus coagulans MTCC 5856 for the Management of Major Depression with Irritable Bowel Syndrome: A Randomised, Double-Blind, Placebo Controlled, Multi-Centre, Pilot Clinical Study. Food Nutr. Res. 2018, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Olesen, M.; Gudmand-Høyer, E. Efficacy, Safety, and Tolerability of Fructooligosaccharides in the Treatment of Irritable Bowel Syndrome. Am. J. Clin. Nutr. 2000, 72, 1570–1575. [Google Scholar] [CrossRef] [Green Version]
  129. Azpiroz, F.; Dubray, C.; Bernalier-Donadille, A.; Cardot, J.M.; Accarino, A.; Serra, J.; Wagner, A.; Respondek, F.; Dapoigny, M. Effects of ScFOS on the Composition of Fecal Microbiota and Anxiety in Patients with Irritable Bowel Syndrome: A Randomized, Double Blind, Placebo Controlled Study. Neurogastroenterol. Motil. 2017, 29, e12911. [Google Scholar] [CrossRef] [Green Version]
  130. Silk, D.B.A.; Davis, A.; Vulevic, J.; Tzortzis, G.; Gibson, G.R. Clinical Trial: The Effects of a Trans-Galactooligosaccharide Prebiotic on Faecal Microbiota and Symptoms in Irritable Bowel Syndrome. Aliment. Pharmacol. Ther. 2009, 29, 508–518. [Google Scholar] [CrossRef]
  131. Chen, Q.; Ren, Y.; Lu, J.; Bartlett, M.; Chen, L.; Zhang, Y.; Guo, X.; Liu, C. A Novel Prebiotic Blend Product Prevents Irritable Bowel Syndrome in Mice by Improving Gut Microbiota and Modulating Immune Response. Nutrients 2017, 9, 1341. [Google Scholar] [CrossRef] [Green Version]
  132. Niv, E.; Halak, A.; Tiommny, E.; Yanai, H.; Strul, H.; Naftali, T.; Vaisman, N. Randomized Clinical Study: Partially Hydrolyzed Guar Gum (PHGG) versus Placebo in the Treatment of Patients with Irritable Bowel Syndrome. Nutr. Metab. 2016, 13, 10. [Google Scholar] [CrossRef] [Green Version]
  133. Swanson, K.S.; Gibson, G.R.; Hutkins, R.; Reimer, R.A.; Reid, G.; Verbeke, K.; Scott, K.P.; Holscher, H.D.; Azad, M.B.; Delzenne, N.M.; et al. The International Scientific Association for Probiotics and Prebiotics (ISAPP) Consensus Statement on the Definition and Scope of Synbiotics. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 687–701. [Google Scholar] [CrossRef]
  134. Cappello, C.; Tremolaterra, F.; Pascariello, A.; Ciacci, C.; Iovino, P. A Randomised Clinical Trial (RCT) of a Symbiotic Mixture in Patients with Irritable Bowel Syndrome (IBS): Effects on Symptoms, Colonic Transit and Quality of Life. Int. J. Colorectal Dis. 2013, 28, 349–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Shavakhi, A.; Shavakhi, S.; Minakari, M.; Farzamnia, S.; Peykar, M.; Taghipour, G.; Tayebi, A.; Hashemi, H. The Effects of Multi-Strain Probiotic Compound on Symptoms and Quality-of-Life in Patients with Irritable Bowel Syndrome: A Randomized Placebo-Controlled Trial. Adv. Biomed. Res. 2014, 3, 140. [Google Scholar] [CrossRef] [PubMed]
  136. Bogovič Matijašic, B.; Obermajer, T.; Lipoglavšek, L.; Sernel, T.; Locatelli, I.; Kos, M.; Šmid, A.; Rogelj, I. Effects of Synbiotic Fermented Milk Containing Lactobacillus acidophilus La-5 and Bifidobacterium animalis ssp. lactis BB-12 on the Fecal Microbiota of Adults with Irritable Bowel Syndrome: A Randomized Double-Blind, Placebo-Controlled Trial. J. Dairy Sci. 2016, 99, 5008–5021. [Google Scholar] [CrossRef] [PubMed]
  137. Bucci, C.; Tremolaterra, F.; Gallotta, S.; Fortunato, A.; Cappello, C.; Ciacci, C.; Iovino, P. A Pilot Study on the Effect of a Symbiotic Mixture in Irritable Bowel Syndrome: An Open-Label, Partially Controlled, 6-Month Extension of a Previously Published Trial. Tech. Coloproctol. 2014, 18, 345–353. [Google Scholar] [CrossRef]
  138. Asgarshirazi, M.; Shariat, M.; Dalili, H. Comparison of the Effects of Ph-Dependent Peppermint Oil and Synbiotic Lactol (Bacillus coagulans + Fructooligosaccharides) on Childhood Functional Abdominal Pain: A Randomized Placebo-Controlled Study. Iran. Red Crescent Med. J. 2015, 17, e23844. [Google Scholar] [CrossRef] [Green Version]
  139. Moser, A.M.; Spindelboeck, W.; Halwachs, B.; Strohmaier, H.; Kump, P.; Gorkiewicz, G.; Högenauer, C. Effects of an Oral Synbiotic on the Gastrointestinal Immune System and Microbiota in Patients with Diarrhea-Predominant Irritable Bowel Syndrome. Eur. J. Nutr. 2019, 58, 2767–2778. [Google Scholar] [CrossRef] [Green Version]
  140. Lee, S.H.; Cho, D.Y.; Lee, S.H.; Han, K.S.; Yang, S.W.; Kim, J.H.; Lee, S.H.; Kim, S.M.; Kim, K.N. A Randomized Clinical Trial of Synbiotics in Irritable Bowel Syndrome: Dose-Dependent Effects on Gastrointestinal Symptoms and Fatigue. Korean J. Fam. Med. 2019, 40, 2. [Google Scholar] [CrossRef]
  141. Min, Y.W.; Park, S.U.; Jang, Y.S.; Kim, Y.H.; Rhee, P.L.; Ko, S.H.; Joo, N.; Kim, S.I.; Kim, C.H.; Chang, D.K. Effect of Composite Yogurt Enriched with Acacia Fiber and Bifidobacterium lactis. World J. Gastroenterol. 2012, 18, 4563. [Google Scholar] [CrossRef]
  142. Bahrudin, M.F.; Abdul Rani, R.; Tamil, A.M.; Mokhtar, N.M.; Raja Ali, R.A. Effectiveness of Sterilized Symbiotic Drink Containing Lactobacillus helveticus Comparable to Probiotic Alone in Patients with Constipation-Predominant Irritable Bowel Syndrome. Dig. Dis. Sci. 2020, 65, 541–549. [Google Scholar] [CrossRef] [Green Version]
  143. Skrzydło-Radomańska, B.; Prozorow-Król, B.; Cichoż-Lach, H.; Majsiak, E.; Bierła, J.B.; Kosikowski, W.; Szczerbiński, M.; Gantzel, J.; Cukrowska, B. The Effectiveness of Synbiotic Preparation Containing Lactobacillus and Bifidobacterium Probiotic Strains and Short Chain Fructooligosaccharides in Patients with Diarrhea Predominant Irritable Bowel Syndrome—A Randomized Double-Blind, Placebo-Controlled Study. Nutrients 2020, 12, 1999. [Google Scholar] [CrossRef]
  144. Seong, G.; Lee, S.; Min, Y.W.; Jang, Y.S.; Park, S.Y.; Kim, C.H.; Lee, C.; Hong, S.N.; Chang, D.K. Effect of a Synbiotic Containing Lactobacillus paracasei and Opuntia Humifusa on a Murine Model of Irritable Bowel Syndrome. Nutrients 2020, 12, 3205. [Google Scholar] [CrossRef]
Table 1. Studies on the impact of probiotics on IBS patients.
Table 1. Studies on the impact of probiotics on IBS patients.
PaperResearch TypeParticipants Selection CriteriaParticipants 1IBS
Subtype
Preparation (Dosage)Placebo/Control (Dosage)DurationOutcomes
Sen et al. (2002) [79]The randomized, double-blind, placebo-controlled studyRome I12 males/females 18–65 y 2wd 3ProViva (oatmeal gruel, 5 × 107 cfu/mL 4 of Lb. plantarum 299V)
125 mL/d 5
Oatmeal gruel
125 mL/d
First 4 weeks of placebo administration, then 4 weeks of treatmentNo beneficial effect
Whorwell et al. (2006) [89]The randomized, double-blind, placebo-controlled, multicenter, dose-ranging studRome II362 females 18–65 yAllExcipient (no data on used compounds), B. infantis 35,624 (three different doses: 1.0 × 106, 1.0 × 108, or 1.0 × 1010 cells/capsules)
1 capsule/d 6
Excipient (no data on used compounds)
1 capsule/d
2 weeks of the run-in period, then 4 weeks of treatment, and 2 weeks of follow upImprovement of IBS symptoms
The effective dose was 1.0 × 108 cells/capsule
Barrett et al. (2008) [113]Uncontrolled pilot studyRome II18 males/females 20–70 yAllYakult® (sucrose, skim milk powder, dextrose, 6.5 × 109 cells/dose L. casei Shirota)
65 mL/d
na 7Up to 2-week run-in period, 6 weeks of treatmentReduction in SIBO 8
Regression of ERBHAL 9, accompanied by improved abdominal symptoms
Sinn et al. (2008) [115]The randomized, double-blind, placebo-controlled human studyRome III40 males/females 18–70 yAllExcipient (no data on used compounds), freeze-dried Lb. acidophilus SDC 2012, and Lb. acidophilus SDC 2013 (a total of 2.0 × 109 cfu/mL)
2 capsules/d
Excipient (no data on used compounds)
2 capsules/d
4 weeksImproved IBS symptoms
Hong et al. (2009) [116]The randomized, double-blinded, placebo-controlled parallel-group clinical studyRome III70 males/females 19–75 yAllB. bifidum BGN4, B. lactis AD011, Lb. acidophilus AD031, Lb. casei IBS041 (a total of 2.0 × 1010 cfu/packet 10 viable, lyophilized probiotics)
2 packets/d 11
Skim milk powder
2 packets/d
8 weeksReduced abdominal pain and defecation discomfort
Ligaarden et al. (2010) [88]The randomized double-blind, placebo-controlled, crossover trialRome II16 males/females 18–75 yAll1010 cfu/capsule 12 Lb. plantarum MF1298 (live, freeze-dried)
1 capsule/d
No data on used compounds
1 capsule/d
1 week run-in period, followed by 3 weeks of treatment (probiotc/placebo), then 4 weeks of wash-out phase, and another 3 weeks of alternate treatment (placebo/probiotic)Adverse effects of probiotic
Pedersen et al. (2010) [81]Placebo-controlled studyRome II61 males/females 18–79 yAllMilk
(a total of 107–109 cfu/g 13 acidifiers: Lb. delbruckeii ssps.
bulgaricus,
S. thermophilus; probiotics: 5 × 107 cfu/mL Lb. paracasei F19,
5 × 107 cfu/mL Lb acidophilus LA-5,
5 × 107 cfu/mL B. lactis BB-12)
400 mL/d
Milk
(acidifiers: D-(+)-gluconic acid, δ-lactone)
400 mL/d
2 weeks of wash-out period, then 8 weeks of treatmentObserved effects of treatment were due to acidified milk itself, not the probiotic
Simrén et al. (2010) [82]The randomized, double-blind, placebo-controlled studyRome II74 males/females 18–70 yAllCultura (fermented with: Lb. bulgaricus, S. thermophilus; containing probiotics: 5.0 × 107 cfu/mL Lb. paracasei F19, 5.0 × 107 cfu/mL Lb. acidophilus La5, 5.0 × 107 cfu/mL B. lactis Bb12)
400 mL/d
Acidified milk
400 mL/d
2 weeks of the run-in period, then 8 weeks of treatment, and 8 weeks of follow-upNo beneficial effect
Guglielmetti et al. (2011) [90]The prospective, multicenter, randomized, double-blind, placebo-controlled, two-arm nutritional studyRome III122 males/females 18–68 ywdExcipient (no data on used compounds), 1 × 109 cfu/capsule B. bifidum MIMBb75
1 capsule/d
Excipient (no data on used compounds), maltodextrin
1 capsule/d
2 weeks of the run-in period, then 4 weeks of treatment, and 2 weeks of wash-out phaseImprovement of IBS symptoms
Maintained beneficial impact of probiotic during the wash-out period
Michail and Kenche (2011) [117]The randomized, double-blind, placebo-controlled trialRome III24 males/females average age of 21.8 ± 17IBS-DVSL#3 (cornstarch, S. thermophilus, B. breve, B. longum, B. infantis, Lb acidophilus, Lb plantarum, Lb paracasei, Lb. bulgaricus)
9.0 × 1011 cells/d 14
Cornstarch8 weeksNo impact on fecal microbiota
No influence on BMI
Søndergaard et al. (2011) [83]The randomized, double-blind, placebo-controlled, parallel-group trialRome II64 males/females 18–70 ywdCultura
1000 mL/d
Acidified milk
1000 mL/d
2-week run-in period, then 8 weeks of treatmentNo effect on IBS symptoms
Cui and Hu (2012) [118]The double-blind, placebo-controlled studyRome III60 males/females average age between 44.38 ± 15.08 and 48.45 ± 14.08AllBifid triple viable capsule (B. longum, Lb. acidophilus)
6 capsules/d
No data on used compounds
600 mg/d 15
4 weeksImprovement in overall IBS symptoms
Higher abundance of gene copies of Bifidobacterium spp. and Lactobacillus spp. in feces
Dapoigny et al. (2012) [100]The prospective, multicenter, randomized, double-blind, placebo-controlled, parallel-group pilot trialRome III50 males/females 18–70 yAll2 × 108 cfu/capsule Lb. rhamnosus (total freeze-dried culture)
3 capsules/d
No data on used compounds
3 capsules/d
2 weeks of the run-in period, then 4 weeks of treatment, and 2-week follow-upImprovement of symptoms only among IBS-D patients
Ducrotté et al. (2012) [80]The multicenter, parallel-group, double-blind, placebo-controlled studyRome III216 males/females 18–70 yAllExcipients (potato starch, magnesium stearate) and 1 × 1010 cfu/capsule Lb. plantarum 299V DSM 9843 Potato starch and magnesium stearate4 weeks of treatment, then 3 weeks of follow-upPotentially beneficial in the management of IBS
Kruis et al. (2012) [107]The randomized, double-blind, parallel-group, monocenter studyRome II120 males/females 18–65IBS-C, IBS-DMUTAFLOR® (2.5–25 × 109 cfu/capsule Escherichia coli Nissle 1917)
1 capsule/d (first 4 days)
2 capsules/d (the rest of the trial)
No data on used compounds
1 capsule/d (first 4 days)
2 capsules/d (the rest of the trial)
12 weeksTherapeutic potential for PI-IBS 16 PA-IBS 17 subjects
Murakami et al. (2012) [91]The placebo-controlled, double-blind, crossover trialRome III35 males/females ≥ 6 y wdCorn starch, maltitol syrup, hydroxypropyl methylcellulose, calcium stearate, ≥1.0 × 1010 cfu/capsule Lb. brevis KB290
1 capsule/d
Corn starch, maltitol syrup, hydroxypropyl methylcellulose, calcium stearate1 capsule/d4-week run-in period, followed by 4 weeks of treatment (probiotic/placebo), then 4 weeks of wash-out phase, and another 4 weeks of alternate treatment (placebo/probiotic)Increased abundance of Bifidobacterium spp. and decrease of Clostridium spp. in fecal samples
Improvement of IBS symptoms and subjects’ quality of life
Amirimani et al. (2013) [84]The randomized parallel-group, single-blind, placebo-controlled studyRome III72 subjects (no information on sex, and age)wdBiogaia® (1 × 108 viable cells of Lb. reuteri)
1 tablet/d 18
1 tablet/d4 weeksIncreased frequency of defecation
No vital differences in IBS symptoms
Roberts et al. (2013) [85]The randomized, double-blind, placebo-controlled trialRome III179 males/females 18–65 ywdYogurt with 1.2 × 109 cfu/cup 19 of standard strains (S. thermophilus CNCM I-1630, Lb. bulgaricus CNCM I-1632, and I-1519) with the addition of 1.25 × 1010 cfu/cap B. lactis DN-173 010
2 cups/d 20
Milk-based non-fermented dairy product without probiotics and with similar lactose content to the test product2 cups/d12 weeksNo impact of tested probiotic on IBS symptoms severity
Abbas et al. (2014) [110]The randomized, double-blind, placebo-controlled trialRome III72 males/females 18–60 yIBS-DS. boulardii
750 mg/d
No data on used compounds
750 mg/d
2 weeks run-in period, then 6 weeks of treatmentDecreased blood levels of pro-inflammatory cytokines (IL 21-8, TNF-α 22)
Increased tissue levels of anti-inflammatory cytokines (IL-10)
Lorenzo-Zúñiga et al. (2014) [86]The multicenter, randomized, double-blind, placebo-controlled intervention clinical trialRome III84 males/females 20–70 yIBS-DLb. plantarum CECT7484, Lb. plantarum CECT7485, and P. acidilactici CECT7483 (ratio 1:1:1; a total of 1–3 × 1010 cfu/capsule in high dose or 3–6 × 109 cfu/capsule in low dose)
1 capsule/d
No data on used compounds
1 capsule/d
6 weeksImprovement of IBS-related life quality
No significant relief in the severity of IBS symptoms
Lack of significant improvement of IBS symptoms
Amelioration of quality of subjects’ life
Sisson et al. (2014) [119]The single-center, randomized, double-blind, placebo-controlled trialRome III186 males/females 18–65 yAllSymprove (water-based barley extract, Lb. rhamnosus NCIMB 30174, Lb. plantarum NCIMB 30173, Lb. acidophilus NCIMB 30175, E. faecium NCIMB 30176; total of 1.0 × 1010 bacteria/50 µ)
1 mL/kg a day 23
Water, flavorings
1 mL/kg a day
12 weeks of treatment, and 4 weeks of follow-upSignificantly improved symptoms of IBS, especially pain and bowel habits
Ameliorated symptoms severity
The effect was not sustained during the follow-up period
Urgesi et al. (2014) [101]The monocentric, double-blind, placebo-controlled, parallel-group clinical trialRome III52 males/females 18–75 ywdColinox® (excipient—no data on used compounds, simethicone, 1.5 × 109 spores/g 24 B. coagulans)
3 tablets/d
Excipient (no data on used compounds) 3 tablets/d4 weeksSignificant improvement of IBS symptom
de Chambrun et al. (2015) [108]The randomized, single-center, double-blind, placebo-controlled, parallel-group clinical studyRome III179 males/females 18–75 yAll8 × 109 cfu/g S. cerevisiae CNCM I-3856
1 capsule/d
Dibasic calcium phosphate
1 capsule/d
2 weeks of the run-in period, then 8 weeks of treatment, and 3 weeks of follow-upReduced severity of abdominal pain/discomfort
The relief of symptoms was limited to the duration of treatment
Yoon et al. (2015) [120] The randomized, double-blind, placebo-controlled trialRome III81 males/females 19–75 yAllLacClean Gold-S® (maltodextrin, corn starch, silicon dioxide, B. bifidum KCTC 12199BP, B. lactis KCTC11904BP, B. longum KCTC 12200BP, Lb. acidophilus KCTC 11906BP, Lb. rhamnosus KCTC 12202BP, S. thermophilus KCTC 11870BP; a total of 5 × 109 viable cells/capsule)
2 capsules/d
Maltodextrin, corn starch, silicon dioxide 2 capsules/d4 weeksIncreased abundance of administrated probiotic strains in fecal samples
Amelioration of diarrhea-predominant symptoms of IBS
Majeed et al. (2016) [102]The randomized, double-blind, parallel-group, placebo-controlled, multi-centered studyRome III36 males/females 18–55 yIBS-DMicrocrystalline cellulose, starch, sodium starch glycolate, magnesium stearate, and B. coagulans MTCC 5856
2 × 109 spores/tablet 25
1 tablet/d
Maltodextrin
1 tablet/d
90 daysAttenuation of symptoms (bloating, vomiting, stool frequency, abdominal pain, diarrhea)
Improved quality of life
Mezzasalma et al. (2016) [121]The randomized, double-blind, placebo-controlled studyRome III157 males/females 18–65 yIBS-CProduct 1: 5 × 109 cfu Lb. acidophilus DSM 24936, 5 × 109 cfu Lb. reuteri DSM 25175, inulin, silica, talc; product 2: 5 × 109 cfu Lb. rhamnosus DSM 25568, 5 × 109 cfu Lb. plantarum DSM 24937, 5 × 109 cfu B. lactis DSM 25566, inulin, silica, talcInulin, silica, talc60 days of treatment, then 30 days of follow-upIncreasing abundance of tested strains in fecal samples during treatment
Probiotic strains remained in the stool samples during follow-up period, except B. lactis
Both products diminish severity of IBS-C symptoms
Spiller et al. (2016) [109]The multi-center, randomized, double-blind, placebo-controlled trialRome III379 males/females 18–75 yAllS. cerevisiae I-3856
2 capsules/d
No data on used compounds
2 capsules/d
2-week run-in period, then 12 weeks of treatmentImproved GIT symptoms of IBS-C subjects
Compare et al. (2017) [98]Ex vivo studyRome III20 males/females 18–70 yPI-IBS/IBS-DLb. casei DG and its postbioticHealthy controls (10 out of 20)naDecreased inflammatory mucosal response in ex vivo model of PI-IBS-D subjects
Cremon et al. (2018) [87]The multicenter, randomized, double-blind, cross-over, placebo-controlled, pilot trialRome III criteria40 males/females 18–65 yAllGelatin capsule containing 2.4 × 1010 viable cells of Lb. paracasei CNCM I-1572
2 capsules/d
No data on used compounds
2 capsules/d
2 weeks of the run-in period, next 4 weeks of treatment (probiotic/placebo), followed by 4 weeks of wash-out phase, and another 4 weeks of alternate treatment (placebo/probiotic), then 4-week follow-upNo significant improvement of IBS symptoms compared to placebo
Reduced Ruminococcus spp.
Diminished levels of pro-inflammatory cytokines IL-6, and IL-15
Increased levels of butyrate
Hod et al. (2018) [122]The randomized, double-blind, placebo-controlled, parallel-group clinical trialRome III97 females 18–70IBS-DBIO-25 (3.0 × 109 cfu/capsule Lb. rhamnosus LR5; 2.0 × 109 cfu/capsule Lb. casei LC5; 1.0 × 109 cfu/capsule Lb paracasei LPC5; 1.0 × 109 cfu/capsule Lb. plantarum LP3; 5.0 × 109 cfu/capsule Lb. acidophilus LA1; 4.0 × 109 cfu/capsule B. bifidum BF3; 1.0 × 109 cfu/capsule B. longum BG7; 2.0 × 109 cfu/capsule B. breve BR3; 1.0 × 109 cfu/capsule B infantis BT1; 2.0 × 109 cfu/capsule S. thermophilus ST3; 3.0 × 109 cfu/capsule Lb. bulgaricus LG1, 3.0 × 109 cfu/capsule L. lactis SL6)
2 capsules/d
Cellulose2 capsules/d2 weeks of run-in period, then 8 weeks of treatmentNo effect on fecal microbiota diversity
Increased abundance of Lactobacillus spp. and Lactococcus spp. in stool samples of subjects whose abdominal pain and bloating were reduced
Individuals with decreased abdominal pain and improved stool consistency showed a decrease in Bilophila genus prevalence
Ishaque et al. (2018) [123]The randomized, double-blind, placebo-controlled, equal allocation ratio, parallel-group, clinical trialRome III360 males/females 18–55 yIBS-DBio-Kult® (B. subtilis PXN 21, B. bifidum PXN 23, B. breve PXN 25, B. infantis PXN 27, B. longum PXN 30, Lb. acidophilus PXN 35, Lb. delbrueckii spp. bulgaricus PXN39, Lb. casei PXN 37, Lb. plantarum PXN 47, Lb. rhamnosus PXN 54, Lb.helveticus PXN 45, Lb. salivarius PXN 57, L. lactis PXN 63, S. thermophilus PXN 66; a total of 2.0 × 109 cfu/capsule)
4 capsules/d
Microcrystalline cellulose, hydroxypropyl methylcellulose4 capsules/d16 weeksSignificantly improved IBS symptoms and their severity
Lee et al. (2018) [114]The single-arm, open-label, pilot studyRome III11 males 19–70 yIBS-DTher-Biotic® Complete (Lb. rhamnosus 6.0 × 109 cfu/capsule, B. bifidum 5.0 × 109 cfu/capsule, Lb. acidophilus 3.0 × 109 cfu/capsule, Lb. casei 2.5 × 109 cfu/capsule, Lb. plantarum 2.0 × 109 cfu/capsule, Lb. salivarius 2.0 × 109 cfu/capsule, B. longum 1.0 × 109 cfu/capsule, S. thermophilus 1.0 × 109 cfu/capsule, Lb. bulgaricus 1.0 × 109 cfu/capsule, Lb. paracasei 5.0 × 108 cfu/capsule, B. lactis 5.0 × 108 cfu/capsule, B. breve 5.0 × 108 cfu/capsule)No placebo or control group8 weeksAmeliorated abdominal discomfort, dyspepsia, flatulence, and stool consistency
Decreased SIBO prevalence
Beneficial impact on GIT microbiota
Kim et al. (2018) [96]The double-blind, randomized, placebo-controlled, parallel studyindividual protocol42 males/females 20–54 ywdLb. gasseri BNR17
Low dose: 2 × 108 cfu/d 26
Medium dose: 2 × 109 cfu/d
High dose: 2 × 5 × 109 cfu/d
Dextrin7 days of the run-in period, then 4 weeksAmelioration of IBS symptoms
The abundance of probiotic strain in fecal samples of treated subjects
The optimal dosage appeared to be high dose (2 × 5 × 109 cfu/d)
Majeed et al. (2018) [127]The randomized, double-blind, placebo-controlled, multicenter, parallel-group clinical studyRome III40 males/females 20–65 ywdMicrocrystalline cellulose, starch, sodium starch glycolate, magnesium stearate, 2.0 × 109 spores/tablet B. coagulans MTCC 5856
1 tablet/d
Microcrystalline cellulose, starch, sodium starch glycolate, magnesium stearate1 tablet/d90 days of treatment, then 15 days of follow-upDecrease in depression, along with diminished IBS symptoms
Reduced sleeplessness
Decreased serum levels of myeloperoxidase
Shin et al. (2018) [97]The single-center, randomized, double-blind, placebo-controlled clinical trialRome III51 males/females 20–55 yIBS-DMaltodextrin, microcrystalline cellulose, magnesium stearate, Lb. gasseri BNR17 (total of 1010 cfu/d)
4 capsules/d
Maltodextrin, microcrystalline cellulose, magnesium stearate4 capsules/d8 weeksImproved bowel habits (longer colon transit time)
Positive impact on intestinal microbiota (decreased Firmicutes, increased Actinobacteria, and Bacteroidetes)
Sun et al. (2018) [105]The prospective, multicenter, randomized, double-blind, placebo-controlled trialRome III criteria200 males/females 18–65 yIBS-D1.5 × 107 cfu/capsule C. butyricum
9 capsules/d
No data on used compounds
9 capsules/d
4 weeksImprovement in overall IBS symptoms and quality of patients’ life
No significant changes in intestinal microbiota diversity
Reduction of Clostridium sensu stricto in microbial community
Improvement of alanine and tryptophan metabolism
Hong et al. (2019) [111]Animal studyna 54 males C57L/B6 mice (15.5 ± 1.0 g) aged 4 weeksna
PI-IBS model (Trichinella)
Formulated probiotics (DW; Lb. acidophilus LA5, B. lactis BB12, S. boulardii)
0.2 mL of the solution of 5.0 × 109 cfu/g
VSL#3
0.2 mL of the solution of 5.0 × 109 cfu/g
No data on used compounds
0.2 mL of the solution of 5.0 × 109 cfu/g
4 weeksDecreased levels of serum pro-inflammatory cytokines in uninfected and infected mice treated with probiotics (DW and VSL#3)
Diminished visceral hypersensitivity (DW)
Leventogiannis et al. (2019) [112]Open-label clinical studyRome III/SIBO-positive or SIBO-negativeMales/Females ≥ 18 yAllLactolevure® (1.5 × 109 cfu/capule S. boulardii, 1.75 × 109 cfu/capsule B. lactis BB-12, 1.5 × 109 cfu/capsule Lb. acidophilus LA-5, 5.0 × 108 cfu/capsule Lb. plantarum)
2 capsules/d
No placebo/control group30 daysAmelioration of bloating
More significant improvement of symptoms among IBS patients with SIBO
Decreased abdominal pain severity
Madempudi et al. (2019) [103]The randomized, double-blind, placebo-controlled trialRome III108 males/females 18–60 ywdExcipient (maltodextrin), 2.0 × 109 cfu/capsule B. coagulans Unique IS2Excipient (maltodextrin)8 weeksRelief in the severity of symptoms (bloating, incomplete evacuation, urgency, straining, the passage of gas, bowel habit satisfaction, and stool consistency)
Reduced abdominal pain
Increased number of complete spontaneous bowel movement
Oh et al. (2019) [124]The randomized, double-blind, placebo-controlled trialRome III50 males/females 19–60 yWith the exclusion of IBS-CFoodis Lactobacillus—excipients (olive oil, pine oil), and Lb. paracasei, Lb. salivarius, 1 × 109 cfu/mL Lb. plantarum (ratio 5:4:1)Excipients (olive oil, pine oil)1 week screening period, then 4 weeks of treatmentRelief in global IBS symptoms
Decreased severity of abdominal pain
Zhang et al. (2019) [125]Pilot studyRome III15 males/females 18–65 yIBS-DBifico® (B. longum, Lb. acidophilus, E. faecalis; a total of ≥1.0 × 107 cfu)20 mg 3 times a dayAntidepressant (Duloxetine)
30 mg/d for 4 days, then 60 mg/d
8 weeksChanges in gut microbiota and SCFAs 27 concentrations (probiotic and antidepressant)
Reduced severity of abdominal symptoms (probiotic and antidepressant)
Decreased plasma levels of cytokines (probiotic and antidepressant)
Zhao et al. (2019) [106]Animal studiesna24 C57BL/6 male mice aged 6–8 weeksna
PI-IBS model
(TNBS 28)
1 × 108 cfu/mL C. butyricum
200 µL/d
Saline4 weeks of model preparation, one week of treatmentAttenuated intestinal visceral hypersensitivity
Diminished low-grade mucosal inflammation (suppressed production of cytokines, decreased number of lamina propria dendritic cells)
Caviglia et al. (2020) [93]The prospective studyRome IV16 males/females 16–65 yIBS-DB. longum ES1 1 × 109 cfuLack of control8–12 weeksImprovement in general IBS symptoms
Amelioration of the immune-inflammatory condition (reduced cytokines, and zonulin levels)
Increased integrity of the intestinal barrier
Lewis et al. (2020) [95]The randomized, double-blind, placebo-controlled, 3-arm parallel-group studyRome III285 males/females ≥ 18 yAllExcipients (potato starch, and magnesium stearate), and 10 × 109 cfu/capsule of B. longum HA-196, or Lb. paracasei R0175Potato starch, and magnesium stearate2-week run-in period, next 8 weeks of treatmentSignificantly improved bowel habits, and stool consistency in IBS-D, and IBS-C subjects (Lb. paracasei R0175)
The positive impact of social aspects of life (both probiotic strains)
Increased number of Bifidobacterium ssp. in fecal samples (B. langum HA-196)No vital changes in A. muciniphila, and F. prausnitzii abundance
Distinct placebo effect
Martoni et al. (2020) [92]The randomized, double-blind, placebo-controlled, multicenter studyRome IV336 males/females 18–70 ywdMicrocrystalline cellulose, and ≥1 × 1010 cfu/capsule of Lb. acidophilus DDS®-1, or B. animalis subsp. lactis UABla-12™
1 capsule/d
Microcrystalline cellulose
1 capsule/d
2-week run-in period (only placebo), 6 weeks of treatment (probiotic/placebo)Improvement in stool consistency
Reduced abdominal pain severity
Vital amelioration of IBS symptoms
Diminished stress levels (Lb. acidophilus DDS®-1)
Zhou et al. (2020) [94]Animal studynaMale Sprague-Dawley rats (weight: 225~260 g)na
WAS model
B. longum 1 × 109 cfu/mL once a day0.9% saline10 daysInfluenced Paneth cells function—enhanced lysozyme production, and repair of mucus
No difference in serum cytokines levels
Beneficial alteration of GIT microbiota
Improved intestinal permeability
Gupta et al. (2021) [104]The prospective, interventional, randomized, double-blind, placebo-controlled clinical studyRome IV40 males/females 18–65 ywdExcipient with B. coagulans LBSC 2 × 109 spores/sachet
3 sachets/d 29
Excipient with maltodextrin
3 sachets/d
Up to 80 daysImprovement in abdominal symptoms (pain, stomach rumbling)
Attenuation of bloating, cramping, nausea, vomiting, diarrhea, anxiety
Beneficial modulation of GIT microbiota
Amelioration in stool consistency
Seong et al. (2021) [99]Animal studyna14 male Wistar rats (weight: 304 ± 1.4 g) aged 8 weeksna
IBS induced by chronic restraint stress
Maltodextrin and heat-killed 1 × 1011 cfu Lb. casei DKGF7Maltodextrin4 weeksImprovement of IBS symptoms in the animal model
Decrease in serum corticosterone levels, inflammatory cytokines in colonic tissue
Enhanced expression of tight junction proteins
Skrzydło-Radomańska et al. (2021) [126]The randomized, double-blind, placebo-controlled, parallel-group trialRome III51 males/females 8–75 yIBS-DNordBiotic™ (B. breve BB010
B. longum BL020,
B. bifidum BF030,
B. lactis BL040,
Lb. rhamnosus LR110,
Lb paracasei LPC100,
Lb acidophilus LA120,
Lb. casei LC130,
Lb plantarum LP140,
S. thermophilus ST250; Total 2.5 × 109 cfu/capsule)
2 capsules/d
Maltodextrin
2 capsules/d
8 weeksImproved quality of patients’ life
Diminished severity of abdominal pain
1 the final (after screening) number of participants who received treatment is given; 2 years old; 3 without differentiation; 4 colony-forming units per milliliter, 5 milliliters per day; 6 capsules per day; 7 not applicable; 8 small intestinal bacterial overgrowth; 9 early rise in breath hydrogen after lactulose; 10 colony-forming units per packet; 11 packets per day; 12 colony-forming unit per capsule; 13 colony-forming units per gram; 14 cells per day; 15 milligrams per day; 16 post-infection IBS; 17 post-antibiotic IBS; 18 tablets per day; 19 colony-forming units per cup; 20 cups per day; 21 interleukins; 22 tumor necrosis factor α; 23 milliliters per kilogram; 24 spores per gram; 25 spores per tablet; 26 colony-forming units per day; 27 short-chain fatty acids; 28 2,4,6-trinitrobenzenesulfonic acid; 29 sachets per day.
Table 2. Studies on the effect of prebiotics on IBS individuals.
Table 2. Studies on the effect of prebiotics on IBS individuals.
PaperResearch TypeParticipants Selection CriteriaParticipants 1IBS SubtypePreparation (Dosage)Placebo/Control (Dosage)DurationOutcomes
Hunter et al. (1999) [52]The randomized, controlled, double-blind, crossover studyIndividual protocol21 males/females 18–65 y 2wd 3Oligofructose (Raftilose P95) three times 2 g/d 4 Sucrose three times 1 g/dFirst 2 weeks of normal diet, then 2 weeks of controlled standard UK diet (45% carbohydrate, 40% fat, and 15% protein)No significant results
Olesen et al. (2000) [128]The prospective, randomized, placebo-controlled, single-blind, and double-blind phasesManning98 males/females 18–70 ywdFOS 5 10 g/d (2 weeks)/20 g/d (next 10 weeks)Glucose 10 g (2 weeks)/20 g/d (next 10 weeks)First 2 weeks of single-blind phase (only placebo), then 12 weeks of double-blind phase (prebiotic/placebo)FOS may increase the severity of IBS symptoms; however, patients might adapt after prolonged usage
Silk et al. (2009) [130]The single-center, parallel, patient blinded, randomized, crossover, placebo-controlled trialRome II44 males/females 20–79 yAlltrans-GOS 6
3.5 or 7.0 g/d
Maltodextrins
DE 20
3.5 or 7.0 g/d
Baseline period—2 weeks (only placebo); next phase—3 months (prebiotic/placebo)Increased number of Bifidobacterium spp. to a level comparable with healthy people (both doses of GOS)
Increased number of Eubacterium rectale/C.coccoides (GOS dose of 3.5 g/d)
Reduced number of C. perfringens (GOS dose of 7.0 g)
Changes in the consistency of feces, flatulence, and overall improvement of IBS symptoms (GOS)
The reduced anxiety level in the IBS-D group (GOS dose of 7.0 g/d)
Azpiroz et al. (2016) [129]The parallel, placebo-controlled, randomized, double-blind studyRome III79 males/females 18–60 yAllscFOS two times 2.5 g/dMaltodextrins two times 2.5 g/d28 daysIncreased number of Bifidobacterium spp.
Decreased level of anxiety
Attenuated severity of IBS symptoms
No effect on rectal hypersensitivity
Niv et al. (2016) [132]The prospective, randomized, double-blind, placebo-controlled studyRome III108 males/females 18–77 yAllPHGG 3 g/d (first week)/6 g/d (11 weeks) Maltodextrin 3 g/d (first week)/6 g/d (11 weeks)First 2 weeks without prebiotic/placebo, next 12 weeks of product administration, then 4 weeks of follow-upImprovement on bloating and gasses
Chen et al. (2017) [131]Animal studyna 760 four-week-old female specific pathogen-free (SPF) C57BL/6 micena
PI-IBS 8 model
The PB (FOS, GOS, inulin, and anthocyanins)
1.26 mg/g body weight
Saline/healthy control group8 weeks of preventive administration of the PB/saline before infection with Trichinella spiralis larvae, then 8 weeks for recovery without prebioticFaster recovery from body weight loss
Pretreatment with PB could help to improve the well-being of PI-IBS patients
PB can diminish inflammation both in the Caco-2 cells and the IBS mice model
Protection of intestinal barrier integrity
PB could help protect the homeostasis of GIT microbiota
1 the final (after screening) number of participants who received treatment is given; 2 years old; 3 without differentiation; 4 gram per day; 5 fructooligosaccharides; 6 galactooligosaccharides; 7 not applicable; 8 post-infection IBS.
Table 3. Researches on the influence of synbiotics on IBS patients.
Table 3. Researches on the influence of synbiotics on IBS patients.
PaperResearch TypeParticipants Selection CriteriaParticipants 1IBS SubtypePreparation
(Dosage)
Placebo/Control (Dosage)DurationOutcomes
Min et al. (2012) [141]The randomized, double-blind, controlled trialRome III117 males/females 18–70 y 2AllYogurt with standard strains S. thermophilus (≥3 × 109 cfu 3/bottle) and Lb. acidophilus (≥109 cfu/bottle) with the addition of Bifidobacterium animalis subsp. lactis Bb-12 (≥1011 cfu/bottle), Bifidobacterium enhancer, and acacia dietary fiberYogurt with standard strains S. thermophilus (≥3 × 109 cfu/bottle) and Lb. acidophilus (≥109 cfu/bottle) with the addition of Bifidobacterium animalis subsp. lactis Bb-12 (≥1011 cfu/bottle)8 weeksImprovement in bowel habits and IBS symptoms
Cappello et al. (2013) [134]The parallel-group, double-blinded, placebo-controlled studyRome III64 males/females 18–75 ywd 4Probinul ® (5 × 109 Lb. plantarum, 2 × 109 Lb. casei subp. rhamnosus, 2 × 109 Lb. gasseri, 1 × 109 B. infantis, 1 × 109 B. longum, 1 × 109 Lb. acidophilus, 1 × 109 Lb. salivarus, 1 × 109 Lb. sporogenes, 5 × 109 S. thermophilus, 2 g inulin, 1.3 g tapioca-resistant starch) twice 5 g/d 5No data on used compounds
product by CaDi Group (Rome, Italy)
2 weeks prior to synbiotic/placebo administration, then 4 weeks of treatmentDecreased flatulenceThe increased transition time of intestinal content
Improved in self-scored quality of IBS patients’ life
No overall relief of symptoms
Bucci et al. (2014) [137]The parallel-group, double-blinded, randomized, placebo-controlled study (core study) and the open-label prospective, partially controlled Rome III64 males/females 18–75 ywdProbinul®
twice 5 g/d (during extension period only 2 weeks/month)
Lack of information4 weeks of core study, then 6 months of the extension periodDecreased flatulence even during cyclic administration
Rogha et al. (2014) [54]The randomized, double-blinded, placebo-controlled trialRome III56 males/females
The average age of 39.8 ± 12.7
wdLactol® (1.5 × 108 spores of B. coagulans and FOS)
100 mg
Lactose starch and tartazine
100 mg
12 weeksRelief from abdominal pain/discomfort and diarrhea
Shavakhi et al. (2014) [135] The randomized, placebo-controlled, triple-blinded studyRome II129 males/females average age of 36.2 ± 9.3 wdBalance® (FOS, magnesium stearate, hydroxypropyl methyl cellulose, Lb. casei, Lb. rhamnosus, Lb. acidophilus, Lb. bulgaricus, B. breve, B. longum, S. thermophilus; a total of 1 × 108 cfu/capsule)
2 capsules a day
No data on used compounds
2 capsules a day
14 daysNo effect of treatment
Bogovič Matijašic et al. (2016) [136]The double-blind, randomized, placebo-controlled multicenter trialRome III30 subjects 18–65 yIBS-CFermented milk (starter culture: S. thermophilus ABT-21, probiotics: 1.8 × 107 cfu/g Lb acidophilus La-5, 2.5 × 107 cfu/g B. lactis BB-12, 2% dietary fiber Beneo Orafti Synergy1—90% inulin, 10% oligofructose)
360 g/d
Heat-treated fermented milk without probiotic bacteria and dietary fibers
360 g/d
2 weeks run-in period, then 4 weeks of treatment, and 2 weeks of follow-upIncreased abundance of used probiotic strains in subjects’ fecal samples
Transient colonization of used probiotics
The abundance of the Enterobacteriaceae family was not affected
No significant changes in fecal microbiota
Moser et al. (2019) [139]Pilot studyIndividual protocol10 males/females 37–53 yIBS-DOMNi-BiOTiC® Stress Repair (corn starch, maltodextrin, inulin, FOS, potassium chloride, magnesium sulfate, mangan sulfate, enzymes, 7.5 × 109 of each strain: Lb. casei W56, Lb. acidophilus W22, Lb. paracasei W20, Lb. salivarius W24, Lb. plantarum W62, L. lactis W19, B. lactis W51, B. lactis W52, B. bifidum W23)na 64 weeksIncreased phylogenetic diversity of gastric and duodenal microbiota
Reduced number of CD4+ T cells in the ascending colon
Higher levels of acetate and butyrate in fecal samples
Lee et al. (2019) [140]The single-center, randomized, double-blind, placebo-controlled clinical trialRome III28 males/females
≥19 y
AllUltra-Probiotics-500
(1 × 1013 cfu of probiotic strains: Lactobacillus (rhamnosus, acidophilus, casei, bulgaricus, plantarum, and salivarius), Bifidobacterium (bifidum and longum, 175 mg of FOS, 150 mg of Ulmus davidiana, 10 mg of Geum urbanum, and 100 mg of inulin)
1 capsule/d (low-dose group)2 capsules/d (high-dose group)
The same material used for encapsulation. No data on used compounds.
1 capsule/d (low-dose group)
2 capsules/d (placebo group)
8 weeksNo dose-dependent effects
Decrease the fatigue in IBS individuals (high-dose)
Relief in abdominal pain/discomfort, bloating, stool patterns
Bahrudin et al. (2020) [142]The prospective, double-blind, randomized, controlled trialRome III163 males/females
22–37 y
IBS-CSynbiotic drink (water, sugar, skimmed milk powder (cow), stabilizers (polydextrose), fermented milk (water, acidity regulator, skimmed milk powder (cow), and lactobacillus), acidity regulators, soybean fiber, and flavoring) with Lb. helveticus and 1.5 g/100 mL polydextrose
350 mL/d 5
Probiotic drink ((water, sugar, skimmed milk powder (cow), stabilizers (polydextrose), fermented milk (water, acidity regulator, skimmed milk powder (cow), and lactobacillus), acidity regulators, soybean fiber, and flavoring)) with Lb. helveticus
350 mL/d
1 weekShortened intestinal transition time
Reduced fecal pH
Relief in constipation-related symptoms
No difference between synbiotic and control group—probiotic alone conferred a health benefit
Seong et al. (2020) [144]Animal studyna20 male Wistar rats (weight: 350 ± 50 g) aged 8 weeksnaTreatment group 1—maltodextrin, 1 × 1010 cfu/g Lb. paracasei DKGF; Treatment group 2—maltodextrin, 1 × 1010 cfu/g Lb. paracasei DKGF, 10.0 mg (w/w) Opuntia extract; Treatment group 3—maltodextrin, 1 × 1010 cfu/g Lb. paracasei DKGF, 30.0 mg (w/w) Opuntia extractMaltodextrin4 weeksImproved stool consistency (better in synbiotic groups)
Decreased serum corticosterone levels (lower in synbiotic groups)
Low levels of TNF-α 7 in the colonic mucosa (both synbiotic and probiotic groups) Increased expression of the tight junction proteins (higher in synbiotic groups)
Higher abundance of Lb. paracasei in fecal samples (more significant difference in synbiotic groups)
Skrzydło-Radomańska et al. (2021) [143]The randomized, double-blind, placebo-controlled, parallel group trial Rome III68 males/females 18–60 yIBS-DSynbiotic containing a total of 5.0 × 109 probiotic strains (B. lactis DSMZ 32269,
B. longum DSMZ 32946,
B. bifidum DSMZ 32403,
Lb. rhamnosus FloraActive 19070-2,
Lb. acidophilus DSMZ 32418) and 947 mg of scFOS
2 sachet/d
978 mg of maltodextrin2 sachet/d2 weeks screening period, then 8 weeks of treatmentAttenuation of IBS symptoms (pain, flatulence, stool pressure, feeling of incomplete bowel movements)
Decreased severity of IBS symptoms
1 the final (after screening) number of participants who received treatment is given; 2 years old; 3 colony-forming units; 4 without differentiation; 5 not applicable; 6 milliliters per day; 7 tumor necrosis factor α.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chlebicz-Wójcik, A.; Śliżewska, K. Probiotics, Prebiotics, and Synbiotics in the Irritable Bowel Syndrome Treatment: A Review. Biomolecules 2021, 11, 1154. https://doi.org/10.3390/biom11081154

AMA Style

Chlebicz-Wójcik A, Śliżewska K. Probiotics, Prebiotics, and Synbiotics in the Irritable Bowel Syndrome Treatment: A Review. Biomolecules. 2021; 11(8):1154. https://doi.org/10.3390/biom11081154

Chicago/Turabian Style

Chlebicz-Wójcik, Agnieszka, and Katarzyna Śliżewska. 2021. "Probiotics, Prebiotics, and Synbiotics in the Irritable Bowel Syndrome Treatment: A Review" Biomolecules 11, no. 8: 1154. https://doi.org/10.3390/biom11081154

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

Article Metrics

Back to TopTop