Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Efficacy and safety of a food supplement with standardized menthol, limonene, and gingerol content in patients with irritable bowel syndrome: A double-blind, randomized, placebo-controlled trial

  • Vladimir T. Ivashkin,

    Roles Conceptualization, Writing – review & editing

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Anna V. Kudryavtseva,

    Roles Methodology, Resources, Software, Supervision

    Affiliation Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation

  • George S. Krasnov,

    Roles Data curation, Methodology, Resources, Software

    Affiliation Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation

  • Yuri M. Poluektov ,

    Roles Data curation, Supervision, Writing – review & editing

    yuripoul@gmail.com

    Affiliation Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation

  • Margarita A. Morozova,

    Roles Data curation, Formal analysis

    Affiliation Mental Health Research Center, Moscow, Russian Federation

  • Oleg S. Shifrin,

    Roles Data curation, Formal analysis, Resources

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Allan G. Beniashvili,

    Roles Conceptualization, Data curation, Investigation, Methodology

    Affiliation Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russian Federation

  • Zarina A. Mamieva,

    Roles Investigation, Validation, Writing – original draft

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Alexandra L. Kovaleva,

    Roles Investigation, Resources, Writing – original draft

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Anatoly I. Ulyanin,

    Roles Data curation, Formal analysis, Methodology

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Elizaveta A. Trush,

    Roles Investigation, Resources

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

  • Alexander G. Erlykin,

    Roles Formal analysis, Methodology, Resources

    Affiliation Moscow State Linguistic University, Moscow, Russian Federation

  • Elena A. Poluektova

    Roles Conceptualization, Data curation, Project administration, Supervision, Writing – review & editing

    Affiliation I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation

Abstract

Background

Irritable bowel syndrome (IBS) affects 9,2% of the global population and places a considerable burden on healthcare systems. Most medications for treating IBS, including spasmolytics, laxatives, and antidiarrheals, have low efficacy. Effective and safe therapeutic treatments have yet to be developed for IBS.

Purpose

This study assessed the efficacy and safety of a food supplement containing standardized menthol, limonene, and gingerol in human participants with IBS or IBS/functional dyspepsia (FD).

Design

A double-blind, randomized, placebo-controlled trial.

Methods

We randomly assigned 56 patients with IBS or IBS/FD to an intervention group (Group 1) or control group (Group 2) that were given supplement or placebo, respectively, in addition to the standard treatment regimen for 30 d. Three outpatient visits were conducted during the study. Symptom severity was measured at each visit using a 7×7 questionnaire. Qualitative and quantitative composition of the intestinal microbiota were assessed at visits 1 and 3 based on 16S rRNA gene sequencing.

Results

At visit 1 (before treatment), the median total 7×7 questionnaire score was in the moderately ill range for both groups, with no difference between the groups (p = 0.1). At visit 2, the total 7×7 score decreased to mildly ill, with no difference between the groups (p = 0.4). At visit 3, the total score for group 1 indicated borderline illness and for group 2 remained indicated mild illness (p = 0.009). Even though we observed some variations in gut microbiota between the groups, we did not find any statistically significant changes.

Conclusion

The food supplement with standardized menthol, limonene, and gingerol content increased the efficacy of standard therapy in IBS and FD patients. The use of the supplement did not cause any obvious side effects.

Registration

ClinicalTrials.gov Identifier: NCT04484467

Introduction

Irritable bowel syndrome (IBS) is the most prevalent functional disorder worldwide and 27.0%–82.6% of patients with IBS exhibit symptoms of functional dyspepsia (FD) [1, 2]. These diseases impair the patient’s quality of life, reduce their capacity for social activity, and create significant healthcare expenses [3]. Based on diagnosis by Rome III criteria, the prevalence of IBS is estimated at 10,7% in Bangladesh and 9,2% worldwide [4].

Most medications for IBS, including spasmolytics, laxatives, and antidiarrheals, have relatively low efficacy and optimal treatment approaches for IBS and FD have yet to be developed [5, 6]. Tricyclic antidepressants (TCAs) and selective serotonin reuptake inhibitors have shown superior efficacy in the treatment of IBS and FD in placebo control studies, though adverse effects have a high prevalence, particularly among patients treated with TCAs [7, 8].

Recently, a relationship was established between gut microbiota and a functional gastrointestinal tract disorder [9]. The composition of the intestinal microbiota in patients suffering from IBS is characterized by decreased content of bacterial cells producing short-chain fatty acids (SCFAs) [10], which regulates the expression of tight contact proteins, T-lymphocytes, and cytokines and ensures adequate permeability of the intestinal barrier [11]. Disruption of intestinal barrier permeability leads to inflammatory responses, changes in sensitivity and motility, and the development of disease symptoms in the intestinal wall [1214]. Therefore, the inclusion of probiotics in the treatment regimen promotes overall therapeutic efficacy [15, 16].

IBS pathogenesis represents a complex interplay among changes in intestinal permeability and microbial dysbiosis within the gut, altered mucosal immune function, visceral hypersensitivity, impaired gut motility [17]. Given the comprehensively described pathology of IBS, we aimed to analyze the efficacy of treatment with a food supplement and how its components affect each pathogenetic stage as well as the composition of intestinal microbiota.

Some herbal ingredients have been reported to relieve IBS symptoms. Menthol has an antispasmodic, analgesic effect and suppresses the growth of pathogenic microorganisms; D-limonene participates in esophagus and stomach mucus barrier recovery; and gingerol has an antispasmodic and prokinetic effect (Table 1). Thus, the proposed treatment consists of these components and should cover almost all known pathophysiological links of IBS disorder.

thumbnail
Table 1. Components of the supplement and their mechanisms of action.

https://doi.org/10.1371/journal.pone.0263880.t001

We designed a double-blind, randomized, placebo-controlled trial to assess the efficacy and safety of a food supplement standardized for menthol, limonene, and gingerol content (hereinafter referred to as “the supplement”) in IBS and IBS/FD patients.

Methods

Trial design

We designed an interventional (clinical trial), randomized, double-blind controlled trial protocol that was approved by the Ethics Committee of the Mental Health Research Center, Moscow, Russian Federation (no. 418, 01/31/2018). Written informed consent was obtained from all participants.

Participants

Participants with IBS or IBS and FD were recruited during their outpatient medical treatment at the Mental Health Research Center and Sechenov University, Moscow, Russia. Symptoms were validated according to the Rome IV criteria. Non-functional causes for the symptoms were excluded after a detailed evaluation of medical history, physical examination, extensive panel of blood tests, stool analysis, and colonoscopy with biopsies [28, 29]. The exclusion criteria were: patients younger than 18 years, older than 59 years, with organic bowel disease, with renal disease, with hepatic insufficiency, or with mental illness significantly impairing self-report (e.g., schizophrenia, bipolar disorder, or epilepsy) (Fig 1).

Interventions

Standard treatment regimen.

Diarrhea-predominant IBS patients (IBS-D) and mixed bowel habits IBS patients (IBS-M) were treated with spasmolytics. Constipation-predominant IBS patients (IBS-C) were treated with spasmolytics and laxatives. IBS/FD patients were treated with spasmolytics and proton pump inhibitors.

Additional treatment regimen.

At visit 1, all patients were randomly assigned into two groups and were assigned a 30-day standard treatment regimen and 1 capsule, 730 mg, once a day in a double-blind manner. The intervention group (group 1) received the supplement “Standart Zdorovya GASTRO” and the control group (group 2) received a placebo (Table 2). During the study period, three outpatient visits were conducted (visit 1 at day 1, visit 2 at 15 ± 2 d, and visit 3 at 30 ± 2 d).

thumbnail
Table 2. Components in the supplement and placebo capsules.

https://doi.org/10.1371/journal.pone.0263880.t002

Outcomes

Primary outcome measures.

Changes in symptom severity of IBS and FD (constipation-predominant, diarrhea-predominant, mixed-type IBS and IBS/FD) were assessed at each visit using a 7×7 questionnaire.

Secondary outcome measures.

Changes in the number of SCFA producing bacteria as well as the qualitative and quantitative composition of intestinal microbiota were assessed at visits 1 and 3, based on 16S rRNA gene sequencing data.

Sample size

Due to a lack of comparable studies, we were unable to determine an appropriate sample size. Using GPower 3.1 software, a sample size of 38 patients was calculated according to Jacob Cohen’s effect size estimation method: statistical significance α = 0.05, power (1-β) = 0.80, and expected effect size d = 0.70 (moderate to strong) [30]. To increase the statistical power of the study, a total of 56 patients were recruited.

Randomization

According to symptom severity, the patients were assigned to one of six groups, determined by the sum of the 7×7 scores: 0–1, normal (healthy); 2–6, borderline ill; 7–12, mildly ill; 13–18, moderately ill; 19–24, markedly ill; >25, severely ill. Effective therapy of IBS greatly decreased the sum of the 7×7 scores (by more than 6 points) in previous studies [31] and effect size (Cohen`s d) can reach 2. The effect size values normally range from 0.3 (low effectiveness of treatment) to 0.5 (moderate effectiveness) and up to 0.8 (high effectiveness). We assumed that the use of food supplements would show a high effect size and reduced the overall 7×7 scores. Thus, clinically significant decreases in the total 7×7 score (5.4 points on average) should have an effect size of 0.07.

Blinding

Being a double-blind trial, both participants and investigators were blinded to the treatment. Placebo and food supplement “Standart Zdorovya GASTRO” were delivered to the center in non-transparent packages. The containers were numbered by the manufacturer and neither the researchers nor the patients knew what number belonged to placebo or food supplement “Standart Zdorovya GASTRO”.

Assessment tools

At each visit, patients completed the 7×7 questionnaire [32] as an assessment of symptom severity. At visits 1 and 3, the qualitative and quantitative composition of intestinal microbiota were determined based on 16S rRNA gene sequencing.

7×7 Questionnaire

The 7×7 questionnaire allows patients to record the main symptoms of FD and IBS and the physician to obtain a quantitative description of symptom frequency and intensity for objective evaluation. The following points are assigned according to symptom frequency: none, 0 points; once a week, 1 point; two to three times a week, 2 points; daily, 3 points; and several times a day, 4 points. The intensity is estimated as mild (1 point), moderate (2 points), and severe (3 points). Solid/“nuts” stool once a week and two to three times a week are scored at 5 points and 2 points, respectively. Liquid or mushy stool with other symptoms (pain in the stomach area, a feeling of burning in the stomach area, fullness in the stomach, early satiety, abdominal pain that decreases after a bowel movement, or bloating) is assigned 0 to 4 points. The scores reflecting symptom presence and intensity are summarized, and the scores obtained for each symptom are summed. According to the total score, patients would be assigned to one of six groups: 0–1 points, healthy; 2–6 points, borderline ill; 7–12 points, mildly ill; 13–18 points, moderately ill; 19–24 points, markedly ill; and 25 or more points, severely ill [19].

Safety

The safety of treatment was assessed at Visits 1, 2 and 3 by interviewing the patients about the additional complaints. Blood samples were collected for the routine blood tests (hemoglobin level, red blood cell count, white blood cells count, erythrocyte sedimentation rate, aspartate aminotransferase level, alanine aminotransferase level, alkaline phosphatase level, gamma-glutamyltransferase level, creatinine level, pancreatic alpha-amylase level) at visit 1 and 3.

DNA isolation, 16S library preparation, and qualitative and quantitative composition of intestinal microbiota based on 16S rRNA gene sequencing

Total DNA was isolated with an AmpliPrimeDNA-sorb-AM kit (NextBio, Moscow, Russia) for clinical specimens, according to the manufacturer’s protocol, and stored at –20°C. For qualitative and quantitative assessment of the isolated DNA, we used a NanoDrop 1000 spectrophotometer (Thermo Fisher Scientific, Waltham, MA, USA). Next, a 16S library was prepared according to the Illumina MiSeq protocol for 16S Metagenomic Sequencing Library Preparation (Illumina, San Diego, CA, USA). The first round of 16S rDNA amplification of the V3–V4 variable regions was performed using the following primers: forward 5′-TCGTCGGCAGCGTCAGATGTGTATAAGAGACAG-CCTACGGGNGGCWGCAG-3′ and reverse 5′-GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAG-GACTACHVGGGTATCTAATCC-3′. These primers are aimed at the amplification of bacterial (more than 90% taxonomic coverage) but not archaeal (less than 5%) rRNA genes. The amplification cycle (2720 Thermal Cycler, Applied Biosystems, Waltham, MA, USA) was programmed as: 1) 95°C for 3 min; 2) 30 cycles of 95°C for 30 s, 55°C for 30 s, and 72°C for 30 s; 3) 72°C for 5 min; and 4) 4°C.

The derived amplicons were purified using Agencourt AMPure XP beads (Beckman Coulter, Brea, CA, USA) according to the manufacturer’s protocol. The second amplification round was used to double-index the samples with a combination of specific primers. The amplification program was set to: 1) 95°C for 3 min; 2) 8 cycles of 95°C for 30 s, 55°C for 30 s, and 72°C for 30 s; 3) 72°C for 5 min; and 4) 4°C.

The products of the second PCR round were purified using Agencourt AMPure XP beads. The concentrations of the 16S rDNA libraries were measured with a Qubit 2.0 fluorimeter (Invitrogen, Waltham, MA, USA) utilizing a Quant-iT dsDNA High-Sensitivity Assay Kit. The purified amplicons were mixed equimolarly according to the concentration values. The quality of the libraries was evaluated using an Agilent 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA) and an Agilent DNA 1000 Kit. Sequencing was carried out on a MiSeq system (Illumina) with a MiSeq Reagent Kit v2 (paired-end reads, 2 × 250 nt, Illumina).

The analysis of 16S rRNA gene sequencing data

Since the reads overlap was small (the average amplicon length was about 440–460 bp at sequencing 2 × 250 nt), the reads were pre-merged (before analysis, in DADA2) using the MeFiT tool [33]. For most samples, more than 99.5% of the reads were successfully merged and analyzed by the DADA2 package for R [34]. For the analysis: 1) primer sequences were removed using cutadapt; 2) the reads were filtered by quality; 3) error distribution models were derived based on read quality profiles; 4) sequencing errors were corrected; 5) ribosomal sequence variants (RSV), OTU analogues, were inferred; and 6) chimeric RSVs, accounting for 62% and 4.6% of all RSV and all reads, respectively, were eliminated. Next, taxonomic annotation of the RSVs (7,551 after the removal of chimeras) was carried out using DADA and the 16S RDP reference sequence database [35]. The data obtained were also analyzed utilizing the Piphillin web service [36], which predicts the metabolic potential of a bacterial community based on the analysis of representative 16S rRNA gene sequences (OTU or RSV). Further analysis was carried out using our own R (including the vegan, fossil, ggplot2, and pheatmap packages) and Python scripts. A Mann-Whitney test was used for comparison between groups, a Wilcoxon test for comparison between visits to the same patient, and a Student’s t-test for comparison of normally distributed data. Spearman correlation coefficients were calculated.

Statistical analysis

Statistical analysis was carried out using standard functions and additional packages in the R environment [37, 38]. To assess differences between groups of patients, analysis of contingency tables (including Fisher’s exact test) and a non-parametric Mann-Whitney test were used. The differences between visits in each group of patients was determined using a Friedman Rank test and a post-hoc Wilcoxon signed-rank test for paired samples, with p-values adjusted according to Bonferroni multiple testing (FDR). The null hypothesis assumed the absence of a relationship between the type of therapy/stage of the study and the change in the analyzed parameter. Two-sided p-values <0.05 were considered statistically significant.

Deviations from the original protocol

There were no deviation from the original protocol.

Study

As this was originally a pilot study, we assumed that the data would not be sufficient for a full-scale clinical trial or publication. Thus, the study was registered only after the initiation of patient recruitment. The authors can confirm that all ongoing and related trials have been registered.

Results

The trial was performed from February 2018 to December 2019 at the Mental Health Research Center, Moscow, Russia. Two groups of 56 patients collectively completed the study (Table 3). There were no significant differences in gender, age, diagnosis, disease duration, and 7×7 questionnaire results between the groups at the first visit.

thumbnail
Table 3. Comparison of patients in groups 1 and 2 by sex, age, and disease duration.

https://doi.org/10.1371/journal.pone.0263880.t003

7×7 Questionnaire

There was no statistically significant difference in the total 7×7 questionnaire scores between the groups at visit 1 and patients in both groups were moderately ill (see Table 2). Throughout all three visits, the treatment effect on the total 7×7 questionnaire scores was tested using the Friedman test (Table 1 in S4 File) followed by the Wilcoxon signed-rank test (Table 2 in S4 File). There was a statistically significant decrease in the total 7×7 questionnaire score in both groups to scores indicating mild illness at visit 2 (mean difference, –4.04; 95% CI, –6.10 to –1.97; p = 0.001 in group 1 and mean difference, –4.86; 95% CI, –6.75 to –2.96; p = 0.0004 in group 2). There were no significant differences (p = 0.42) between groups at visit 2. At visit 3, the total score in group 1 decreased to a score indicating borderline illness (mean difference, –2.68; 95% CI, –4.29 to –1.07; p = 0.009). Group 2 remained in the score range indicating mild illness (Fig 2).

thumbnail
Fig 2. Total 7×7 questionnaire scores in group 1 and group 2 at visits 1, 2, and 3.

https://doi.org/10.1371/journal.pone.0263880.g002

The total 7×7 questionnaire score decreased significantly in group 1 from 13 (95% CI 11.4–15.0) at visit 1 to 9.18 (95% CI 7.07–11.3) at visit 2 and 6.5 (95% CI 4.58–8.42) at visit 3. In group 2, the 7×7 questionnaire score decreased significantly between visits 1 and 2 (15.9 [95% CI 13.4–18.5] to 11.1 [95% CI 8.4–13.7]). * The differences between the groups are significant (p < 0.05); ns–non-significant.

Between visit 1 to visit 3, there was a decrease in the number of group 1 patients with epigastric pain (p = 0.029), solid/”nuts” stool (p = 0.005), abdominal pain that decreases after a bowel movement (p < 0.0001), and bloating (p = 0.015) (S1–S6 Figs in S3 File).

Qualitative and quantitative composition of the intestinal microbiota based on 16S rRNA gene sequencing.

At visit 1, we observed some differences in the gut microbiota composition between groups 1 and 2. For several genera, we noticed 1.5–2.5-fold change in the average bacterial content (normalized read counts per taxon) between the groups, some of which passed the Mann-Whitney U-test p < 0.05 threshold, but none with statistically significant FDR (e.g., Blautia, Anaerostipes, and Paraprevotella). The 60 bacterial genera with the highest abundance in patients of groups 1 and 2 at visit 1 are presented in Fig 3. A high degree of heterogeneity was observed between patients.

thumbnail
Fig 3. Qualitative and quantitative composition of the intestinal microbiota genera in patients of groups 1 and 2 at visit 1.

Columns 2 and 3 present the relative bacterial content of a given genus (r.c., represents the normalized number of reads annotated to the current genus) averaged over all samples of a group. The color brightness reflects the average content, whereas the color hue (blue to orange) indicates the difference between the groups. The heatmaps in columns 4 and 5 show the bacterial content profiles of the genera for individual samples. The genera are sorted by a scoring factor, which reflects the magnitude of differences between groups 1 and 2, statistical significance (based on the Mann-Whitney test), and the average bacterial content across all samples.

https://doi.org/10.1371/journal.pone.0263880.g003

Using the Piphillin web service, we inferred the relative abundance of genes involved in fatty acid biosynthesis (FAB) at visit 1. While the total abundance of all genes involved in FAB was practically identical (differing by less than 1%), significant differences were noted for the fabH gene (Fig 4), though it did not pass the FDR threshold.

thumbnail
Fig 4. Relative content in the metagenome of genes involved in fatty acid biosynthesis (FAB) in patients of groups 1 and 2 at visit 1 (inferred from taxonomic data).

Columns 5 and 6 show the average content of FAB genes in each group. The heatmaps in columns 3 and 4 demonstrate the relative gene abundance per sample. Samples are re-arranged according to the similarity of gene abundance profiles.

https://doi.org/10.1371/journal.pone.0263880.g004

At visit 3, we observed some differences in the composition of gut microbiota between groups 1 and 2. Oscillibacter had higher prevalence in group 1, whereas Veillonella, Collinsella, and Gemmiger were more prevalent in group 2 (Fig 5). However, the overall magnitude of these differences was comparable to that at visit 1. Although p-values (Mann-Whitney U-test) for these events were lower than 0.05, they did not pass the FDR < 0.05 threshold.

thumbnail
Fig 5. Statistically significant differences in the composition of intestinal microbiota genera between groups 1 and 2 at visit 3.

Columns 2 and 3 present the relative content (r.c.) of bacterial genera averaged over all samples in each group. The heatmaps in columns 4 and 5 show the bacterial content profiles for individual samples.

https://doi.org/10.1371/journal.pone.0263880.g005

The same observations were made for the relative content of genes involved in FAB (Fig 6). In this case, very slight (~2%) differences were noted for individual genes (fabZ) in favor of the patients taking dietary supplements. Even though they passed the Mann-Whitney U-test p < 0.05 threshold, they did not pass multiple test adjustments (FDR). The magnitude of inter-group differences at visit 3 was comparable to that at visit 1.

thumbnail
Fig 6. The relative content in the metagenome of genes participating in FAB in patients of groups 1 and 2 at visit 3 (inferred from taxonomic data).

Columns 5 and 6 show the average content of FAB genes in each group. The heatmaps in columns 3 and 4 demonstrate the relative gene abundance per sample. The samples are re-arranged according to the similarity of gene abundance profiles.

https://doi.org/10.1371/journal.pone.0263880.g006

Correlation analysis between microbiome composition and 7×7 questionnaire scores.

In order to identify the genera and families of bacteria associated with IBS severity, we carried out a correlation analysis between the relative abundance of various bacterial taxa and the sum of the 7×7 questionnaire scores across all visits. Statistically significant associations (that passed the FDR threshold < 0.05) were noticed only for the genus Fusobacterium and its related family Fusobacteriaceae (r = 0.29, p = 0.0002, FDR = 0.02). However, this family had a relatively low overall abundance (mean of 3 reads per 1 sample, found only in 11% of samples).

Additionally, we identified a number of families statistically associated with IBS severity, though they did not pass the FDR adjustment threshold. Among them were Acidaminococcaceae (Spearman’s r = –0.23, p = 0.005); Enterobacteriaceae (r = –0.16, p = 0.05); Streptococcaceae, Coriobacteriaceae, and Veillonellaceae (r = 0.18 … 0.21, p = 0.01 … 0.03). We noted positive correlations between Butyrivibrio, Catenibacterium, Akkermansia, Alloprevotella, Dialister, and Prevotella abundance with age (r = 0.17 … 0.24); inverse correlations of Roseburia, Megasphaera, Bacteroides, Odoribacter, Parabacteroides, Alistipes, and Parasutterella abundance with age (r = –0.17 … –0.28); and associations of Roseburia, Mitsuokella, Coprobacter, and Escherichia/Shigella abundance with gender.

Finally, we assessed the relationship between the metabolic potential of the collective microbiome (inferred from taxonomy) and disease severity (inferred from the 7×7 questionnaire score). Correlations were observed for 360 predicted bacterial genes, though none of them passed the FDR threshold. Interestingly, none of these genes were related to FAB. However, the 26th place (out of 4,405 genes) was marked by the atoE gene encoding for a short-chain fatty acid transporter. The correlation coefficient of its relative abundance and 7×7 score was r = –0.23 (p = 0.004). When analyzing associations with age, we noted 1,088 or 186 predicted bacterial genes at a given threshold for p < 0.05 or FDR < 0.05.

Correlation analysis between microbiome composition and 7×7 questionnaire scores.

The safety of the food supplement “Standart Zdorovya GASTRO” was measured according to additional complaints recorded during each visit. Also, blood tests were performed at visit 1 and 3, with no deviations from the reference values observed. In group 2 some changes were observed in sedimentation rate and red and white blood cell count, but these values did not exceed the reference ranges (S7–S16 Figs in S3 File). No additional complaints were recorded in either of the groups.

Discussion

The development and maintenance of functional gastrointestinal disorders can be attributed to various psychological and biological factors. Changes in the composition of intestinal microbiota and intestinal permeability that decrease tight junction protein expression and reduce the thickness of the mucus layer are key factors impairing gastrointestinal motility and sensitivity [3941].

Microbial SCFAs are vital to intestinal homeostasis. They suppress the growth of Gram-negative pathogens, function as energy sources for colonocytes, and have anti-inflammatory effects. SCFAs are organic fatty acids with 1–6 carbon atoms and are the principal anions arising from the bacterial fermentation of polysaccharides, oligosaccharides, proteins, peptides, and glycoprotein precursors in the colon [42]. The main SCFAs are produced by bacterial cells using acetyl-CoA by different pathways, such as acrylate and succinate [43].

We used the food supplement “Standart Zdorovya GASTRO” with standardized menthol, limonene, and gingerol content to deliver antispasmodic [18, 25], analgesic [19, 20], anti-inflammatory, and prokinetic [19, 20, 22, 2527] effects. The supplement has also been shown to contribute to the restoration of intestinal microbiota [21] and gastrointestinal mucosal function [23, 24] (Table 1). Thus, the inclusion of this supplement in IBS or IBS/FD treatment regimens can improve the efficacy of standard therapy. Patients who received the supplement showed significant symptom reduction. A significant decrease in symptom severity to borderline ill was observed in group 1 at visit 3 (7×7 score –6.5, 95% CI 4.58–8.42), whereas group 2 remained mildly ill (total 7×7 score 10.8, 95% CI 8.00–13.6). Furthermore, the number of IBS and FD symptoms decreased in more patients of group 1 than group 2.

The greater treatment efficacy in group 1 (standard treatment regimen and dietary supplement) might be associated with modifications in the intestinal microbiome and metabolome. However, we generally observed heterogeneous taxonomic profiles of microbiome community structure between patients, both at visits 1 and 3. This complicates the interpretation of changes caused by the intake of the dietary supplement. Similarly, we found a very small increase in the content of specific genes involved in the biosynthesis of fatty acids at visit 3. However, here it is also impossible to confirm that the changes were due to the effects of the dietary supplement. Nevertheless, the analysis of clinical indicators (7×7 questionnaire) showed an improvement in the condition of patients who had taken dietary supplements together with conventional therapy.

With regard to microbiome composition, there was no variation observed between groups 1 and 2. Even though several bacterial genera passed the p < 0.05 threshold, the number of these genera was comparable between visit 1 and 3. However, among these genera, we noticed Oscillibacter was dominant in group 1 and Veillonella, Gemmiger, and Collinsella were dominant in Group 2. Previously, an animal study showed higher Oscillibacter prevalence in healthy controls than in patients with IBS [44, 45] and high Veillonella prevalence in IBS-C patients [46, 47]. Although Oscillibacter can promote fiber degradation by an array of cellulases and produce butyric acid from glucose, ribose, and xylose, the main metabolite produced is valeric acid [48, 49]. Previously, it was shown that the abundance of Collinsella was associated with low a fiber diet and obesity [50], type 2 diabetes [51], and atherosclerosis [52]. Lower content of the Collinsella family (Coriobacteriaceae) was associated with a healthier microbiome composition [53]. No data exists for Gemmiger prevalence in patients with functional disorders or healthy controls.

We noticed slight changes between group 1 and 2 in the number of bacteria containing genes involved in the synthesis of fatty acids. However, these changes were not statistically significant after correcting for FDR and no correlations were observed with symptom severity. In general, an increased number of bacteria implicated in fatty acid biosynthesis could improve intestinal permeability through mucus layer generation [54].

When analyzing associations with the severity of IBS, we found positive correlations with the abundance of Fusobacterium. Moreover, the presence/abundance of these bacteria was strongly associated with increased discomfort and pain. Previous reports found that Fusobacterium increased severity of diarrhea-predominant IBS (IBS-D) [55], and Fusobacterium nucleatum was involved in the pathogenesis of IBS by causing microbial dysbiosis and exacerbating visceral hypersensitivity [55]. The metabolites of Fusobacterium contain butyric acid, which promotes visceral hypersensitivity in an IBS-like model via an enteric glial cell-derived nerve growth factor [56]. However, some studies have shown that Fusobacterium was decreased in IBS-D patients [57].

The present study revealed negative correlations between Acidaminococcaceae and Enterobacteriaceae abundance and positive correlations of Streptococcaceae, Coriobacteriaceae, and Veillonellaceae abundance with IBS severity. Except for Enterobacteriaceae, all of these families have been associated with symptom severity in IBS patients (especially IBS-D) compared to healthy controls or patients with other gastrointestinal diseases [5862].

The current study has several limitations. First, the sample size was relatively small and comprised only 56 patients. Further studies with more participants are needed to validate the trends observed. Moreover, we did not perform follow-up assessments to record the long-term treatment effects. This limitation will also be addressed in future trials.

Conclusion

The components of the food supplement “Standart Zdorovya GASTRO” standardized for menthol, limonene, and gingerol influence the main pathogenetic mechanisms of IBS and FD and increase the efficacy of standard therapy for IBS and FD. The use of the supplement is safe and does not cause side effects.

Acknowledgments

We would like to thank “РМЛ ИНВЕСТ” (RML INVEST), Torkhovsky passage 10, Tula, Russian Federation, for providing the food supplement “Стандарт Здоровья ГАСТРО” (Standart Zdorovya GASTRO); registration number: RU.77.99.88.003.E.004127.09.17; standardized for menthol, limonene, and gingerol for scientific purposes.

References

  1. 1. Perveen I, Rahman MM, Saha M, Rahman MM, Hasan MQ. Prevalence of irritable bowel syndrome and functional dyspepsia, overlapping symptoms, and associated factors in a general population of Bangladesh. Indian J Gastroenterol. 2014;33: 265–273. pmid:24664445
  2. 2. Vakil N, Stelwagon M, Shea EP, Miller S. Symptom burden and consulting behavior in patients with overlapping functional disorders in the US population. United Eur Gastroenterol J. 2016. pmid:27403308
  3. 3. Lee BJ, Bak YT. Irritable bowel syndrome, gut microbiota and probiotics. J Neurogastroenterol Motil. 2011;17: 252–266. pmid:21860817
  4. 4. Oka P, Parr H, Barberio B, Black CJ, Savarino E V., Ford AC. Global prevalence of irritable bowel syndrome according to Rome III or IV criteria: a systematic review and meta-analysis. Lancet Gastroenterol Hepatol. 2020. pmid:32702295
  5. 5. Lacy BE, Mearin F, Chang L, Chey WD, Lembo AJ, Simren M, et al. Bowel disorders. Gastroenterology. 2016;150: 1393–1407. pmid:27144627
  6. 6. Irritable bowel syndrome: A mild disorder; Purely symptomatic treatment. Prescrire Int. 2009;18: 75–79. pmid:19585728
  7. 7. Ford AC, Lacy BE, Harris LA, Quigley EMM, Moayyedi P. Effect of Antidepressants and Psychological Therapies in Irritable Bowel Syndrome. Am J Gastroenterol. 2019;114: 21–39. pmid:30177784
  8. 8. Kleinstäuber M, Witthöft M, Steffanowski A, van Marwijk H, Hiller W, Lambert MJ. Pharmacological interventions for somatoform disorders in adults. Cochrane Database Syst Rev. 2014; CD010628. pmid:25379990
  9. 9. Dale HF, Rasmussen SH, Asiller ÖÖ, Lied GA. Probiotics in irritable bowel syndrome: An up-to-date systematic review. Nutrients. 2019;11: 2048. pmid:31480656
  10. 10. Pittayanon R, Lau JT, Yuan Y, Leontiadis GI, Tse F, Surette M, et al. Gut Microbiota in Patients With Irritable Bowel Syndrome—A Systematic Review. Gastroenterology. 2019. pmid:30940523
  11. 11. Morrison DJ, Preston T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes. 2016;7: 189–200. pmid:26963409
  12. 12. Coss-Adame E, Rao SSC. Brain and gut interactions in irritable bowel syndrome: New paradigms and new understandings. Curr Gastroenterol Rep. 2014;16: 1–8. pmid:24595616
  13. 13. Simrén M, Törnblom H, Palsson OS, Van Oudenhove L, Whitehead WE, Tack J. Cumulative Effects of Psychologic Distress, Visceral Hypersensitivity, and Abnormal Transit on Patient-reported Outcomes in Irritable Bowel Syndrome. Gastroenterology. 2019;157: 391-402.e2. pmid:31022401
  14. 14. Duan R, Zhu S, Wang B, Duan L. Alterations of gut microbiota in patients with irritable bowel syndrome based on 16s rRNA-targeted sequencing: A systematic review. Clin Transl Gastroenterol. 2019;10: e00012. pmid:30829919
  15. 15. Trush EA, Poluektova EA, Beniashvilli AG, Shifrin OS, Poluektov YM, Ivashkin VT. The Evolution of Human Probiotics: Challenges and Prospects. Probiotics and Antimicrobial Proteins. 2020. pmid:31907861
  16. 16. Didari T, Mozaffari S, Nikfar S, Abdollahi M. Effectiveness of probiotics in irritable bowel syndrome: Updated systematic review with meta-analysis. World J Gastroenterol. 2015;21: 3072–3084. pmid:25780308
  17. 17. Ford AC, Lacy BE, Talley NJ. Irritable Bowel Syndrome. N Engl J Med. 2017;376: 2566–2578. pmid:28657875
  18. 18. Amato A, Liotta R, Mulè F. Effects of menthol on circular smooth muscle of human colon: Analysis of the mechanism of action. Eur J Pharmacol. 2014;740: 295–301. pmid:25046841
  19. 19. Gaudioso C, Hao J, Martin-Eauclaire MF, Gabriac M, Delmas P. Menthol pain relief through cumulative inactivation of voltage-gated sodium channels. Pain. 2012;153: 473–484. pmid:22172548
  20. 20. Pan R, Tian Y, Gao R, Li H, Zhao X, Barrett JE, et al. Central mechanisms of menthol-induced analgesia. J Pharmacol Exp Ther. 2012;343: 661–672. pmid:22951274
  21. 21. Imai H, Osawa K, Yasuda H, Hamashima H, Arai T, Sasatsu M. Inhibition by the essential oils of peppermint and spearmint of the growth of pathogenic bacteria. Microbios. 2001;106: 31–39. pmid:11549238
  22. 22. Patrick L. Gastroesophageal reflux disease (GERD): a review of conventional and alternative treatments. Altern Med Rev. 2011;16: 116–133. Available: http://search.ebscohost.com/login.aspx?direct=true&profile=ehost&scope=site&authtype=crawler&jrnl=10895159&AN=61445441&h=Po9FKCHAth5IC0RATltSqXC3FKSxDGA3LB%2FxS1zJR75kiTBjsKt3NhESxGtQabGE36UGKZ4E8h3b0iq4HLGYWg%3D%3D&crl=c pmid:21649454
  23. 23. D’Alessio PA, Ostan R, Bisson JF, Schulzke JD, Ursini M V., Béné MC. Oral administration of d-Limonene controls inflammation in rat colitis and displays anti-inflammatory properties as diet supplementation in humans. Life Sci. 2013;92: 1151–1156. pmid:23665426
  24. 24. Rehman MU, Tahir M, Khan AQ, Khan R, Oday-O-Hamiza , Lateef A, et al. d-limonene suppresses doxorubicin-induced oxidative stress and inflammation via repression of COX-2, iNOS, and NFκB in kidneys of Wistar rats. Exp Biol Med. 2014;239: 465–476. pmid:24586096
  25. 25. Ghayur MN, Gilani AH. Pharmacological basis for the medicinal use of ginger in gastrointestinal disorders. Dig Dis Sci. 2005;50: 1889–1897. pmid:16187193
  26. 26. Giacosa A, Morazzoni P, Bombardelli E, Riva A, Porro GB, Rondanelli M. Can nausea and vomiting be treated with Ginger extract? Eur Rev Med Pharmacol Sci. 2015;19: 1291–1296. pmid:25912592
  27. 27. Simon A, Darcsi A, Kéry Á, Riethmüller E. Blood-brain barrier permeability study of ginger constituents. J Pharm Biomed Anal. 2020;177: 112820. pmid:31476432
  28. 28. Ivashkin VT, Shelygin Y., Baranskaya YK, Belousova YA, Beniashvili AG, Vasilyev S., et al. Diagnosis and treatment of the irritable bowel syndrome: clinical guidelines of the Russian gastroenterological association and Russian association of coloproctology. gastro-j.ru. 2017;27: 76–93. https://doi.org/10.22416/1382-4376-2017-27-5-76-93
  29. 29. Ivashkin VT, Mayev IV, Sheptulin AA, Lapina TL, Trukhmanov AS, Kartavenko IM, et al. Diagnosis and treatment of the functional dyspepsia: clinical guidelines of the Russian Gastroenterological Association. Ross Z Gastroenterol Gepatol Koloproktol. 2017;27: 50–61. https://doi.org/10.22416/1382-4376-2017-27-1-50-61
  30. 30. Faul F, Erdfelder E, Lang AG, Buchner A. G*Power 3: A flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behavior Research Methods. 2007. pmid:17695343
  31. 31. Ivashkin VT, Poluektova EA, Glazunov AB, Putilovskiy MA, Epstein OI. Pathogenetic approach to the treatment of functional disorders of the gastrointestinal tract and their intersection: Results of the Russian observation retrospective program COMFORT. BMC Gastroenterol. 2019. pmid:31892312
  32. 32. Ivashkin V, Sheptulin A, Shifrin O, Poluektova E, Pavlov C, Ivashkin K, et al. Clinical validation of the “7 × 7” questionnaire for patients with functional gastrointestinal disorders. J Gastroenterol Hepatol. 2019;34: 1042–1048. pmid:30462850
  33. 33. Parikh HI, Koparde VN, Bradley SP, Buck GA, Sheth NU. MeFiT: Merging and filtering tool for illumina paired-end reads for 16S rRNA amplicon sequencing. BMC Bioinformatics. 2016;17: 491. pmid:27905885
  34. 34. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: High-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13: 581–583. pmid:27214047
  35. 35. Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, et al. Ribosomal Database Project: Data and tools for high throughput rRNA analysis. Nucleic Acids Res. 2014;42: D633–D642. pmid:24288368
  36. 36. Narayan NR, Weinmaier T, Laserna-Mendieta EJ, Claesson MJ, Shanahan F, Dabbagh K, et al. Piphillin predicts metagenomic composition and dynamics from DADA2-corrected 16S rDNA sequences. BMC Genomics. 2020;21: 56. pmid:31952477
  37. 37. R Development Core Team. R: A language and environment for statistical computing. Vienna, Austria. 2017. doi:R Foundation for Statistical Computing, Vienna, Austria. ISBN 3-900051-07-0, URL http://www.R-project.org.
  38. 38. Wickham H. ggplot2: Elegant Graphics for Data Analysis. Second Edition. Springer. Media. 2016. https://doi.org/10.1007/978-0-387-98141-3
  39. 39. Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. Psychobiotics and the Manipulation of Bacteria–Gut–Brain Signals. Trends Neurosci. 2016;39: 763–781. pmid:27793434
  40. 40. König J, Wells J, Cani PD, García-Ródenas CL, MacDonald T, Mercenier A, et al. Human intestinal barrier function in health and disease. Clin Transl Gastroenterol. 2016;7: e196. pmid:27763627
  41. 41. Ivashkin VT, Mayev IV, Ivashkin KV, Korochanskaya NV, Lopina OD, Lapina TL, et al. The role of protective factors disorders of acid-related diseases pathogenesis. Ross Z Gastroenterol Gepatol Koloproktol. 2016;26: 115–116. https://doi.org/10.22416/1382-4376-2016-26-3
  42. 42. Hijova E, Chmelarova A. Short chain fatty acids and colonic health. Bratisl Lek Listy. 2007;108: 354–358. pmid:18203540
  43. 43. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat Rev Microbiol. 2014;12: 661–672. pmid:25198138
  44. 44. Tap J, Derrien M, Törnblom H, Brazeilles R, Cools-Portier S, Doré J, et al. Identification of an Intestinal Microbiota Signature Associated With Severity of Irritable Bowel Syndrome. Gastroenterology. 2017;152: 111–123.e8. pmid:27725146
  45. 45. Wang X, Qi Q, Wang Y, Wu H, Jin X, Yao H, et al. Gut microbiota was modulated by moxibustion stimulation in rats with irritable bowel syndrome. Chin Med. 2018;13: 63. pmid:30574173
  46. 46. Tana C, Umesaki Y, Imaoka A, Handa T, Kanazawa M, Fukudo S. Altered profiles of intestinal microbiota and organic acids may be the origin of symptoms in irritable bowel syndrome. Neurogastroenterol Motil. 2010;22: 512–e115. pmid:19903265
  47. 47. Lee KJ, Tack J. Altered intestinal microbiota in irritable bowel syndrome. Neurogastroenterol Motil. 2010;22: 493–498. pmid:20414959
  48. 48. Lee GH, Kumar S, Lee JH, Chang DH, Kim DS, Choi SH, et al. Genome sequence of oscillibacter ruminantium strain GH1, isolated from rumen of korean native cattle. J Bacteriol. 2012;194: 6362. pmid:23105088
  49. 49. Katano Y, Fujinami S, Kawakoshi A, Nakazawa H, Oji S, Iino T, et al. Complete genome sequence of Oscillibacter valericigenes Sjm18-20 T (= NBRC 101213 T). Stand Genomic Sci. 2012;6: 406–414. pmid:23408234
  50. 50. Gomez-Arango LF, Barrett HL, Wilkinson SA, Callaway LK, McIntyre HD, Morrison M, et al. Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes. 2018;9: 189–201. pmid:29144833
  51. 51. Lambeth SM, Carson T, Lowe J, Ramaraj T, Leff JW, Luo L, et al. Composition Diversity and Abundance of Gut Microbiome in Prediabetes and Type 2 Diabetes. J Diabetes Obes. 2015;2: 1–7. pmid:26756039
  52. 52. Karlsson FH, Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun. 2012;3. pmid:23212374
  53. 53. Astbury S, Atallah E, Vijay A, Aithal GP, Grove JI, Valdes AM. Lower gut microbiome diversity and higher abundance of proinflammatory genus Collinsella are associated with biopsy-proven nonalcoholic steatohepatitis. Gut Microbes. 2019;11: 569–580. pmid:31696774
  54. 54. Mariadason JM, Barkla DH, Gibson PR. Effect of short-chain fatty acids on paracellular permeability in Caco- 2 intestinal epithelium model. Am J Physiol—Gastrointest Liver Physiol. 1997;272: G705–G712. pmid:9142899
  55. 55. Gu X, Song LJ, Li LX, Liu T, Zhang MM, Li Z, et al. Fusobacterium nucleatum Causes Microbial Dysbiosis and Exacerbates Visceral Hypersensitivity in a Colonization-Independent Manner. Front Microbiol. 2020. pmid:32733392
  56. 56. Long X, Li M, Li LX, Sun YY, Zhang WX, Zhao DY, et al. Butyrate promotes visceral hypersensitivity in an IBS-like model via enteric glial cell-derived nerve growth factor. Neurogastroenterol Motil. 2018. pmid:29052293
  57. 57. Mei L, Zhou J, Su Y, Mao K, Wu J, Zhu C, et al. Gut microbiota composition and functional prediction in diarrhea-predominant irritable bowel syndrome. BMC Gastroenterol. 2021. pmid:33663411
  58. 58. Carco C, Young W, Gearry RB, Talley NJ, McNabb WC, Roy NC. Increasing Evidence That Irritable Bowel Syndrome and Functional Gastrointestinal Disorders Have a Microbial Pathogenesis. Frontiers in Cellular and Infection Microbiology. 2020. pmid:33014892
  59. 59. Barandouzi ZA, Lee J, Maas K, Starkweather AR, Cong XS. Altered gut microbiota in irritable bowel syndrome and its association with food components. J Pers Med. 2021. pmid:33429936
  60. 60. Durbán A, Abellán JJ, Jiménez-Hernández N, Artacho A, Garrigues V, Ortiz V, et al. Instability of the faecal microbiota in diarrhoea-predominant irritable bowel syndrome. FEMS Microbiol Ecol. 2013. pmid:23889283
  61. 61. Si JM, Yu YC, Fan YJ, Chen SJ. Intestinal microecology and quality of life in irritable bowel syndrome patients. World J Gastroenterol. 2004. pmid:15188510
  62. 62. Liu Y, Yuan X, Li L, Lin L, Zuo X, Cong Y, et al. Increased Ileal Immunoglobulin A Production and Immunoglobulin A-Coated Bacteria in Diarrhea-Predominant Irritable Bowel Syndrome. Clin Transl Gastroenterol. 2020. pmid:32352710