Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Jan 11.
Published in final edited form as: Probiotics Antimicrob Proteins. 2011 Mar;3(1):1–7. doi: 10.1007/s12602-010-9059-y

Gut microbiota is not modified by Randomized, Double-blind, Placebo-controlled Trial of VSL#3 in Diarrhea-predominant Irritable Bowel Syndrome

Sonia Michail, Harshavardhan Kenche
PMCID: PMC3255476  NIHMSID: NIHMS347351  PMID: 22247743

Abstract

Irritable Bowel Syndrome (IBS) is a common condition that negatively impacts the quality of life for many individuals. The exact etiology of this disorder is largely unknown; however, emerging studies suggest that the gut microbiota is a contributing factor. Several clinical trials show that probiotics, such as VSL#3, can have a favorable effect on IBS. This double-blind, randomized placebo-controlled study has been conducted in diarrhea-predominant IBS subjects in order to investigate the effect of VSL#3 on the fecal microbiota. The bacterial composition of the fecal microbiota was investigated using high-throughput microarray technology to detect 16S RNA. Twenty four subjects were randomized to receive VSL#3 or placebo for 8 weeks. IBS symptoms were monitored using GSRS and quality of life questionnaires. A favorable change in Satiety subscale was noted in the VSL #3 groups. However, the consumption of the probiotic did not change the gut microbiota. There were no adverse events or any safety concerns encountered during this study. To summarize, the use of VSL#3 in this pilot study was safe and showed improvement in specific GSRS-IBS scores in diarrhea-predominant IBS subjects. The gut microbiota was not affected by VSL#3 consumption suggesting that the mechanism of action is not directly linked to the microbiota.

Keywords: Irritable Bowel Syndrome, Diarrhea, Probiotics, Microbiota

Introduction

Irritable Bowel Syndrome (IBS) is the most common disorder encountered by gastroenterologists. It can be disabling with significant impact on the quality of life, yet the exact etiology and pathophysiology remain undefined. Many theories suggest immune activation with low-grade intestinal inflammation and an altered host brain-gut response. Recently, the gut microbiota is being considered an important contributor to the development of IBS; based on observations of post-infectious symptoms, response to probiotics or specific antimicrobials, the relapse of symptoms with the use of antibiotics that change the microbiota, and finally, the suspicion of small bowel bacterial overgrowth. It is estimated that the human colon contains up to 1014 bacteria [1]. Although this diverse microbial load is largely beneficial, it has been postulated that altered bacterial populations or products of bacterial metabolism may contribute to human disease. The mechanisms by which altered fecal microbiota induce disease are poorly understood. Fecal short chain fatty acids produced by microbiota are critical for colonic epithelial maintenance, yet they are clearly different in IBS [2]. Maximal gas production and hydrogen excretion after lactulose is increased in IBS as colonic-gas production, particularly of hydrogen, is greater in patients with IBS than in controls, and both symptoms and gas production are reduced by an exclusion diet. This reduction may be associated with alterations in the activity of hydrogen-consuming bacteria further emphasizing the importance of fermentation in the pathogenesis of IBS [3]. Moreover, increased bacterial methane production has been linked to constipation in IBS [4]. In this population, serotonin release is blunted, suggesting a possible neurochemical basis for impaired motor function[5]. Now, several studies have demonstrated that patients with IBS have altered gut microbiota[6-9] leading to the development of probiotic studies that suggest a favorable effect on IBS[10-12]. One of the questions that remain unanswered is how the probiotic products influence the gut microbiota. VSL#3 is a polymicrobial probiotic product that shows promise in treating IBS[13-15]. This pilot study was designed to investigate the safety, efficacy and the effect of VSL#3 on the gut microbiota of patients with diarrhea-predominant IBS.

Methods

Clinical Study

Protocol and assignment

Twenty four subjects with diarrhea-predominant irritable bowel syndrome that met the Rome III criteria were randomly and blindly assigned to consume VSL#3, 900 billion bacteria per day or placebo composed of cornstarch (base ingredient for probiotic product) for eight weeks. Subjects were excluded if they have been on probiotics or antibiotics for three months prior to enrollment. The safety of the product was monitored closely throughout the study. IBS symptoms were monitored using GSRS; a clinical rating scale for gastrointestinal symptoms in patients with irritable bowel syndrome on a weekly basis. Quality of life (QOL) was monitored using QOL questionnaires at baseline and week 8. Fecal samples were collected at baseline, week 4 and week 8. The primary goal of this study was to examine the effect of the probiotic VSL#3 on the gut microbiota in diarrhea-predominant. This study was approved by the Institutional Review Board. Subjects were electronically randomized through the pharmacy department. Compliance was monitored by asking subjects to bring all used and unused study medication sachets.

Masking

The group assignment of subjects was concealed to the subject and all the investigating team.

Microbiota study

The bacterial make up of the gut microbiota was investigated using high-throughput microarray technology as we previously reported[16]. All fecal specimens were homogenized and immediately frozen at −80°C.

DNA isolation

Fecal DNA was isolated using ZR Fecal DNA kit (Zymo Research Corporation, Orange, CA). Frozen fecal samples (150 mg) were used following the manufacturer’s protocol. Eluted DNA was checked for quality by 1% agarose gel electrophoresis. The eluted DNA was then quantified on a NanoVue Spectrophotometer (GE Health care, NJ).

PCR reaction

The genomic DNA (gDNA) was diluted to a concentration of 400 ng/μl and universal primers “16S rRNA for (forward AGA GTT TGA TCC TGG CTC AG)” and “16S rRNA Rev (reverse ACG GCT ACC TTG TTA CGA CTT)” from Integrated DNA technologies, were used to amplify the whole 1.5kb fragment of 16S ribosomal RNA gene.

The reaction was carried out for 30 cycles on StepOne Real-time PCR system (Applied Biosystems, Foster City, CA) as a normal thermo-cycling reaction, for 90 seconds at 75°C. The amplified product was cleaned up using Qiagen MinElute PCR Purification Kit as per the manufacturer’s suggested protocol. The amplified product was verified by 1% agarose electrophoresis alongside 100 bp Low Scale DNA Ladder (Fisher Scientific, Pittsburgh, PA) for the presence of a distinct band at 1500 bp position. The amplified DNA was then quantified on a NanoVue Spectrophotometer.

DNA fragmentation

Amplified 16S rDNA (400 ng) was used as target for the microarrays. DNase I enzyme (New England Biolabs, Ipswich, MA) was diluted to a concentration of 0.05 units/μl in DNase reaction buffer. Ribosomal DNA fragmentation was carried out using working concentration of enzyme at 0.05 units DNase I/ μg of amplified DNA as this concentration has been shown to produce ideal fragment size for microarray hybridization16. The DNA was incubated at 37°C along with the enzyme for 10 minutes; thereafter the enzyme was heat-inactivated at 75°C for another 10 minutes.

DNA labeling reaction

The single color protocol from Agilent Technologies (version 5.0, June 2007) was adopted for DNA labeling reaction using Agilent Genomic DNA Enzymatic Labeling Kit (Agilent Technologies, Santa Clara, CA) and using single dye (either with Cy-3 or Cy-5). The thermo-cycler was programmed for the following temperatures: 95°C, 37°C and 65°C. To 13 μl of the fragmented DNA, 2.5 μl of Agilent primers were added and the mixture was incubated at 95°C for 3 minutes and then cooled to 4°C. The samples were briefly spun in a microcentrifuge to collect the contents at the bottom. Reaction buffer, dNTPs, dye (Cy-3 or Cy-5) and Exo-Klenow fragment was added to the annealed primer-template mix and incubated at 37°C for 2 hours followed by heat inactivation with the enzyme at 65°C for 10 minutes. The labeled DNA was mixed of 1X TE buffer (pH 8.0) and passed through Microcon YM-30 filters to clean the DNA. There were 38,340 probes on the array with more probes than signals obtained.

Microarray hybridization

The following components were added in the order indicated: Cy-3 or Cy-5 labeled DNA, Cot-1 DNA, Agilent 10X blocking agent, and Agilent hybridization buffer. Components were mixed thoroughly, quickly spun and then incubated at 95°C for 3 minutes and 37°C for 30 minutes. The reaction mixture (40 μl) was loaded onto a gasket chamber and the printed microarray was placed over the gasket with the active side of the array facing down. The sandwich assembly was thoroughly inspected for any leaks then hybridized for 24 hours. Washing and scanning (Agilent G2565CA Microarray scanner with SureScan High resolution technology, Agilent Technologies, Santa Clara, CA) were then carried out as per manufacturer’s protocol and the raw data was extracted using Agilent Feature Extraction software (Version 10.1).

Statistical Analyses

Microarray Data

Signals from 852 separate bacteria were obtained from the 15 VSL patients at baseline, week 4, and week 8. The bacteria were classified into 120 different genera, 53 families, 25 orders, 14 classes, and 8 phyla. The signal from each bacterium for each patient was then divided by its genus average copy number to obtain an adjusted signal for each bacterium. The 852 adjusted signals were summed and the percent of the total adjusted signal was determined for all bacteria. Adjusted signals were then summed by genus, family, order, class, and phylum and the percents of the total signal were determined for each hierarchical classification level.

For statistical analyses, all adjusted signals were transformed using the natural logarithmic transformation. Comparisons between the baseline, week 4 and week 8 signals were made with repeated measures analysis of variance. P values were adjusted for multiple comparisons using Bonferroni corrections.

Results

Clinical Outcomes

Twenty four subjects were enrolled in this study; 15 were randomly assigned to the VSL #3 group and 9 to the placebo group. Two thirds of the patients in each group were females, twenty two patients were white, and two were African-American. The mean (±SD) age was 21.8±17 years, and mean weight was 80±27 kg (range 52-132 kg). There were no differences between the groups for age and weight, and both groups’ weights remained stable throughout the treatment period.

GSRS-IBS pain subscale scores decreased significantly over the treatment period in both groups, but there was no difference between the groups in the degree of change (Table 1). The mean baseline pain score for both groups combined was 3.5±1.2, and decreased by 1.6±1.3 points at the end of the treatment period. Significant decreases also occurred in the combined groups in the bloating, diarrhea, satiety, and global scores. The decrease was significantly greater in the VSL #3 group for the satiety subscale.

Table 1.

GSRS-IBS scores and comparisons between the VSL #3 and placebo groups

Subscale Group Baseline W1 W2 W3 W4 W5 W6 W7 W8 P Values1
Pain VSL #3 4.0±1.3 1.9±0.7 2.7±1.2 1.7±0.8 2.4±0.7 2.6±1.0 2.1±1.0 2.0±0.6 1.9±0.9 .81, <.001, .28
Placebo 3.0±1.0 2.0±1.2 2.6±1.3 2.4±1.1 2.0±1.0 2.2±1.0 1.9±0.7 2.1±1.0 2.0±0.9
Bloating VSL #3 2.7±0.9 1.1±0.2 2.1±0.9 1.9±0.6 1.7±0.3 1.9±0.2 1.5±0.3 1.9±1.0 1.6±0.3 .52, .01, .15
Placebo 1.6±0.4 1.3±0.5 1.7±0.7 1.7±1.1 1.5±0.8 1.7±1.0 1.6±1.2 1.5±0.9 1.5±0.9
Constipation VSL #3 1.8±0.8 1.5±0.5 1.4±0.5 2.0±1.0 1.5±0.5 1.1±0.2 1.4±0.5 1.5±0.9 1.2±0.4 .58, .32, .42
Placebo 2.2±1.2 1.2±0.4 2.3±2.2 1.4±0.9 1.8±0.6 1.5±0.5 1.9±1.2 1.5±0.7 1.5±0.7
Diarrhea VSL #3 3.1±1.6 1.6±0.5 2.6±1.3 1.9±0.8 1.7±0.7 2.2±1.2 1.5±0.4 1.5±0.5 1.6±0.5 .56, <.001, .89
Placebo 3.1±1.2 2.5±1.3 2.6±1.5 2.4±1.5 2.1±0.9 2.3±0.9 1.9±0.9 1.9±0.9 1.8±0.8
Satiety VSL #3 2.3±0.9 1.2±0.3 1.8±1.2 1.2±0.4 1.3±0.4 1.1±0.2 1.1±0.2 1.1±0.2 1.0±0.0 .65, .01, .02
Placebo 1.8±1.3 1.6±0.9 1.4±0.9 1.7±1.3 1.7±1.6 1.5±1.1 1.8±1.8 1.5±1.1 1.5±1.1
Global score VSL #3 2.8±0.7 1.5±0.2 2.2±0.9 1.8±0.3 1.7±0.2 1.8±0.4 1.5±0.2 1.6±0.3 1.5±0.3 .81, .001, .51
Placebo 2.4±0.6 1.8±0.7 2.2±1.1 2.0±1.2 1.8±0.8 1.9±0.7 1.8±1.0 1.7±0.8 1.7±0.8

Values in table are the mean±SD. W1-W8 are treatment weeks 1-8.

1

P values are the group, treatment, and interaction p values respectively from two-way ANOVAs.

Significant decreases in scores were also observed in the combined groups for 3 of 7 QOL statements and the overall QOL score, with no differences between the groups (Table 2). The baseline QOL score was 2.7±1.1 and decreased to 1.9±0.7 after 8 weeks. The patients reported that they missed from 0-4 activities and 0-2 work days, and experienced from 0-2 significant life events during the 8 weeks prior to enrollment. There were no differences between the groups, and no changes occurred during the 8-week treatment period.

Table 2.

Quality of life (QOL) scores and comparisons between the VSL #3 and placebo groups

VSL #3 Placebo

QOL statement Baseline W8 Baseline W8 P Values1
I am bothered by how much time I spend on the toilet 2.8±1.8 2.0±0.7 2.5±0.6 2.5±1.7 .90, .44, .44
I feel I get less done because of my bowel problems 3.0±1.2 1.6±0.5 2.0±0.8 1.5±0.6 .22, .05, .30
I avoid stressful situations because of my bowel problems 3.2±1.3 2.0±0.7 2.3±1.5 1.8±1.0 .41, .05, .36
I watch the kind of food I eat because of my bowel problems 4.2±1.3 2.8±1.3 3.3±1.5 2.0±0.0 .25, .01, .86
It is important to be near a toilet because of my bowel problems 3.0±1.4 2.0±1.2 2.0±1.2 1.5±0.6 .31, .09, .53
I worry about losing control of my bowels 2.2±1.8 1.6±0.9 2.5±1.7 1.5±0.6 .89, .19, .73
Overall average score 3.0±1.3 2.1±0.8 2.4±1.0 1.8±0.6 .41, .05, .63

Values in table are the mean±SD. W8 is treatment week 8.

1

P values are the group, treatment, and interaction p values respectively from two-way ANOVAs.

Safety

There were no adverse events noted during the study.

Microarray Data

Of the 852 phylospecies analyzed, no differences were noted after adjustment for multiple comparisons. The number of species and percents of total signals by phylum and class are shown in Table 3. The class Clostridia contained the largest number of species (564) and comprised over 60% of the total signals at baseline, week 4, and week 8. There were no differences noted at baseline or with probiotic consumption at the Phylum, Class, Order, Family or Genus levels. Similarly, there was no change in the gut microbiota after placebo consumption (table 4).

Table 3.

Microarray analysis data for the VSL #3 subjects

Phylum Class (Subclass) No. of species Baseline W4 (p-value) W8 (p-value)
Actinobacteria Actinobacteria (Actinobacteridae) 20 2.1±0.5 1.6±0.2 1.6±0.3
Actinobacteria (Coriobacteridae) 9 2.6±0.6 2.3±0.3 2.4±0.3
Bacteroidetes Bacteroidia 142 16.2±6.1 19.6±2.4 19.7±1.9
Firmicutes Bacilli 20 3.7±2.7 3.1±0.9 3.1±0.4
Clostridia 564 62.7±5.3 62.4±2.9 62.6±2.5
Erysipelotrichi 39 5.4±2 5.0±0.4 4.8±0.5
Fusobacteria Fusobacteria 3 0.3±0.1 0.3±0.2 0.3±0.1
Lentisphaerae Lentisphaeria 1 0.5±0.2 0.4±0.1 0.3±0.1
Proteobacteria Alphaproteobacteria 9 0.6±0.2 0.6±0.2 0.5±0.1
Betaproteobacteria 18 1.9±1.7 1.2±0.5 1.2±0.7
Deltaproteobacteria 5 0.8±0.4 0.7±0.2 0.7±0.1
Epsilonproteobacteria 6 0.9±0.3 0.9±0.0 0.9±0.1
Gammaproteobacteria 11 1.3±0.8 1.0±0.1 0.9±0.1
Spirochaetes Spirochaetes 4 0.8±0.2 0.8±0.1 0.8±0.0
Verrucomicrobia Verrucomicrobiae 1 0.2±0.1 0.1±0.1 0.1±0.1

Values in table are the percents of the total adjusted signals (mean±SD). W4 and W8 are treatment weeks 4 and 8. None of the comparisons had a statistically significant p-value (adjusted p-value<0.05).

Table 4.

Microarray analysis data for the placebo subjects

Phylum Class (Subclass) No. of
species
Baseline W4 (p) W8 (p)
Actinobacteria Actinobacteria (Actinobacteridae) 20 1.1±0.1 1.2±0.1 (0.91) 1.3±0.1 (0.43)
Actinobacteria (Coriobacteridae) 9 1.0±0.07 1.1±0.09 (0.91) 1.1±0.09 (0.61)
Bacteroidetes Bacteroidetes 142 14.9±1.4 14.9±1.03 (0.91) 16.4±2.3 (0.43)
Firmicutes Bacilli 20 1.9±0.4 1.7±0.5 (0.91) 1.8±0.5 (0.43)
Clostridia 564 75.7±2.3 75.8±1.4 (0.91) 73.9±2.7 (0.43)
Erysipelotrichi 39 2.3±0.3 2.5±0.4 (0.94) 2.6±0.3 (0.43)
Fusobacteria Fusobacteria 3 0.1±0.05 0.12±0.01 (0.96) 0.12±0.03 (0.43)
Lentisphaerae Lentisphaerae 1 0.08±0.02 0.07±0.01 (0.91) 0.06±0.02 (0.68)
Proteobacteria Alphaproteobacteria 9 0.1±0.04 0.17±0.02 (0.91) 0.2±0.03 (0.43)
Betaproteobacteria 18 0.8±0.2 0.6±0.1 (0.75) 0.6±0.2 (0.43)
Deltaproteobacteria 5 0.2±0.04 0.2±0.0 (0.91) 0.29±0.02 (0.96)
Epsilonproteobacteria 6 0.2±0.02 0.2±0.01 (0.91) 0.3±0.03 (0.43)
Gammaproteobacteria 11 0.7±0.1 0.7±0.08 (0.91) 0.8±0.1 (0.43)
Spirochaetes Spirochaetes 4 0.1±0.02 0.1±0.01 (0.91) 0.1±0.1 (0.43)
Verrucomicrobia Verrucomicrobiae 1 0.07±0.05 0.07±0.05 (0.91) 0.05±0.05 (0.65)

Values in table are the percents of the total adjusted signals (mean±SD). W4 and W8 are treatment weeks 4 and 8. None of the comparisons had a statistically significant p-value (adjusted p-value<0.05).

Discussion

This study is the first placebo-controlled trial reporting culture-independent analysis of the gut microbiota after consumption of VSL#3. Abnormalities in intestinal microbiota have recently surfaced as a contributing factor to the development of irritable bowel syndrome. A significant reduction in bifidobacteria of the gut microbiota can result in an increased production of gas, dysmotility, and enteric neuromuscular dysfunction[17] which in turn can contribute to the development of IBS. Since gut microbiota in IBS is different from that of healthy subjects[18-20], probiotic-related changes in the enteric flora could reduce the non-desirable effects of bacteria in the gut[3] and favorably influence gut function. Probiotics, defined as live or attenuated bacteria or bacterial products that confer a significant health benefit to the host[21] have the potential to serve as a tool to explore gut microbial interactions. Probiotics, especially VSL#3 have therapeutic potential as they possess antibacterial and antiviral effects that could potentially prevent or ameliorate post-infectious IBS[22, 23]. They also have mucosal anti-inflammatory properties[24, 25] with an increase in mucus[26] production and reduction of the migration of neutrophils to the intestinal epithelium[27]. Reduction of mucosal inflammation can ameliorate the consequences of inflammation and reduce the neurochemical and impaired motor function found in many subjects with IBS. Furthermore, probiotics could alter stool and gas formation[28-30] or increase intestinal mucus secretion[31, 32], which could ameliorate symptoms such as constipation and diarrhea. Indeed, many probiotic studies suggest a favorable outcome in ameliorating symptoms of irritable bowel syndrome[10, 33, 34]. While results between studies are difficult to compare because of differences in study design, probiotic dose, and strain, there has been evidence of symptom improvement[14, 15, 35-38]. Several of these studies have involved either lactobacilli or bifidobacteria[39]. One of the probiotic products studied with success in this disorder is VSL#3[14, 15], a product that combines both bacterial groups. However, many of the studies are limited by lack of documentation of stool recovery of the probiotic agent and lack of knowledge of how the probiotic agent can modify the gut microbiota.

Whether probiotics exert their effect on the host through alteration of the gut microbiota remains a controversial issue. Animal studies confirm the ability of VSL#3 to modify the microbiota. VSL#3 strains survived in the gastrointestinal tract, increased the cecal concentrations of bifidobacteria, and modified cecal microflora metabolic activity in mice [40]. In human subjects with irritable bowel syndrome, the data is scarce. Only one study could be identified in the literature that has addressed this issue. Brigidi et al[13] investigated the gut microbiota in 10 human subjects with irritable bowel syndrome and functional diarrhea but used culture methodology. Most of the intestinal microbiota species are obligate anaerobes [41-43] making it difficult and inaccurate to depend on culture techniques to investigate the microbiota. Nevertheless, Brigidi could not find any significant changes in the gut microbiota as a result of VSL#3 use. Similarly in our study, the fecal gut microbiota did not appear to be affected by VSL#3 suggesting that alteration of the gut microbiota is not the mechanism by which this probiotic product can offer benefit to the host relevant to the findings in this study showing a beneficial effect on the “Satiety” parameter.

Recent studies suggest that obesity is associated with a change in the gut microbiota[44]. Therefore, analysis of the BMI of subjects was performed to determine if there was a difference between the two groups. We found no statistical difference in the BMI of subjects receiving placebo versus those receiving the probiotic product. Therefore, changes based on BMI would not be a contributing variable in this study.

This study is the first to describe a culture-independent molecular approach to the interrogation of the gut microbiota in human subjects with diarrhea-predominant IBS consuming VSL#3.

Acknowledgement

This work was supported by NIH grant 5R21 AT003400 and The Children’s Medical Center Research Foundation. The authors would like to acknowledge Ms. Stolfi for her help with the statistical analysis and Mrs. Debra Cunningham for her effort in patient recruitment.

References

  • 1.Suau A. Molecular tools to investigate intestinal bacterial communities. J Pediatr Gastroenterol Nutr. 2003;37(3):222–224. doi: 10.1097/00005176-200309000-00003. [DOI] [PubMed] [Google Scholar]
  • 2.Treem WR, Ahsan N, Kastoff G, Hyams JS. Fecal short-chain fatty acids in patients with diarrhea-predominant irritable bowel syndrome: in vitro studies of carbohydrate fermentation. J Pediatr Gastroenterol Nutr. 1996;23(3):280–286. doi: 10.1097/00005176-199610000-00013. [DOI] [PubMed] [Google Scholar]
  • 3.King TS, Elia M, Hunter JO. Abnormal colonic fermentation in irritable bowel syndrome. Lancet. 1998;352(9135):1187–1189. doi: 10.1016/s0140-6736(98)02146-1. [DOI] [PubMed] [Google Scholar]
  • 4.Pimentel M, Mayer AG, Park S, Chow EJ, Hasan A, Kong Y. Methane production during lactulose breath test is associated with gastrointestinal disease presentation. Dig Dis Sci. 2003;48(1):86–92. doi: 10.1023/a:1021738515885. [DOI] [PubMed] [Google Scholar]
  • 5.Pimentel M, Kong Y, Park S. IBS subjects with methane on lactulose breath test have lower postprandial serotonin levels than subjects with hydrogen. Dig Dis Sci. 2004;49(1):84–87. doi: 10.1023/b:ddas.0000011607.24171.c0. [DOI] [PubMed] [Google Scholar]
  • 6.Codling C, O’Mahony L, Shanahan F, Quigley EM, Marchesi JR. A molecular analysis of fecal and mucosal bacterial communities in irritable bowel syndrome. Dig Dis Sci. 2009;55(2):392–397. doi: 10.1007/s10620-009-0934-x. [DOI] [PubMed] [Google Scholar]
  • 7.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. 2009;22(5):512–519. e114–515. doi: 10.1111/j.1365-2982.2009.01427.x. [DOI] [PubMed] [Google Scholar]
  • 8.Parkes GC, Brostoff J, Whelan K, Sanderson JD. Gastrointestinal microbiota in irritable bowel syndrome: their role in its pathogenesis and treatment. Am J Gastroenterol. 2008;103(6):1557–1567. doi: 10.1111/j.1572-0241.2008.01869.x. [DOI] [PubMed] [Google Scholar]
  • 9.Kassinen A, Krogius-Kurikka L, Makivuokko H, Rinttila 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(1):24–33. doi: 10.1053/j.gastro.2007.04.005. [DOI] [PubMed] [Google Scholar]
  • 10.Hoveyda N, Heneghan C, Mahtani KR, Perera R, Roberts N, Glasziou P. A systematic review and meta-analysis: probiotics in the treatment of irritable bowel syndrome. BMC Gastroenterol. 2009;9:15. doi: 10.1186/1471-230X-9-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Nikfar S, Rahimi R, Rahimi F, Derakhshani S, Abdollahi M. Efficacy of probiotics in irritable bowel syndrome: a meta-analysis of randomized, controlled trials. Dis Colon Rectum. 2008;51(12):1775–1780. doi: 10.1007/s10350-008-9335-z. [DOI] [PubMed] [Google Scholar]
  • 12.McFarland LV, Dublin S. Meta-analysis of probiotics for the treatment of irritable bowel syndrome. World J Gastroenterol. 2008;14(17):2650–2661. doi: 10.3748/wjg.14.2650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brigidi P, Vitali B, Swennen E, Bazzocchi G, Matteuzzi D. Effects of probiotic administration upon the composition and enzymatic activity of human fecal microbiota in patients with irritable bowel syndrome or functional diarrhea. Res Microbiol. 2001;152(8):735–741. doi: 10.1016/s0923-2508(01)01254-2. [DOI] [PubMed] [Google Scholar]
  • 14.Kim HJ, Camilleri M, McKinzie S, Lempke MB, Burton DD, Thomforde GM, Zinsmeister AR. A randomized controlled trial of a probiotic, VSL#3, on gut transit and symptoms in diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2003;17(7):895–904. doi: 10.1046/j.1365-2036.2003.01543.x. [DOI] [PubMed] [Google Scholar]
  • 15.Kim HJ, Roque MI Vazquez, Camilleri M, Stephens D, Burton DD, Baxter K, Thomforde G, Zinsmeister AR. A randomized controlled trial of a probiotic combination VSL# 3 and placebo in irritable bowel syndrome with bloating. Neurogastroenterol Motil. 2005;17(5):687–696. doi: 10.1111/j.1365-2982.2005.00695.x. [DOI] [PubMed] [Google Scholar]
  • 16.Paliy O, Kenche H, Abernathy F, Michail S. High-throughput quantitative analysis of the human intestinal microbiota with a phylogenetic microarray. Appl Environ Microbiol. 2009;75(11):3572–3579. doi: 10.1128/AEM.02764-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Collins SM. The immunomodulation of enteric neuromuscular function: implications for motility and inflammatory disorders. Gastroenterology. 1996;111(6):1683–1699. doi: 10.1016/s0016-5085(96)70034-3. [DOI] [PubMed] [Google Scholar]
  • 18.Drossman DA, Camilleri M, Mayer EA, Whitehead WE. AGA technical review on irritable bowel syndrome. Gastroenterology. 2002;123(6):2108–2131. doi: 10.1053/gast.2002.37095. [DOI] [PubMed] [Google Scholar]
  • 19.Quigley EM. Current concepts of the irritable bowel syndrome. Scand J Gastroenterol Suppl. 2003;(237):1–8. doi: 10.1080/00855910310001403. [DOI] [PubMed] [Google Scholar]
  • 20.Bradley HK, Wyatt GM, Bayliss CE, Hunter JO. Instability in the faecal flora of a patient suffering from food-related irritable bowel syndrome. J Med Microbiol. 1987;23(1):29–32. doi: 10.1099/00222615-23-1-29. [DOI] [PubMed] [Google Scholar]
  • 21.Gorbach SL. Probiotics in the third millennium. Dig Liver Dis. 2002;34(Suppl 2):S2–7. doi: 10.1016/s1590-8658(02)80155-4. [DOI] [PubMed] [Google Scholar]
  • 22.von Wright A, Salminen S. Probiotics: established effects and open questions. Eur J Gastroenterol Hepatol. 1999;11(11):1195–1198. [PubMed] [Google Scholar]
  • 23.Isolauri E, Kirjavainen PV, Salminen S. Probiotics: a role in the treatment of intestinal infection and inflammation? Gut. 2002;50(Suppl 3):III54–59. doi: 10.1136/gut.50.suppl_3.iii54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.McCarthy J, O’Mahony L, O’Callaghan L, Sheil B, Vaughan EE, Fitzsimons N, Fitzgibbon J, O’Sullivan GC, Kiely B, Collins JK, et al. Double blind, placebo controlled trial of two probiotic strains in interleukin 10 knockout mice and mechanistic link with cytokine balance. Gut. 2003;52(7):975–980. doi: 10.1136/gut.52.7.975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.O’Mahony L, Feeney M, O’Halloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G, O’Sullivan G, Shanahan F, Collins JK. Probiotic impact on microbial flora, inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol Ther. 2001;15(8):1219–1225. doi: 10.1046/j.1365-2036.2001.01027.x. [DOI] [PubMed] [Google Scholar]
  • 26.Mack DR, Michail S, Wei S, McDougall L, Hollingsworth MA. Probiotics inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression. Am J Physiol. 1999;276(4 Pt 1):G941–950. doi: 10.1152/ajpgi.1999.276.4.G941. [DOI] [PubMed] [Google Scholar]
  • 27.Michail S, Abernathy F. Lactobacillus plantarum inhibits the intestinal epithelial migration of neutrophils induced by enteropathogenic Escherichia coli. J Pediatr Gastroenterol Nutr. 2003;36(3):385–391. doi: 10.1097/00005176-200303000-00017. [DOI] [PubMed] [Google Scholar]
  • 28.Jiang T, Savaiano DA. In vitro lactose fermentation by human colonic bacteria is modified by Lactobacillus acidophilus supplementation. J Nutr. 1997;127(8):1489–1495. doi: 10.1093/jn/127.8.1489. [DOI] [PubMed] [Google Scholar]
  • 29.Jiang T, Mustapha A, Savaiano DA. Improvement of lactose digestion in humans by ingestion of unfermented milk containing Bifidobacterium longum. J Dairy Sci. 1996;79(5):750–757. doi: 10.3168/jds.S0022-0302(96)76422-6. [DOI] [PubMed] [Google Scholar]
  • 30.Jiang T, Savaiano DA. Modification of colonic fermentation by bifidobacteria and pH in vitro. Impact on lactose metabolism, short-chain fatty acid, and lactate production. Dig Dis Sci. 1997;42(11):2370–2377. [Google Scholar]
  • 31.Ouwehand AC, Isolauri E, Kirjavainen PV, Tolkko S, Salminen SJ. The mucus binding of Bifidobacterium lactis Bb12 is enhanced in the presence of Lactobacillus GG and Lact. delbrueckii subsp. bulgaricus. Lett Appl Microbiol. 2000;30(1):10–13. doi: 10.1046/j.1472-765x.2000.00590.x. [DOI] [PubMed] [Google Scholar]
  • 32.Ouwehand AC, Lagstrom H, Suomalainen T, Salminen S. Effect of probiotics on constipation, fecal azoreductase activity and fecal mucin content in the elderly. Ann Nutr Metab. 2002;46(3-4):159–162. doi: 10.1159/000063075. [DOI] [PubMed] [Google Scholar]
  • 33.Ford AC, Talley NJ, Quigley EM, Moayyedi P. Efficacy of probiotics in irritable bowel syndrome: a meta-analysis of randomized, controlled trials. Dis Colon Rectum. 2009;52(10):1805. doi: 10.1007/DCR.0b013e3181ae0ab8. author reply 1806. [DOI] [PubMed] [Google Scholar]
  • 34.Brenner DM, Moeller MJ, Chey WD, Schoenfeld PS. The utility of probiotics in the treatment of irritable bowel syndrome: a systematic review. Am J Gastroenterol. 2009;104(4):1033–1049. doi: 10.1038/ajg.2009.25. quiz 1050. [DOI] [PubMed] [Google Scholar]
  • 35.Bausserman M, Michail S. The use of Lactobacillus GG in irritable bowel syndrome in children: a double-blind randomized control trial. J Pediatr. 2005;147(2):197–201. doi: 10.1016/j.jpeds.2005.05.015. [DOI] [PubMed] [Google Scholar]
  • 36.Nobaek S, Johansson ML, Molin G, Ahrne S, Jeppsson B. Alteration of intestinal microflora is associated with reduction in abdominal bloating and pain in patients with irritable bowel syndrome. Am J Gastroenterol. 2000;95(5):1231–1238. doi: 10.1111/j.1572-0241.2000.02015.x. [DOI] [PubMed] [Google Scholar]
  • 37.Tsuchiya J, Barreto R, Okura R, Kawakita S, Fesce E, Marotta F. Single-blind follow-up study on the effectiveness of a symbiotic preparation in irritable bowel syndrome. Chin J Dig Dis. 2004;5(4):169–174. doi: 10.1111/j.1443-9573.2004.00176.x. [DOI] [PubMed] [Google Scholar]
  • 38.Halpern GM, Prindiville T, Blankenburg M, Hsia T, Gershwin ME. Treatment of irritable bowel syndrome with Lacteol Fort: a randomized, double-blind, cross-over trial. Am J Gastroenterol. 1996;91(8):1579–1585. [PubMed] [Google Scholar]
  • 39.Hamilton-Miller J. Probiotics in the management of irritable bowel syndrome: a review of clinical trials. Microb Ecol Health Dis. 2001;13:212–216. [Google Scholar]
  • 40.Gaudier E, Michel C, Segain JP, Cherbut C, Hoebler C. The VSL# 3 Probiotic Mixture Modifies Microflora but Does Not Heal Chronic Dextran-Sodium Sulfate-Induced Colitis or Reinforce the Mucus Barrier in Mice. J Nutr. 2005;135(12):2753–2761. doi: 10.1093/jn/135.12.2753. [DOI] [PubMed] [Google Scholar]
  • 41.Langendijk PS, Schut F, Jansen GJ, Raangs GC, Kamphuis GR, Wilkinson MH, Welling GW. Quantitative fluorescence in situ hybridization of Bifidobacterium spp. with genus-specific rRNA-targeted probes and its application in fecal samples. Appl Environ Microbiol. 1995;61(8):3069–3075. doi: 10.1128/aem.61.8.3069-3075.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wilson KH, Blitchington RB. Human colonic biota studied by ribosomal DNA sequence analysis. Appl Environ Microbiol. 1996;62(7):2273–2278. doi: 10.1128/aem.62.7.2273-2278.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Suau A, Bonnet R, Sutren M, Godon JJ, Gibson GR, Collins MD, Dore J. Direct analysis of genes encoding rRNA from complex communities reveals many novel molecular species within the human gut. Appl Environ Microbiol. 1999;65(11):4799–4807. doi: 10.1128/aem.65.11.4799-4807.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi: 10.1038/nature05414. [DOI] [PubMed] [Google Scholar]

RESOURCES