51
|
Moser AM, 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 2018; 58:2767-2778. [PMID: 30251020 PMCID: PMC6768888 DOI: 10.1007/s00394-018-1826-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 09/18/2018] [Indexed: 12/22/2022]
Abstract
Purpose Diarrhea-predominant irritable bowel syndrome (IBS-D) is a common functional gastrointestinal disorder. Probiotics and synbiotics have been shown to improve symptoms of IBS, although mechanisms of action are currently not understood. Methods We investigated the effects of a 4-week oral synbiotic treatment (OMNi-BiOTiC® Stress Repair) in ten IBS-D patients on gastrointestinal mucosal and fecal microbiota, mucosa-associated immune cells, and fecal short-chain fatty acids. The upper and lower gastrointestinal tracts were compared before and after a 4-week synbiotic treatment using endoscopic evaluation to collect mucosal specimens for FACS analysis and mucosal 16S rRNA gene analysis. In stool samples, analysis for fecal SCFAs using GC–MS, fecal zonulin using ELISA, and fecal 16S rRNA gene analysis was performed. Results Synbiotics led to an increased microbial diversity in gastric (p = 0.008) and duodenal (p = 0.025) mucosal specimens. FACS analysis of mucosal immune cells showed a treatment-induced reduction of CD4+ T cells (60 vs. 55%, p = 0.042) in the ascending colon. Short-chain fatty acids (acetate 101 vs. 202 µmol/g; p = 0.007) and butyrate (27 vs. 40 µmol/g; p = 0.037) were elevated in fecal samples after treatment. Furthermore, treatment was accompanied by a reduction of fecal zonulin concentration (67 vs. 36 ng/ml; p = 0.035) and disease severity measured by IBS-SSS (237 vs. 54; p = 0.002). Conclusions Our findings indicate that a short-course oral synbiotic trial may influence the human gastrointestinal tract in IBS-D patients on different levels which are region specific. Electronic supplementary material The online version of this article (10.1007/s00394-018-1826-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adrian Mathias Moser
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Walter Spindelboeck
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Bettina Halwachs
- Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036, Graz, Austria
| | - Heimo Strohmaier
- Center for Medical Research, Medical University of Graz, Stiftingtalstraße 24, 8010, Graz, Austria
| | - Patrizia Kump
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Gregor Gorkiewicz
- Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.,Institute of Pathology, Medical University of Graz, Auenbruggerplatz 25, 8036, Graz, Austria
| | - Christoph Högenauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria. .,Theodor Escherich Laboratory for Microbiome Research, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| |
Collapse
|
52
|
Pietrzak A, Skrzydło-Radomańska B, Mulak A, Lipiński M, Małecka-Panas E, Reguła J, Rydzewska G. Guidelines on the management of irritable bowel syndrome: In memory of Professor Witold Bartnik. PRZEGLAD GASTROENTEROLOGICZNY 2018; 13:259-288. [PMID: 30581501 PMCID: PMC6300851 DOI: 10.5114/pg.2018.78343] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 09/09/2018] [Indexed: 12/15/2022]
Abstract
These guidelines constitute an update of the previous "Recommendations on the management of irritable bowel syndrome" issued in 2008. They have been developed by a Task Force organized by the Governing Board of the Polish Society of Gastroenterology. They discuss, with particular emphasis on new scientific data covering papers published since 2008, the aetiology, epidemiology, clinical presentation, diagnostic principles and criteria for the diagnosis, and recommendations for the treatment of irritable bowel syndrome (IBS). The English-language acronym for the syndrome (IBS) has become popular in medical and popular scientific language. It is also widely recognized by patients who identify with this diagnosis. Therefore, in the discussed guidelines, this is what we will use.
Collapse
Affiliation(s)
- Anna Pietrzak
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie Memorial Cancer Center, Institute of Oncology, Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | | - Agata Mulak
- Department of Gastroenterology and Hepatology, Wroclaw Medical University, Wroclaw, Poland
| | - Michał Lipiński
- Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Subdivision, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
| | - Ewa Małecka-Panas
- Department of Digestive Tract Diseases, Medical University of Lodz, Lodz, Poland
| | - Jarosław Reguła
- Department of Oncological Gastroenterology, Maria Sklodowska-Curie Memorial Cancer Center, Institute of Oncology, Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Grażyna Rydzewska
- Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Subdivision, Central Clinical Hospital of the Ministry of the Interior, Warsaw, Poland
- Department of the Prevention of Alimentary Tract Diseases, Faculty of Medicine and Health Science, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
53
|
Negi YK, Pandey C, Saxena N, Sharma S, Garg FC, Garg SK. Isolation of antibacterial protein from Lactobacillus spp. and preparation of probiotic curd. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2018; 55:2011-2020. [PMID: 29892101 PMCID: PMC5976584 DOI: 10.1007/s13197-018-3115-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 02/24/2018] [Accepted: 03/12/2018] [Indexed: 12/26/2022]
Abstract
The study was aimed to isolate antagonistic lactobacilli and the molecules responsible for their antagonistic ability from curd. Preparation of probiotic curd and the ability of the selected lactobacilli to suppress the pathogen therein was also assessed. All the 116 isolates were identified as Lactobacillus spp. based on morphological, biochemical and curdling assays. Five of these lactobacilli (Lb-17, Lb-33, Lb-108, Lb-112, and Lb-N3) were found most promising to inhibit all test pathogens (Staphylococcus aureus, Escherichia coli, Klebsiella pneumoniae, Salmonella typhi and Shigella sonnei). The cell-free culture supernatants of these five lactobacilli were recorded as thermo-tolerant when subjected to heat treatment at 100 °C for 20 min. The loss in the activity after protease treatment indicated the proteinaceous nature of the antimicrobial molecule present in the culture supernatants. Active protein (19 kDa) produced by lactobacilli was confirmed by SDS-PAGE followed by agar-overlay method. Antibiotic sensitivity assay revealed that the selected Lactobacillus spp. isolates were resistant to methicillin and vancomycin. Probiotic curd prepared by using Lb-108 and Lb-N3 was found to be superior to rest of the three isolates based on organoleptic tests and shelf-life. Complete inhibition of all the test pathogens in curd was shown by Lb-108 and Lb-N3. Inhibition spectrum, production of thermostable protein and preparation of quality curd suggest Lb-108 and Lb-N3 as promising candidates to prepare probiotic curd.
Collapse
Affiliation(s)
- Yogesh Kumar Negi
- Department of Basic Sciences, College of Forestry (VCSG UUHF), Ranichauri, Tehri Garhwal, Uttarakhand India
| | - Chitra Pandey
- Department of Basic Sciences, College of Forestry (VCSG UUHF), Ranichauri, Tehri Garhwal, Uttarakhand India
| | - Neha Saxena
- Department of Microbiology, SBS PG Institute of Biomedical Sciences and Research, Balawala, Dehradun India
| | - Sonika Sharma
- Department of Microbiology, SBS PG Institute of Biomedical Sciences and Research, Balawala, Dehradun India
| | - F. C. Garg
- Department of Microbiology, SBS PG Institute of Biomedical Sciences and Research, Balawala, Dehradun India
| | - S. K. Garg
- Department of Microbiology, RML Avadh University Faizabad, Faizabad, India
| |
Collapse
|
54
|
Ford AC, Moayyedi P, Chey WD, Harris LA, Lacy BE, Saito YA, Quigley EMM. American College of Gastroenterology Monograph on Management of Irritable Bowel Syndrome. Am J Gastroenterol 2018; 113:1-18. [PMID: 29950604 DOI: 10.1038/s41395-018-0084-x] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alexander C Ford
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds and Leeds Gastroenterology Institute, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Paul Moayyedi
- Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - William D Chey
- Division of Gastroenterology, Department of Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | | | | | - Eamonn M M Quigley
- Division of Gastroenterology and Hepatology, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA.
| | | |
Collapse
|
55
|
Ishaque SM, Khosruzzaman SM, Ahmed DS, Sah MP. 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. [PMID: 29801486 PMCID: PMC5970461 DOI: 10.1186/s12876-018-0788-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/26/2018] [Indexed: 02/07/2023] Open
Abstract
Background Accumulating evidence supports the view that an imbalance of gut bacteria contributes to IBS, and that increasing the mass of beneficial species may reduce the numbers of pathogenic bacteria and help alleviate symptoms. Methods In this double-blind trial 400 adult patients with moderate-to-severe symptomatic diarrhea-predominant IBS (IBS-D) were randomized to treatment with the multi-strain probiotic Bio-Kult® (14 different bacterial strains) or placebo for 16 weeks. The change in severity and frequency of abdominal pain was the primary outcome measure. Results Probiotic treatment significantly improved the severity of abdominal pain in patients with IBS-D. A 69% reduction for probiotic versus 47% for placebo (p < 0.001) equates to a 145 point reduction on the IBS-severity scoring system (IBS-SSS). The proportion of patients who rated their symptoms as moderate-to-severe was reduced from 100% at baseline to 14% for the multi-strain probiotic at follow-up (month 5) versus 48% for placebo (p < 0.001). Also, the number of bowel motions per day from month 2 onwards was significantly reduced in the probiotic group compared with the placebo group (p < 0.05). In addition to relieving symptoms, the probiotic markedly improved all dimensions of quality of life in the 34-item IBS-Quality of Life (IBS-QoL) questionnaire. No serious adverse events were reported. Conclusions The multi-strain probiotic was associated with significant improvement in symptoms in patients with IBS-D and was well-tolerated. These results suggest that probiotics confer a benefit in IBS-D patients which deserves further investigation. Trial registration [Clinicaltrials.gov NCT03251625; retrospectively registered on August 9, 2017]. Electronic supplementary material The online version of this article (doi:10.1186/s12876-018-0788-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shamsuddin M Ishaque
- Department of Gastroenterology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh.
| | - S M Khosruzzaman
- Department of Gastroenterology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Dewan Saifuddin Ahmed
- Department of Gastroenterology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Mukesh Prasad Sah
- Department of Gastroenterology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| |
Collapse
|
56
|
Hungin APS, Mitchell CR, Whorwell P, Mulligan C, Cole O, Agréus L, Fracasso P, Lionis C, Mendive J, Philippart de Foy J, Seifert B, Wensaas K, Winchester C, de Wit N. Systematic review: probiotics in the management of lower gastrointestinal symptoms - an updated evidence-based international consensus. Aliment Pharmacol Ther 2018; 47:1054-1070. [PMID: 29460487 PMCID: PMC5900870 DOI: 10.1111/apt.14539] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/15/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND In 2013, a systematic review and Delphi consensus reported that specific probiotics can benefit adult patients with irritable bowel syndrome (IBS) and other gastrointestinal (GI) problems. AIM To update the consensus with new evidence. METHODS A systematic review identified randomised, placebo-controlled trials published between January 2012 and June 2017. Evidence was graded, previously developed statements were reassessed by an 8-expert panel, and agreement was reached via Delphi consensus. RESULTS A total of 70 studies were included (IBS, 34; diarrhoea associated with antibiotics, 13; diarrhoea associated with Helicobacter pylori eradication therapy, 7; other conditions, 16). Of 15 studies that examined global IBS symptoms as a primary endpoint, 8 reported significant benefits of probiotics vs placebo. Consensus statements with 100% agreement and "high" evidence level indicated that specific probiotics help reduce overall symptom burden and abdominal pain in some patients with IBS and duration/intensity of diarrhoea in patients prescribed antibiotics or H. pylori eradication therapy, and have favourable safety. Statements with 70%-100% agreement and "moderate" evidence indicated that, in some patients with IBS, specific probiotics help reduce bloating/distension and improve bowel movement frequency/consistency. CONCLUSIONS This updated review indicates that specific probiotics are beneficial in certain lower GI problems, although many of the new publications did not report benefits of probiotics, possibly due to inclusion of new, less efficacious preparations. Specific probiotics can relieve lower GI symptoms in IBS, prevent diarrhoea associated with antibiotics and H. pylori eradication therapy, and show favourable safety. This study will help clinicians recommend/prescribe probiotics for specific symptoms.
Collapse
|
57
|
Ren C, Dokter‐Fokkens J, Figueroa Lozano S, Zhang Q, de Haan BJ, Zhang H, Faas MM, de Vos P. Lactic Acid Bacteria May Impact Intestinal Barrier Function by Modulating Goblet Cells. Mol Nutr Food Res 2018; 62:e1700572. [PMID: 29333697 PMCID: PMC5900975 DOI: 10.1002/mnfr.201700572] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/11/2017] [Indexed: 12/19/2022]
Abstract
SCOPE Lactic acid bacteria (LAB) are recognized to promote gastrointestinal health by mechanisms that are not fully understood. LABs might modulate the mucus and thereby enhance intestinal barrier function. Herein, we investigate effects of different LAB strains and species on goblet cell genes involved in mucus synthesis. METHODS AND RESULTS Gene expression profiles of goblet-cell-associated products (mucin MUC2, trefoil factor 3, resistin-like molecule β, carbohydrate sulfotransferase 5, and galactose-3-O-sulfotransferase 2) induced by LAB or their derived conditioned medium in human goblet cell line LS174T are studied. Effects of LAB on gene transcription are assessed with or without exposure to TNF-α, IL-13, or the mucus damaging agent tunicamycin. LAB do impact the related genes in a species- and strain-specific fashion and their effects are different in the presence of the cytokines and tunicamycin. Bioactive factors secreted by some strains are also found to regulate goblet cell-related genes. CONCLUSION Our findings provide novel insights in differences in modulatory efficacy on mucus genes between LAB species and strains. This study further unravels direct interactions between LAB and intestinal goblet cells, and highlights the importance of rationally selecting appropriate LAB candidates to achieve specific benefits in the gut.
Collapse
Affiliation(s)
- Chengcheng Ren
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Jelleke Dokter‐Fokkens
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Susana Figueroa Lozano
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Qiuxiang Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Bart J. de Haan
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Hao Zhang
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Marijke M. Faas
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| | - Paul de Vos
- ImmunoendocrinologyDivision of Medical BiologyDepartment of Pathology and Medical BiologyUniversity of Groningen and University Medical Center GroningenHanzeplein 1The Netherlands
| |
Collapse
|
58
|
Abstract
With the advent of the scientific realization that the microbiota of the gastrointestinal tract was more than the cells that exist in the body, the full importance of prebiotics and probiotics has come forth. The importance has been stressed and is available in the new textbook entitled, "The Microbiota in Gastrointestinal Pathophysiology: Implication for Human Health, Prebiotics, Probiotics and Dysbiosis." There is enough evidence now published in the literature so that the scientific world now believes that prebiotics and probiotics are important in gastrointestinal disease.
Collapse
Affiliation(s)
- Martin H Floch
- Section of Digestive Diseases, Yale University School of Medicine, 333 Cedar Street, 1089 LMP, New Haven, CT 06850, USA.
| |
Collapse
|
59
|
A Systematic Review of Probiotic Interventions for Gastrointestinal Symptoms and Irritable Bowel Syndrome in Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME). Probiotics Antimicrob Proteins 2018; 10:466-477. [DOI: 10.1007/s12602-018-9397-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
60
|
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. [PMID: 29445178 PMCID: PMC5813237 DOI: 10.1038/s41598-018-21241-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/29/2018] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common disorder in gastrointestinal system and impairs the quality of life of the patients. Clostridium butyricum (CB) is a probiotics that has been used in several gastrointestinal diseases. The efficacy of CB in treating IBS is still unknown. This prospective, multi-centre, randomized, double-blind, placebo-controlled trial aimed to assess the efficacy and safety of CB in treating diarrhea-predominant IBS (IBS-D) and analyze the fecal microbiota after treatment. Two hundred patients with IBS-D were recruited and were given CB or placebo for 4 weeks. End points included change from baseline in IBS symptoms, quality of life, stool consistency and frequency. Compared with placebo, CB is effective in improving the overall IBS-D symptoms (-62.12 ± 74.00 vs. -40.74 ± 63.67, P = 0.038) as well as quality of life (7.232 ± 14.06 vs. 3.159 ± 11.73, P = 0.032) and stool frequency (-1.602 ± 1.416 vs. -1.086 ± 1.644, P = 0.035). The responder rates are found higher in CB compared with the placebo (44.76% vs. 30.53%, P = 0.042). The change in fecal microbiota was analyzed and function pathways of CB in treating IBS-D were predicted. In conclusion, CB improves overall symptoms, quality of life and stool frequency in IBS-D patients and is considered to be used as a probiotics in treating IBS-D clinically.
Collapse
|
61
|
Harper A, Naghibi MM, Garcha D. The Role of Bacteria, Probiotics and Diet in Irritable Bowel Syndrome. Foods 2018; 7:E13. [PMID: 29373532 PMCID: PMC5848117 DOI: 10.3390/foods7020013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome is a highly prevalent gastrointestinal disorder that threatens the quality of life of millions and poses a substantial financial burden on healthcare systems around the world. Intense research into the human microbiome has led to fascinating discoveries which directly and indirectly implicate the diversity and function of this occult organ in irritable bowel syndrome (IBS) pathophysiology. The benefit of manipulating the gastrointestinal microbiota with diet and probiotics to improve symptoms has been demonstrated in a wealth of both animal and human studies. The positive and negative mechanistic roles bacteria play in IBS will be explored and practical probiotic and dietary choices offered.
Collapse
Affiliation(s)
- Ashton Harper
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| | - Malwina M Naghibi
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| | - Davinder Garcha
- Protexin, Medical Affairs, Probiotics International Ltd., Lopen Head, Somerset TA13 5JH, UK.
| |
Collapse
|
62
|
Mokkala K, Pussinen P, Houttu N, Koivuniemi E, Vahlberg T, Laitinen K. The impact of probiotics and n-3 long-chain polyunsaturated fatty acids on intestinal permeability in pregnancy: a randomised clinical trial. Benef Microbes 2018; 9:199-208. [PMID: 29345158 DOI: 10.3920/bm2017.0072] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A disruption in intestinal barrier integrity may predispose individuals to metabolic aberrations, particularly during the vulnerable period of pregnancy. We investigated whether intestinal permeability, as measured by serum zonulin concentration, changes over the duration of pregnancy and whether this change is reflected in lipopolysaccharide (LPS) activity. Second, we tested in a randomised double-blind placebo controlled clinical trial the impact of consuming dietary probiotics and/or long chain polyunsaturated fatty acid (LC-PUFA) supplements in lowering serum zonulin concentration and LPS activity. The probiotic supplement was a combination of two bacteria, Bifidobacterium animalis ssp. lactis 420 and Lactobacillus rhamnosus HN001. This study included 200 overweight pregnant women participating in an on-going study; participants were randomised to consume either (1) probiotics, (2) LC-PUFA, (3) probiotics and LC-PUFA, or (4) placebo for each supplement. Blood samples were obtained at early, the baseline, and late pregnancy (mean 14 and 35 weeks of gestation, respectively). Serum zonulin concentration increased from early (mean (standard deviation): 62.7 (12.9) ng/ml) to late pregnancy by 5.3 (95%CI 3.7-6.9) ng/ml, and LPS activity increased from (0.16 (0.04) EU/ml) by 0.04 (95%CI 0.03-0.05) EU/ml. No differences among the intervention groups were detected in the change from early to late pregnancy in serum zonulin concentration (P=0.8) or LPS activity (P=0.2). The change in serum zonulin concentration during the pregnancy was associated with the weeks of follow up (r=0.25, P<0.001). Serum LPS activity was correlated with higher maternal weight gain (r=0.19, P=0.008). As a conclusion, intestinal permeability increased with the progression of pregnancy in overweight and obese women and was reflected in LPS activity. No efficacy of supplementation with probiotics and/or LC-PUFA was demonstrated in pregnancy-induced changes in serum zonulin concentration or LPS activity.
Collapse
Affiliation(s)
- K Mokkala
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| | - P Pussinen
- 2 Oral and Maxillofacial Diseases, University of Helsinki and University Hospital of Helsinki, 00014 Helsinki, Finland
| | - N Houttu
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| | - E Koivuniemi
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland.,3 Turku University Hospital, Department of Obstetrics and Gynaecology, 20014 Turku, Finland
| | - T Vahlberg
- 4 Department of Clinical Medicine, Biostatistics, University of Turku and Turku University Hospital, 20014 Turku, Finland
| | - K Laitinen
- 1 Institute of Biomedicine, University of Turku, 20014 Turku, Finland
| |
Collapse
|
63
|
Principi N, Cozzali R, Farinelli E, Brusaferro A, Esposito S. Gut dysbiosis and irritable bowel syndrome: The potential role of probiotics. J Infect 2017; 76:111-120. [PMID: 29291933 DOI: 10.1016/j.jinf.2017.12.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To discuss the role of gut dysbiosis in the development of irritable bowel syndrome (IBS) and the impact of probiotics as a potential therapeutic measure. METHODS PubMed was used to search for all of the studies published over the last 15 years using the key words: "irritable bowel syndrome" and "gut dysbiosis" or "probiotic". More than 800 articles were found, but only those published in English or providing evidence-based data were included in the evaluation. RESULTS IBS is a common disease for which no resolutive therapy is presently available. In recent years, strong evidence of a possible relationship between modifications of the gut microbiota composition and development of IBS has been collected. Moreover, the evidence showed that attempts to treat acute infectious and post-antibiotic gastroenteritis with some probiotics were significantly effective in a great number of patients, leading many experts to suggest the use of probiotics to address all of the clinical problems associated with IBS. CONCLUSION The available data are promising, but presently, a precise definition of which probiotic or which mixture of probiotics is effective cannot be made. Moreover, the dose and duration of treatment has not been established. Finally, we do not know whether probiotic treatment should be different according to the type of IBS. Further studies are needed before probiotics can be considered a reliable treatment for IBS.
Collapse
Affiliation(s)
| | - Rita Cozzali
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Edoardo Farinelli
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Andrea Brusaferro
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy
| | - Susanna Esposito
- Pediatric Clinic, Department of Surgical and Biomedical Sciences, Università degli Studi di Perugia, Perugia, Italy.
| |
Collapse
|
64
|
Low-fat yogurt consumption reduces biomarkers of chronic inflammation and inhibits markers of endotoxin exposure in healthy premenopausal women: a randomised controlled trial. Br J Nutr 2017; 118:1043-1051. [DOI: 10.1017/s0007114517003038] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AbstractThe anti-inflammatory mechanisms of low-fat dairy product consumption are largely unknown. The objective of this study was to determine whether low-fat yogurt reduces biomarkers of chronic inflammation and endotoxin exposure in women. Premenopausal women (BMI 18·5–27 and 30–40 kg/m2) were randomised to consume 339 g of low-fat yogurt (yogurt non-obese (YN); yogurt obese (YO)) or 324 g of soya pudding (control non-obese; control obese (CO)) daily for 9 weeks (n 30/group). Fasting blood samples were analysed for IL-6, TNF-α/soluble TNF II (sTNF-RII), high-sensitivity C-reactive protein, 2-arachidonoyl glycerol, anandamide, monocyte gene expression, soluble CD14 (sCD14), lipopolysaccharide (LPS), LPS binding protein (LBP), IgM endotoxin-core antibody (IgM EndoCAb), and zonulin. BMI, waist circumference and blood pressure were also determined. After 9-week yogurt consumption, YO and YN had decreased TNF-α/sTNFR-RII. Yogurt consumption increased plasma IgM EndoCAb regardless of obesity status. sCD14 was not affected by diet, but LBP/sCD14 was lowered by yogurt consumption in both YN and YO. Yogurt intervention increased plasma 2-arachidonoylglycerol in YO but not YN. YO peripheral blood mononuclear cells expression of NF-κB inhibitor α and transforming growth factor β1 increased relative to CO at 9 weeks. Other biomarkers were unchanged by diet. CO and YO gained approximately 0·9 kg in body weight. YO had 3·6 % lower diastolic blood pressure at week 3. Low-fat yogurt for 9 weeks reduced biomarkers of chronic inflammation and endotoxin exposure in premenopausal women compared with a non-dairy control food. This trial was registered as NCT01686204.
Collapse
|
65
|
Sinagra E, Morreale GC, Mohammadian G, Fusco G, Guarnotta V, Tomasello G, Cappello F, Rossi F, Amvrosiadis G, Raimondo D. New therapeutic perspectives in irritable bowel syndrome: Targeting low-grade inflammation, immuno-neuroendocrine axis, motility, secretion and beyond. World J Gastroenterol 2017; 23:6593-6627. [PMID: 29085207 PMCID: PMC5643283 DOI: 10.3748/wjg.v23.i36.6593] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/15/2017] [Accepted: 07/04/2017] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic, recurring, and remitting functional disorder of the gastrointestinal tract characterized by abdominal pain, distention, and changes in bowel habits. Although there are several drugs for IBS, effective and approved treatments for one or more of the symptoms for various IBS subtypes are needed. Improved understanding of pathophysiological mechanisms such as the role of impaired bile acid metabolism, neurohormonal regulation, immune dysfunction, the epithelial barrier and the secretory properties of the gut has led to advancements in the treatment of IBS. With regards to therapies for restoring intestinal permeability, multiple studies with prebiotics and probiotics are ongoing, even if to date their efficacy has been limited. In parallel, much progress has been made in targeting low-grade inflammation, especially through the introduction of drugs such as mesalazine and rifaximin, even if a better knowledge of the mechanisms underlying the low-grade inflammation in IBS may allow the design of clinical trials that test the efficacy and safety of such drugs. This literature review aims to summarize the findings related to new and investigational therapeutic agents for IBS, most recently developed in preclinical as well as Phase 1 and Phase 2 clinical studies.
Collapse
Affiliation(s)
- Emanuele Sinagra
- Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy
- Euro-Mediterranean Institute of Science and Technology, 90100 Palermo, Italy
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90100 Palermo, Italy
| | | | - Ghazaleh Mohammadian
- Department of Medicine, Division of Gastroenterology and Hepatology, Karolinska Institutet, Karolinska University Hospital, Huddinge, 17176 Stockholm, Sweden
| | - Giorgio Fusco
- Unit of Internal Medicine, Ospedali Riuniti Villa Sofia-Vincenzo Cervello, 90100 Palermo, Italy
| | - Valentina Guarnotta
- Section of Cardio-Respiratory and Endocrine-Metabolic Diseases, Biomedical Department of Internal and Specialist Medicine, University of Palermo, Palermo 90127, Italy
| | - Giovanni Tomasello
- Euro-Mediterranean Institute of Science and Technology, 90100 Palermo, Italy
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90100 Palermo, Italy
| | - Francesco Cappello
- Euro-Mediterranean Institute of Science and Technology, 90100 Palermo, Italy
- Department of Experimental Biomedicine and Clinical Neuroscience, Section of Human Anatomy, University of Palermo, 90100 Palermo, Italy
| | - Francesca Rossi
- Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy
| | - Georgios Amvrosiadis
- Unit of Gastroenterology, Ospedali Riuniti Villa Sofia-Vincenzo Cervello, 90100 Palermo, Italy
| | - Dario Raimondo
- Gastroenterology and Endoscopy Unit, Fondazione Istituto Giuseppe Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy
| |
Collapse
|
66
|
Bailey J, Vince V, Williams N, Cogan T. Streptococcus thermophilus NCIMB 41856 ameliorates signs of colitis in an animal model of inflammatory bowel disease. Benef Microbes 2017; 8:605-614. [DOI: 10.3920/bm2016.0110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Treatment of inflammatory bowel disease (IBD) is mainly based on suppression of symptoms, often with numerous side effects. Trials of probiotics in IBD have frequently produced disappointing results. The majority of probiotics are unusual, since they do not require iron for growth, unlike many bacteria resident in the intestine. The IBD intestine is iron-rich due to bleeding and use of oral iron supplements; conventional probiotics would be rapidly outcompeted. We have evaluated an iron-responsive Streptococcus thermophilus strain for its potential to reduce signs of colitis. Efficacy of S. thermophilus was evaluated in the dextran sodium sulphate mouse model of colitis. Treated animals were given 1×108 cfu S. thermophilus per day and clinical observations were taken daily. At termination, gross and histopathological signs of disease, cellular infiltration, location of bacteria, and cytokine expression in the intestine were determined. S. thermophilus delayed onset of colitis and reduced clinical signs of disease, including bodyweight loss and gastrointestinal bleeding. It reduced bacterial translocation into the colonic tissue. Increased numbers of CD8+ intraepithelial lymphocytes were seen in control animals treated with S. thermophilus. S. thermophilus had no effect on gross pathology, histopathology or cytokine production in either colitic or control animals. We propose that S. thermophilus promotes maintenance of mucosal barrier function which reduces bacterial translocation, thereby reducing immune stimulation and associated inflammation. This allows mucosal healing, reducing gastrointestinal bleeding and weight loss. This could be studied as a locally-acting adjunct or alternative to current IBD treatments.
Collapse
Affiliation(s)
- J.R. Bailey
- Mucosal Microbiology, School of Veterinary Sciences, University of Bristol, BS40 5DU, United Kingdom
| | - V. Vince
- KWS BioTest Ltd., Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - N.A. Williams
- KWS BioTest Ltd., Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - T.A. Cogan
- Mucosal Microbiology, School of Veterinary Sciences, University of Bristol, BS40 5DU, United Kingdom
| |
Collapse
|
67
|
Hod K, Sperber AD, Ron Y, Boaz M, Dickman R, Berliner S, Halpern Z, Maharshak N, Dekel R. A double-blind, placebo-controlled study to assess the effect of a probiotic mixture on symptoms and inflammatory markers in women with diarrhea-predominant IBS. Neurogastroenterol Motil 2017; 29. [PMID: 28271623 DOI: 10.1111/nmo.13037] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Micro-inflammation is considered an element in the pathogenesis of irritable bowel syndrome (IBS). High-sensitivity C reactive protein (hs-CRP) was previously shown to be higher in IBS compared to healthy controls, albeit within the normal range. Since probiotics may suppress micro-inflammation in the gut, we tested if they reduce symptoms and inflammatory markers (hs-CRP and fecal calprotectin (FC) in diarrhea-predominant IBS (IBS-D). The aim of this study was to assess the clinical and laboratory effects of BIO-25, a multispecies probiotic, in women with IBS-D. METHODS A double-blind, placebo-controlled study. Following a 2-week run-in, eligible women were assigned at random to a probiotic capsule or an indistinguishable placebo, twice daily for 8 weeks. IBS symptoms and stool consistency were rated daily by Visual Analogue Scales (VAS) and the Bristol Stool Scale (BSS). High-sensitivity C reactive protein was tested at baseline, 4 and 8 weeks. FC was tested at baseline and 8 weeks. KEY RESULTS One hundred and seventy-two IBS-D patients were recruited and 107 eligible patients were allocated to the intervention (n=54) or placebo (n=53) group. All symptoms improved in both groups with no significant difference between them in symptom improvement, hs-CRP or FC levels. CONCLUSIONS & INFERENCES An 8-week treatment with BIO-25 improved symptoms in women with IBS-D, but was not superior to placebo. This rigorously designed and executed study supports the findings of other studies that did not demonstrate superiority of probiotics over placebo in IBS. High quality clinical studies are necessary to examine the efficacy of other specific probiotics in IBS-D patients since data are still conflicting.
Collapse
Affiliation(s)
- K Hod
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Research Division, Epidemiology Service, Assuta Medical Centers, Tel Aviv, Israel
| | - A D Sperber
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Y Ron
- Department of Gastroenterology and Liver Diseases, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv, Israel
| | - M Boaz
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - R Dickman
- Department of Gastroenterology and Liver Diseases, Rabin Medical Center, Belinson Hospital, Petach Tikva, Israel
| | - S Berliner
- Department of Medicine E, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Z Halpern
- Department of Gastroenterology and Liver Diseases, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv, Israel
| | - N Maharshak
- Department of Gastroenterology and Liver Diseases, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv, Israel
| | - R Dekel
- Department of Gastroenterology and Liver Diseases, Tel-Aviv Sourasky Medical Center, Affiliated to the Sackler Faculty of Medicine, Tel Aviv, Israel
| |
Collapse
|
68
|
Fu L, Wang C, Wang Y. Seafood allergen-induced hypersensitivity at the microbiota-mucosal site: Implications for prospective probiotic use in allergic response regulation. Crit Rev Food Sci Nutr 2017; 58:1512-1525. [DOI: 10.1080/10408398.2016.1269719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Linglin Fu
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Chong Wang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Yanbo Wang
- Key Laboratory for Food Microbial Technology of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| |
Collapse
|
69
|
Probiotic characteristics of Bacillus strains isolated from Korean traditional soy sauce. Lebensm Wiss Technol 2017. [DOI: 10.1016/j.lwt.2016.08.040] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
70
|
de Roos NM, van Hemert S, Rovers JMP, Smits MG, Witteman BJM. The effects of a multispecies probiotic on migraine and markers of intestinal permeability-results of a randomized placebo-controlled study. Eur J Clin Nutr 2017; 71:1455-1462. [PMID: 28537581 DOI: 10.1038/ejcn.2017.57] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/15/2017] [Accepted: 03/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES Migraine, associated with several gastrointestinal disorders, may result from increased intestinal permeability, allowing endotoxins to enter the bloodstream. We tested whether probiotics could reduce migraine through an effect on intestinal permeability and inflammation. SUBJECTS/METHODS In total, 63 patients were randomly allocated to the probiotic (n=31) or the placebo group (n=32). Participants ingested a multispecies probiotic (5x109 colony-forming units) or placebo daily for 12 weeks. Migraine was assessed with the Migraine Disability Assessment Scale (MIDAS), the Headache Disability Inventory (HDI) and headache diaries. At baseline and 12 weeks, intestinal permeability was measured with the urinary lactulose/mannitol test and fecal and serum zonulin; inflammation was measured from interleukin (IL) -6, IL-10, tumor necrosis factor-α and C-reactive protein in serum. RESULTS The MIDAS migraine intensity score significantly decreased in both groups (P<0.001) and the HDI score significantly decreased in the probiotic group (P=0.032) and borderline in the placebo group (P=0.053). In the probiotics group, patients had a median of 6 migraine days in the first month, 4 in the second month (P=0.002) and 5 in the last month, which was not significantly different from the 5, 4, and 4 days in the placebo group. A ⩾2day reduction in migraine days was seen in 12/31 patients in the probiotics group versus 7/29 in the placebo group (ns). Probiotic use did not significantly affect medication use, intestinal permeability or inflammation compared to placebo. CONCLUSIONS In this study, we could not confirm significant benefit from a multispecies probiotic compared to a placebo on the outcome parameters of migraine and intestinal integrity.
Collapse
Affiliation(s)
- N M de Roos
- Wageningen UR, Division Human Nutrition and Epidemiology, Wageningen University, Wageningen, The Netherlands
| | - S van Hemert
- Winclove b.v., Innovation Department, Amsterdam, The Netherlands
| | - J M P Rovers
- Hospital Gelderse Vallei, Department of Neurology, Ede, The Netherlands
| | - M G Smits
- Hospital Gelderse Vallei, Department of Neurology, Ede, The Netherlands
| | - B J M Witteman
- Wageningen UR, Division Human Nutrition and Epidemiology, Wageningen University, Wageningen, The Netherlands.,Hospital Gelderse Vallei, Department of Gastroenterology and Hepatology, Ede, The Netherlands
| |
Collapse
|
71
|
The effects of Lactobacillus plantarum on small intestinal barrier function and mucosal gene transcription; a randomized double-blind placebo controlled trial. Sci Rep 2017; 7:40128. [PMID: 28045137 PMCID: PMC5206730 DOI: 10.1038/srep40128] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/02/2016] [Indexed: 02/08/2023] Open
Abstract
The aim of this study was to investigate the effects of three Lactobacillus plantarum strains on in-vivo small intestinal barrier function and gut mucosal gene transcription in human subjects. The strains were selected for their differential effects on TLR signalling and tight junction protein rearrangement, which may lead to beneficial effects in a stressed human gut mucosa. Ten healthy volunteers participated in four different intervention periods: 7-day oral intake of either L. plantarum WCFS1, CIP104448, TIFN101 or placebo, proceeded by a 4 weeks wash-out period. Lactulose-rhamnose ratio (an indicator of small intestinal permeability) increased after intake of indomethacin, which was given as an artificial stressor of the gut mucosal barrier (mean ratio 0.06 ± 0.04 to 0.10 ± 0.06, p = 0.001), but was not significantly affected by the bacterial interventions. However, analysis in small intestinal biopsies, obtained by gastroduodenoscopy, demonstrated that particularly L. plantarum TIFN101 modulated gene transcription pathways related to cell-cell adhesion with high turnover of genes involved in tight- and adhesion junction protein synthesis and degradation (e.g. actinin alpha-4, metalloproteinase-2). These effects were less pronounced for L. plantarum WCFS1 and CIP104448. In conclusion, L. plantarum TIFN101 induced the most pronounced probiotic properties with specific gene transcriptional effects on repair processes in the compromised intestine of healthy subjects.
Collapse
|
72
|
González-Castro AM, Martínez C, Salvo-Romero E, Fortea M, Pardo-Camacho C, Pérez-Berezo T, Alonso-Cotoner C, Santos J, Vicario M. Mucosal pathobiology and molecular signature of epithelial barrier dysfunction in the small intestine in irritable bowel syndrome. J Gastroenterol Hepatol 2017; 32:53-63. [PMID: 27087165 DOI: 10.1111/jgh.13417] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/16/2022]
Abstract
Irritable bowel syndrome (IBS) is one of the most prevalent gastrointestinal disorders in developed countries. Its etiology remains unknown; however, a common finding, regardless of IBS subtype, is the presence of altered intestinal barrier. In fact, signaling and location of cell-to-cell adhesion proteins, in connection with increased immune activity, seem abnormal in the intestinal epithelium of IBS patients. Despite that most research is performed on distal segments of the intestine, altered permeability has been reported in both, the small and the large bowel of all IBS subtypes. The small intestine carries out digestion and nutrient absorption and is also the site where the majority of immune responses to luminal antigens takes place. In fact, the upper intestine is more exposed to environmental antigens than the colon and is also a site of symptom generation. Recent studies have revealed small intestinal structural alterations of the epithelial barrier and mucosal immune activation in association with intestinal dysfunction, suggesting the commitment of the intestine as a whole in the pathogenesis of IBS. This review summarizes the most recent findings on mucosal barrier alterations and its relationship to symptoms arising from the small intestine in IBS, including epithelial structural abnormalities, mucosal immune activation, and microbial dysbiosis, further supporting the hypothesis of an organic origin of IBS.
Collapse
Affiliation(s)
- Ana M González-Castro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Martínez
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Department of Human Molecular Genetics, Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Eloísa Salvo-Romero
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Marina Fortea
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Pardo-Camacho
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Teresa Pérez-Berezo
- Inserm, U1043, Toulouse, France.,Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - María Vicario
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive Diseases Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitario Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| |
Collapse
|
73
|
Park S, Ji Y, Jung HY, Park H, Kang J, Choi SH, Shin H, Hyun CK, Kim KT, Holzapfel WH. Lactobacillus plantarum HAC01 regulates gut microbiota and adipose tissue accumulation in a diet-induced obesity murine model. Appl Microbiol Biotechnol 2016; 101:1605-1614. [PMID: 27858139 DOI: 10.1007/s00253-016-7953-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/14/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022]
Abstract
The functional features of Lactobacillus plantarum HAC01 (HAC01), isolated from fermented Korean kimchi, were studied with regard to the fat mass, immunometabolic biomarkers and dysbiosis in a diet-induced obesity (DIO) murine model. L. rhamnosus GG (LGG) served as reference strain and a PBS-treated group as control. The administration of L. plantarum HAC01 resulted in reduction of the mesenteric adipose depot, the conjunctive tissue closely associated with the gastrointestinal tract, where lipid oxidative gene expression was upregulated compared to the control group. Metagenome analysis of intestinal microbiota showed that both strains HAC01 and LGG influenced specific bacterial families such as the Lachnospiraceae and Ruminococcaceae rather than the phyla Firmicutes and Bacteroidetes as a whole. The relative abundance of the Lachnospiraceae (phylum Firmicutes) was significantly higher in both LAB-treated groups than in the control. Comparing the impact of the two Lactobacillus strains on microbial composition in the gut also suggests strain-specific effects. The study emphasises the need for deeper studies into functional specificity of a probiotic organism at the strain level. Alleviation of obesity-associated dysbiosis by modulation of the gut microbiota appears to be associated with "indicator" bacterial taxa such as the family Lachnospiraceae. This may provide further insight into mechanisms basic to the mode of probiotic action against obesity and associated dysbiosis.
Collapse
Affiliation(s)
- Soyoung Park
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, 558 Handong-Ro, 3, Heunghae-eup, Buk-gu, Pohang, Gyungbuk, 37673, South Korea
- R&D Center, Pohang, Gyungbuk, 37668, South Korea
| | - Yosep Ji
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, 558 Handong-Ro, 3, Heunghae-eup, Buk-gu, Pohang, Gyungbuk, 37673, South Korea
| | - Hoe-Yune Jung
- R&D Center, Pohang, Gyungbuk, 37668, South Korea
- Department of Life Science, Division of Integrative Biosciences and Biotechnology, POSTECH, Pohang, Gyungbuk, 37673, South Korea
| | - Hyunjoon Park
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, 558 Handong-Ro, 3, Heunghae-eup, Buk-gu, Pohang, Gyungbuk, 37673, South Korea
| | - Jihee Kang
- AtoGen Co. Ltd., 11-8 Techno 1-ro, Yuseong-gu, Daejeon, 34015, South Korea
| | - Sang-Haeng Choi
- AtoGen Co. Ltd., 11-8 Techno 1-ro, Yuseong-gu, Daejeon, 34015, South Korea
| | - Heuynkil Shin
- School of Life Sciences, Handong Global University, Pohang, Gyungbuk, 791-708, South Korea
| | - Chang-Kee Hyun
- School of Life Sciences, Handong Global University, Pohang, Gyungbuk, 791-708, South Korea
| | - Kyong-Tai Kim
- Department of Life Science, Division of Integrative Biosciences and Biotechnology, POSTECH, Pohang, Gyungbuk, 37673, South Korea
| | - Wilhelm H Holzapfel
- Advanced Green Energy and Environment Institute (AGEE), Handong Global University, 558 Handong-Ro, 3, Heunghae-eup, Buk-gu, Pohang, Gyungbuk, 37673, South Korea.
| |
Collapse
|
74
|
Gallo A, Passaro G, Gasbarrini A, Landolfi R, Montalto M. Modulation of microbiota as treatment for intestinal inflammatory disorders: An uptodate. World J Gastroenterol 2016; 22:7186-202. [PMID: 27621567 PMCID: PMC4997632 DOI: 10.3748/wjg.v22.i32.7186] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/23/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023] Open
Abstract
Alterations of intestinal microflora may significantly contribute to the pathogenesis of different inflammatory and autoimmune disorders. There is emerging interest on the role of selective modulation of microflora in inducing benefits in inflammatory intestinal disorders, by as probiotics, prebiotics, synbiotics, antibiotics, and fecal microbiota transplantation (FMT). To summarize recent evidences on microflora modulation in main intestinal inflammatory disorders, PubMed was searched using terms microbiota, intestinal flora, probiotics, prebiotics, fecal transplantation. More than three hundred articles published up to 2015 were selected and reviewed. Randomized placebo-controlled trials and meta-analysis were firstly included, mainly for probiotics. A meta-analysis was not performed because of the heterogeneity of these studies. Most of relevant data derived from studies on probiotics, reporting some efficacy in ulcerative colitis and in pouchitis, while disappointing results are available for Crohn's disease. Probiotic supplementation may significantly reduce rates of rotavirus diarrhea. Efficacy of probiotics in NSAID enteropathy and irritable bowel syndrome is still controversial. Finally, FMT has been recently recognized as an efficacious treatment for recurrent Clostridium difficile infection. Modulation of intestinal flora represents a very interesting therapeutic target, although it still deserves some doubts and limitations. Future studies should be encouraged to provide new understanding about its therapeutical role.
Collapse
|
75
|
Marasco G, Di Biase AR, Schiumerini R, Eusebi LH, Iughetti L, Ravaioli F, Scaioli E, Colecchia A, Festi D. Gut Microbiota and Celiac Disease. Dig Dis Sci 2016; 61:1461-72. [PMID: 26725064 DOI: 10.1007/s10620-015-4020-2] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/20/2015] [Indexed: 12/18/2022]
Abstract
Recent evidence regarding celiac disease has increasingly shown the role of innate immunity in triggering the immune response by stimulating the adaptive immune response and by mucosal damage. The interaction between the gut microbiota and the mucosal wall is mediated by the same receptors which can activate innate immunity. Thus, changes in gut microbiota may lead to activation of this inflammatory pathway. This paper is a review of the current knowledge regarding the relationship between celiac disease and gut microbiota. In fact, patients with celiac disease have a reduction in beneficial species and an increase in those potentially pathogenic as compared to healthy subjects. This dysbiosis is reduced, but might still remain, after a gluten-free diet. Thus, gut microbiota could play a significant role in the pathogenesis of celiac disease, as described by studies which link dysbiosis with the inflammatory milieu in celiac patients. The use of probiotics seems to reduce the inflammatory response and restore a normal proportion of beneficial bacteria in the gastrointestinal tract. Additional evidence is needed in order to better understand the role of gut microbiota in the pathogenesis of celiac disease, and the clinical impact and therapeutic use of probiotics in this setting.
Collapse
Affiliation(s)
- Giovanni Marasco
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy.
| | | | - Ramona Schiumerini
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| | - Leonardo Henry Eusebi
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| | - Lorenzo Iughetti
- Department of Pediatrics, University of Modena, 41124, Modena, Italy
| | - Federico Ravaioli
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| | - Eleonora Scaioli
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| | - Antonio Colecchia
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| | - Davide Festi
- Department of Medical and Surgical Science, University of Bologna, 40138, Bologna, Italy
| |
Collapse
|
76
|
Zeng J, Wang CT, Zhang FS, Qi F, Wang SF, Ma S, Wu TJ, Tian H, Tian ZT, Zhang SL, Qu Y, Liu LY, Li YZ, Cui S, Zhao HL, Du QS, Ma Z, Li CH, Li Y, Si M, Chu YF, Meng M, Ren HS, Zhang JC, Jiang JJ, Ding M, Wang YP. Effect of probiotics on the incidence of ventilator-associated pneumonia in critically ill patients: a randomized controlled multicenter trial. Intensive Care Med 2016; 42:1018-28. [PMID: 27043237 DOI: 10.1007/s00134-016-4303-x] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 03/02/2016] [Indexed: 02/06/2023]
Abstract
PURPOSE To evaluate the potential preventive effect of probiotics on ventilator-associated pneumonia (VAP). METHODS This was an open-label, randomized, controlled multicenter trial involving 235 critically ill adult patients who were expected to receive mechanical ventilation for ≥48 h. The patients were randomized to receive (1) a probiotics capsule containing live Bacillus subtilis and Enterococcus faecalis (Medilac-S) 0.5 g three times daily through a nasogastric feeding tube plus standard preventive strategies or (2) standard preventive strategies alone, for a maximum of 14 days. The development of VAP was evaluated daily, and throat swabs and gastric aspirate were cultured at baseline and once or twice weekly thereafter. RESULTS The incidence of microbiologically confirmed VAP in the probiotics group was significantly lower than that in the control patients (36.4 vs. 50.4 %, respectively; P = 0.031). The mean time to develop VAP was significantly longer in the probiotics group than in the control group (10.4 vs. 7.5 days, respectively; P = 0.022). The proportion of patients with acquisition of gastric colonization of potentially pathogenic microorganisms (PPMOs) was lower in the probiotics group (24 %) than the control group (44 %) (P = 0.004). However, the proportion of patients with eradication PPMO colonization on both sites of the oropharynx and stomach were not significantly different between the two groups. The administration of probiotics did not result in any improvement in the incidence of clinically suspected VAP, antimicrobial consumption, duration of mechanical ventilation, mortality and length of hospital stay. CONCLUSION Therapy with the probiotic bacteria B. Subtilis and E. faecalis are an effective and safe means for preventing VAP and the acquisition of PPMO colonization in the stomach.
Collapse
Affiliation(s)
- Juan Zeng
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Chun-Ting Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China.
| | - Fu-Shen Zhang
- Department of Intensive Care Unit, Taian Central Hospital, #29 Longtan Road, Taian, Shandong, People's Republic of China
| | - Feng Qi
- Department of Intensive Care Unit, Taian Central Hospital, #29 Longtan Road, Taian, Shandong, People's Republic of China
| | - Shi-Fu Wang
- Department of Intensive Care Unit, Zibo Central Hospital, #54 Gongqingtuan Xi Road, Zibo, Shandong, People's Republic of China
| | - Shuang Ma
- Department of Intensive Care Unit, Zibo Central Hospital, #54 Gongqingtuan Xi Road, Zibo, Shandong, People's Republic of China
| | - Tie-Jun Wu
- Department of Intensive Care Unit, Liaocheng People's Hospital, #67 Dongchang Xi Road, Liaocheng, Shandong, People's Republic of China
| | - Hui Tian
- Department of Intensive Care Unit, Liaocheng People's Hospital, #67 Dongchang Xi Road, Liaocheng, Shandong, People's Republic of China
| | - Zhao-Tao Tian
- Department of Intensive Care Unit, Jinan Military General Hospital, #25 Shifan Road, Jinan, Shandong, People's Republic of China
| | - Shu-Liu Zhang
- Department of Intensive Care Unit, Jinan Military General Hospital, #25 Shifan Road, Jinan, Shandong, People's Republic of China
| | - Yan Qu
- Department of Intensive Care Unit, Qingdao Municipal Hospital, #5 Donghai Zhong Road, Qingdao, Shandong, People's Republic of China
| | - Lu-Yi Liu
- Department of Intensive Care Unit, Yantai Yuhuangding Hospital, Yuhuangding Dong Road 20#, Yantai, Shandong, People's Republic of China
| | - Yuan-Zhong Li
- Department of Intensive Care Unit, Dalian Central Hospital, #42 Xuegong Street, Dalian, Liaoning, People's Republic of China
| | - Song Cui
- Department of Intensive Care Unit, Dalian Central Hospital, #42 Xuegong Street, Dalian, Liaoning, People's Republic of China
| | - He-Ling Zhao
- Department of Intensive Care Unit, Hebei People's Hospital, #348 Heping Xi Road, Shijiazhuang, Hebei, People's Republic of China
| | - Quan-Sheng Du
- Department of Intensive Care Unit, Hebei People's Hospital, #348 Heping Xi Road, Shijiazhuang, Hebei, People's Republic of China
| | - Zhuang Ma
- Department of Intensive Care Unit, General Hospital of Shenyang Military Region, #83 Wenhua Road, Shenyang, Liaoning, People's Republic of China
| | - Chun-Hua Li
- Department of Intensive Care Unit, General Hospital of Shenyang Military Region, #83 Wenhua Road, Shenyang, Liaoning, People's Republic of China
| | - Yun Li
- Department of Intensive Care Unit, Jinan Central Hospital, #105 Jiefang Road, Jinan, Shandong, People's Republic of China
| | - Min Si
- Department of Intensive Care Unit, Jinan Central Hospital, #105 Jiefang Road, Jinan, Shandong, People's Republic of China
| | - Yu-Feng Chu
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Mei Meng
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Hong-Sheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Ji-Cheng Zhang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Jin-Jiao Jiang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Min Ding
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| | - Yu-Ping Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong University, #324 Jingwu Road, Jinan, Shandong, People's Republic of China
| |
Collapse
|
77
|
Altered gastrointestinal microbiota in irritable bowel syndrome and its modification by diet: probiotics, prebiotics and the low FODMAP diet. Proc Nutr Soc 2016; 75:306-18. [PMID: 26908093 DOI: 10.1017/s0029665116000021] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Irritable bowel syndrome (IBS) is a functional bowel disorder characterised by abdominal pain or discomfort with disordered defecation. This review describes the role of the gastrointestinal (GI) microbiota in the pathogenesis of IBS and how dietary strategies to manage symptoms impact on the microbial community. Evidence suggests a dysbiosis of the luminal and mucosal colonic microbiota in IBS, frequently characterised by a reduction in species of Bifidobacteria which has been associated with worse symptom profile. Probiotic supplementation trials suggest intentional modulation of the GI microbiota may be effective in treating IBS. A smaller number of prebiotic supplementation studies have also demonstrated effectiveness in IBS whilst increasing Bifidobacteria. In contrast, a novel method of managing IBS symptoms is the restriction of short-chain fermentable carbohydrates (low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet). Studies consistently demonstrate clinical effectiveness of the low FODMAP diet in patients with IBS. However, one unintentional consequence of this dietary intervention is its impact on the microbiota. This leads to an interesting paradox; namely, increasing luminal Bifidobacteria through probiotic supplementation is associated with a reduction in IBS symptoms while in direct conflict to this, the low FODMAP diet has clinical efficacy but markedly reduces luminal Bifidobacteria concentration. Given the multifactorial aetiology of IBS, the heterogeneity of symptoms and the complex and diverse nature of the microbiome, it is probable that both interventions are effective in patient subgroups. However combination treatment has never been explored and as such, presents an exciting opportunity for optimising clinical management, whilst preventing potentially deleterious effects on the GI microbiota.
Collapse
|
78
|
Distrutti E, Monaldi L, Ricci P, Fiorucci S. Gut microbiota role in irritable bowel syndrome: New therapeutic strategies. World J Gastroenterol 2016; 22:2219-2241. [PMID: 26900286 PMCID: PMC4734998 DOI: 10.3748/wjg.v22.i7.2219] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 12/05/2015] [Accepted: 12/30/2015] [Indexed: 02/07/2023] Open
Abstract
In the last decade the impressive expansion of our knowledge of the vast microbial community that resides in the human intestine, the gut microbiota, has provided support to the concept that a disturbed intestinal ecology might promote development and maintenance of symptoms in irritable bowel syndrome (IBS). As a correlate, manipulation of gut microbiota represents a new strategy for the treatment of this multifactorial disease. A number of attempts have been made to modulate the gut bacterial composition, following the idea that expansion of bacterial species considered as beneficial (Lactobacilli and Bifidobacteria) associated with the reduction of those considered harmful (Clostridium, Escherichia coli, Salmonella, Shigella and Pseudomonas) should attenuate IBS symptoms. In this conceptual framework, probiotics appear an attractive option in terms of both efficacy and safety, while prebiotics, synbiotics and antibiotics still need confirmation. Fecal transplant is an old treatment translated from the cure of intestinal infective pathologies that has recently gained a new life as therapeutic option for those patients with a disturbed gut ecosystem, but data on IBS are scanty and randomized, placebo-controlled studies are required.
Collapse
|
79
|
Hod K, Ringel Y. Probiotics in functional bowel disorders. Best Pract Res Clin Gastroenterol 2016; 30:89-97. [PMID: 27048900 DOI: 10.1016/j.bpg.2016.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 01/07/2016] [Indexed: 01/31/2023]
Abstract
Functional bowel disorders (FBDs) are the most common gastrointestinal (GI) disorders seen by gastroenterologists and primary care physicians. The disorders affect patients functioning and quality of life (QOL) and are associated with significant healthcare burden. The current theory regarding the development of FBDs suggests brain-gut axis dysfunctions associated abnormal GI motility and sensation. Recent data suggest that alterations in the intestinal microbiota may have a role in the pathogenesis of FBDs; or at least have the potential to affect intestinal functions that are thought to be relevant to the development of functional GI symptoms. This has led to growing interest of healthcare providers and patients in targeting the intestinal microbiota for the treatment of FBDs. In this article we discuss the potential role probiotic interventions in the treatment of FBDs. We review the evidence from pre-clinical and clinical studies and discuss the current recommendations for the use of probiotics for FBDs in clinical practice.
Collapse
Affiliation(s)
- Keren Hod
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehuda Ringel
- Department of Gastroenterology, Rabin Medical Center, Petach Tikva, Israel; Department of Medicine, University of North Carolina School of Medicine at Chapel Hill, NC, USA
| |
Collapse
|
80
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015; 9:392. [PMID: 26528128 PMCID: PMC4604320 DOI: 10.3389/fncel.2015.00392] [Citation(s) in RCA: 649] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022] Open
Abstract
The emerging links between our gut microbiome and the central nervous system (CNS) are regarded as a paradigm shift in neuroscience with possible implications for not only understanding the pathophysiology of stress-related psychiatric disorders, but also their treatment. Thus the gut microbiome and its influence on host barrier function is positioned to be a critical node within the brain-gut axis. Mounting preclinical evidence broadly suggests that the gut microbiota can modulate brain development, function and behavior by immune, endocrine and neural pathways of the brain-gut-microbiota axis. Detailed mechanistic insights explaining these specific interactions are currently underdeveloped. However, the concept that a "leaky gut" may facilitate communication between the microbiota and these key signaling pathways has gained traction. Deficits in intestinal permeability may underpin the chronic low-grade inflammation observed in disorders such as depression and the gut microbiome plays a critical role in regulating intestinal permeability. In this review we will discuss the possible role played by the gut microbiota in maintaining intestinal barrier function and the CNS consequences when it becomes disrupted. We will draw on both clinical and preclinical evidence to support this concept as well as the key features of the gut microbiota which are necessary for normal intestinal barrier function.
Collapse
Affiliation(s)
- John R Kelly
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Paul J Kennedy
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Gerard Clarke
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Niall P Hyland
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Pharmacology and Therapeutics, University College Cork Cork, Ireland
| |
Collapse
|
81
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
82
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
83
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
84
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
85
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
86
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
87
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
88
|
Mazurak N, Broelz E, Storr M, Enck P. Probiotic Therapy of the Irritable Bowel Syndrome: Why Is the Evidence Still Poor and What Can Be Done About It? J Neurogastroenterol Motil 2015; 21:471-85. [PMID: 26351253 PMCID: PMC4622129 DOI: 10.5056/jnm15071] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/22/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND/AIMS Despite numerous randomized clinical trials and meta-analyses, there is no increased evidence for the efficacy of probiotics in the treatment of irritable bowel syndrome (IBS). We review this evidence, identify and analyse the reasons for this lack of evidence and propose methodological improvements for future studies. METHODS Based on a literature search, we identified 56 papers that matched the purpose of our analyses. Twenty-seven studies used multi-species bacterial preparations and 29 used single-strain probiotics. They were analysed regarding patients included, treatment duration, probiotic dosage, and outcome measures. RESULTS Trials in both groups suffered from heterogeneity with respect to probiotic concentration, duration of treatment, and other methodological issues (crossover design and underpowered studies). This heterogeneity did not allow the application of a meta-analytic approach and a systematic review was therefore performed instead. Multi-strain preparations combined 2 to 8 different bacterial subspecies, mostly lactobacilli or bifidobacteria, and used variable lengths of treatments. Overall, more than 50% of trials presented negative outcomes. The majority of the single-strain probiotic trials employing lactobacilli or Saccharomyces were negative, whereas trials employing bifidobacteria showed positive results. CONCLUSIONS The heterogeneity of the studies of probiotics in IBS questions the value of meta-analyses. The use of different bacterial strains and different mixtures of these strains, as well as different dosages, are the main contributors to this heterogeneity. Current data provides limited evidence for the efficacy of a small number of single-strain probiotics in IBS (mostly bifidobacteria) and sound studies following strict trial guidelines (Food and Drug Administration and European Medicines Agency guidelines for clinical trials) are needed. We summarised and proposed some methodological issues for future studies in the field.
Collapse
Affiliation(s)
- Nazar Mazurak
- SymbioGruppe GmbH, Herborn,
Germany
- Department of Psychosomatic Medicine, University Hospital Tübingen,
Germany
| | - Ellen Broelz
- Department of Psychosomatic Medicine, University Hospital Tübingen,
Germany
| | | | - Paul Enck
- Department of Psychosomatic Medicine, University Hospital Tübingen,
Germany
| |
Collapse
|
89
|
Dysbiosis of Intestinal Microbiota Associated With Inflammation Involved in the Progression of Acute Pancreatitis. Pancreas 2015; 44:868-75. [PMID: 25931253 DOI: 10.1097/mpa.0000000000000355] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To evaluate alterations of the intestinal bacteria and its associations with the inflammation in acute pancreatitis (AP). METHODS A multihospital prospective clinical study was conducted, and a total of 108 participants were enrolled in our study, including 44 with severe AP (SAP), 32 with mild AP (MAP), and 32 healthy volunteers. The structure of intestinal microbiota, 10 predominant bacteria, plasma endotoxin, and serum cytokines were investigated by polymerase chain reaction-denaturing gradient gel electrophoresis, real-time quantitative polymerase chain reaction, Limulus amebocyte lysate tests, and enzyme-linked immunosorbent assays, respectively. RESULTS Dramatic alterations in the predominant fecal microbiota were observed in most of both MAP and SAP patients. In addition, the rates of the multiorgan failures and infectious complications in the patients with SAP with altered intestinal microbiota were significantly higher than in those whose intestinal microbiota remained unaltered. Enterococcus increased and Bifidobacterium decreased in the patients with SAP compared to the patients with MAP. Serum IL-6 were positively correlated with Enterobacteriaceae and Enterococcus and negatively correlated with Bifidobacterium, whereas plasma endotoxin positively correlated with Enterococcus (P < 0.05). CONCLUSIONS The intestinal bacteria most frequently altered in both the patients with MAP and those with SAP significantly correlated with inflammation, which indicated that the intestinal microbiota may be involved in the progression of AP.
Collapse
|
90
|
Raftery T, Martineau AR, Greiller CL, Ghosh S, McNamara D, Bennett K, Meddings J, O'Sullivan M. Effects of vitamin D supplementation on intestinal permeability, cathelicidin and disease markers in Crohn's disease: Results from a randomised double-blind placebo-controlled study. United European Gastroenterol J 2015; 3:294-302. [PMID: 26137304 DOI: 10.1177/2050640615572176] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 01/17/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Vitamin D (vitD) supplementation may prolong remission in Crohn's disease (CD); however, the clinical efficacy and mechanisms are unclear. AIM To determine changes in intestinal permeability (IP), antimicrobial peptide (AMP) concentrations and disease markers in CD, in response to vitD supplementation. METHODS In a double-blind randomised placebo-controlled study, we assigned 27 CD patients in remission to 2000 IU/day vitD or placebo for 3 mos. We determined IP, plasma cathelicidin (LL-37 in ng/mL), human-beta-defensin-2 (hBD2 in pg/mL), disease activity (Crohn's Disease Activity Index (CDAI)), C-reactive protein (CRP in mg/L), fecal calprotectin (µg/g), Quality of Life (QoL) and serum 25-hydroxyvitamin D (25(OH)D in nmol/L) at 0 and 3 mos. RESULTS At 3 mos., 25(OH)D concentrations were significantly higher in those whom were treated (p < 0.001). Intra-group analysis showed increased LL-37 concentrations (p = 0.050) and maintenance of IP measures in the treated group. In contrast, in the placebo group, the small bowel (p = 0.018) and gastro-duodenal permeability (p = 0.030) increased from baseline. At 3 mos., patients with 25(OH)D ≥ 75 nmol/L had significantly lower CRP (p = 0.019), higher QoL (p = 0.037), higher LL-37 concentrations (p < 0.001) and non-significantly lower CDAI scores (p = 0.082), compared to those with levels <75 nmol/L. CONCLUSION Short-term treatment with 2000 IU/day vitD significantly increased 25(OH)D levels in CD patients in remission and it was associated with increased LL-37 concentrations and maintenance of IP. Achieving 25(OH)D ≥ 75 nmol/l was accompanied by higher circulating LL-37, higher QoL scores and reduced CRP. Registered at ClinicalTrials.gov (NCT01792388).
Collapse
Affiliation(s)
- Tara Raftery
- Department of Clinical Medicine, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Adrian R Martineau
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Claire L Greiller
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Subrata Ghosh
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Deirdre McNamara
- Department of Clinical Medicine, Trinity Centre for Health Sciences, Tallaght Hospital, Dublin, Ireland
| | - Kathleen Bennett
- Department of Pharmacology and Therapeutics, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| | - Jon Meddings
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Maria O'Sullivan
- Department of Clinical Medicine, Trinity Centre for Health Sciences, St. James's Hospital, Dublin, Ireland
| |
Collapse
|
91
|
Heat-killed yogurt-containing lactic acid bacteria prevent cytokine-induced barrier disruption in human intestinal Caco-2 cells. ANN MICROBIOL 2015. [DOI: 10.1007/s13213-015-1093-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
92
|
Abstract
Irritable bowel syndrome (IBS) is one of the most common gastrointestinal disorders. The pathogenesis of IBS is not fully clear up to now, but numerous studies suggest that it is related to multiple factors, such as abnormal intestinal motility, visceral hypersensitivity, intestinal infection and immunoreaction, and imbalanced intestinal microbiota. The dysbiosis of the gastrointestinal tract microbiota is considered one of the most important factors responsible for the development of IBS. In this article, we will discuss the relationship between intestinal microbiota and IBS.
Collapse
|
93
|
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 PMCID: PMC4356930 DOI: 10.3748/wjg.v21.i10.3072] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/24/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the efficacy of probiotics in irritable bowel syndrome (IBS) patients.
METHODS: PubMed, Cochrane library, Scopus, Google Scholar, and Clinicaltrial.gov databases were searched for literature published between September 2007 and December 2013. The applied Mesh terms were “probiotics,”“irritable bowel syndrome,” and “irritable bowel syndrome treatment.” The collected data contained24 clinical trials, of which 15 were eligible for meta-analysis and nine were reviewed systematically. All studies were randomized placebo-controlled trials in patients with IBS that investigated the efficacy of probiotics in IBS improvement. The Jadad score was used to assess the methodological quality of trials. The quality scale ranges from 0 to 5 points, with a score ≤ 2 indicating a low quality report, and a score of ≥ 3 indicating a high quality report. Relative risk (RR), standardized effect size, and 95%CI were calculated using the DerSimonian-Laird method. The Cochran Q test was used to test heterogeneity with P < 0.05. Funnel plots were constructed and Egger’s and Begg-Mazumdar tests were performed to assess publication bias.
RESULTS: A total of 1793 patients were included in the meta-analysis. The RR of responders to therapies based on abdominal pain score in IBS patients for two included trials comparing probiotics to placebo was 1.96 (95%CI: 1.14-3.36; P = 0.01). RR of responders to therapies based on a global symptom score in IBS patients for two included trials comparing probiotics with placebo was 2.43 (95%CI: 1.13-5.21; P = 0.02). For adequate improvement of general symptoms in IBS patients, the RR of seven included trials (six studies) comparing probiotics with placebo was 2.14 (95%CI: 1.08-4.26; P = 0.03). Distension, bloating, and flatulence were evaluated using an IBS severity scoring system in three trials (two studies) to compare the effect of probiotic therapy in IBS patients with placebo, the standardized effect size of mean differences for probiotics therapy was -2.57 (95%CI: -13.05--7.92).
CONCLUSION: Probiotics reduce pain and symptom severity scores. The results demonstrate the beneficial effects of probiotics in IBS patients in comparison with placebo.
Collapse
|
94
|
Lei M, Dai X, Liu M. Biological Characteristics and Safety Examination of Five Enterococcal Strains from Probiotic Products. J Food Saf 2015. [DOI: 10.1111/jfs.12179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Ming Lei
- College of Life Sciences; China Jiliang University; Hangzhou 310018 China
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection and Quarantine; China Jiliang University; Hangzhou 310018 China
| | - Xianjun Dai
- College of Life Sciences; China Jiliang University; Hangzhou 310018 China
| | - Mingqi Liu
- College of Life Sciences; China Jiliang University; Hangzhou 310018 China
| |
Collapse
|
95
|
Gastrointestinal Tract Commensal Bacteria and Probiotics: Influence on End-Organ Physiology. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2015; 70:1-33. [PMID: 26462363 DOI: 10.1007/978-3-0348-0927-6_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Bacteria represent the earliest form of independent life on this planet. Bacterial development has included cooperative symbiosis with plants (e.g., Leguminosae family and nitrogen fixing bacteria in soil) and animals (e.g., the gut microbiome). It is generally agreed upon that the fusion of two prokaryotes evolutionarily gave rise to the eukaryotic cell in which mitochondria may be envisaged as a genetically functional mosaic, a relic from one of the prokaryotes. This is expressed by the appearance of mitochondria in eukaryotic cells (an alpha-proteobacteria input), a significant endosymbiotic evolutionary event. As such, the evolution of human life has been complexly connected to bacterial activities. Hence, microbial colonization of mammals has been a progressively driven process. The interactions between the human host and the microbiome inhabiting the gastrointestinal tract (GIT) for example, afford the human host the necessary cues for the development of regulated signals that in part are induced by reactive oxygen species (ROS). This regulated activity then promotes immunological tolerance and metabolic regulation and stability, which then helps establish control of local and extraintestinal end-organ (e.g., kidneys) physiology. Pharmacobiotics, the targeted administration of live probiotic cultures, is an advancing area of potential therapeutics, either directly or as adjuvants. Hence the continued scientific understanding of the human microbiome in health and disease may further lead to fine tuning the targeted delivery of probiotics for a therapeutic gain.
Collapse
|
96
|
Tiequn B, Guanqun C, Shuo Z. Therapeutic effects of Lactobacillus in treating irritable bowel syndrome: a meta-analysis. Intern Med 2015; 54:243-9. [PMID: 25748731 DOI: 10.2169/internalmedicine.54.2710] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE As the lack of reliable treatment for irritable bowel syndrome (IBS) prompts interest in the development of new therapies, we aimed to systematically evaluate the effect of Lactobacillus in treating this disease. METHODS We searched MEDLIINE, PubMed, Scopus, Web of Science and the Cochrane Central Register of Controlled Trials for the period from 1966 to August 2013 for double-blind, placebo-controlled trials investigating the efficacy of Lactobacillus treatment in the management of IBS. The studies were screened for inclusion based on randomization, controls and reported measurable outcomes. We used the Jadad score to assess the quality of the articles. The STATA 11.0 and Revman 5.0 software packages were used for the meta-analysis. The STATA 11.0 software program was also used to assess indicators of publication bias according to Begg's and Egger's tests. RESULTS Six randomized, placebo-controlled clinical trials met the criteria and were included in the meta-analysis. The Jadad score of the articles was >3, and three articles were of high quality. We analyzed the heterogeneity of the studies and found no heterogeneity in the meta-analysis. In the forest plot, the diamond was on the right side of the vertical line and did not intersect with the line. The pooled relative risk for clinical improvement with Lactobacillus treatment was 7.69 (95% confidence interval: 2.33-25.43, p=0.0008). For adults, the pooled relative risk for clinical improvement with Lactobacillus treatment was 17.62 (95% confidence interval: 5.12-60.65, p<0.00001). For children, the pooled relative risk for clinical improvement with Lactobacillus treatment was 3.71 (95% confidence interval:1.05-13.11, p=0.04). Using the STATA 10.0 and Revman 5.0 software programs, we confirmed that Lactobacillus exhibited significant efficacy in treating IBS. CONCLUSION Compared with the placebo, Lactobacillus treatment was found to be associated with a significantly higher rate of treatment responders in the overall population with IBS, without any side effects. As to limitations of the analysis, additional research is needed.
Collapse
Affiliation(s)
- Bian Tiequn
- Xiaoshan Traditional Chinese Medicine Hospital, China
| | | | | |
Collapse
|
97
|
Intestinal permeability--a new target for disease prevention and therapy. BMC Gastroenterol 2014; 14:189. [PMID: 25407511 PMCID: PMC4253991 DOI: 10.1186/s12876-014-0189-7] [Citation(s) in RCA: 1087] [Impact Index Per Article: 108.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 10/17/2014] [Indexed: 02/06/2023] Open
Abstract
Data are accumulating that emphasize the important role of the intestinal barrier and intestinal permeability for health and disease. However, these terms are poorly defined, their assessment is a matter of debate, and their clinical significance is not clearly established. In the present review, current knowledge on mucosal barrier and its role in disease prevention and therapy is summarized. First, the relevant terms 'intestinal barrier' and 'intestinal permeability' are defined. Secondly, the key element of the intestinal barrier affecting permeability are described. This barrier represents a huge mucosal surface, where billions of bacteria face the largest immune system of our body. On the one hand, an intact intestinal barrier protects the human organism against invasion of microorganisms and toxins, on the other hand, this barrier must be open to absorb essential fluids and nutrients. Such opposing goals are achieved by a complex anatomical and functional structure the intestinal barrier consists of, the functional status of which is described by 'intestinal permeability'. Third, the regulation of intestinal permeability by diet and bacteria is depicted. In particular, potential barrier disruptors such as hypoperfusion of the gut, infections and toxins, but also selected over-dosed nutrients, drugs, and other lifestyle factors have to be considered. In the fourth part, the means to assess intestinal permeability are presented and critically discussed. The means vary enormously and probably assess different functional components of the barrier. The barrier assessments are further hindered by the natural variability of this functional entity depending on species and genes as well as on diet and other environmental factors. In the final part, we discuss selected diseases associated with increased intestinal permeability such as critically illness, inflammatory bowel diseases, celiac disease, food allergy, irritable bowel syndrome, and--more recently recognized--obesity and metabolic diseases. All these diseases are characterized by inflammation that might be triggered by the translocation of luminal components into the host. In summary, intestinal permeability, which is a feature of intestinal barrier function, is increasingly recognized as being of relevance for health and disease, and therefore, this topic warrants more attention.
Collapse
|
98
|
Assessment of the capability of a gelling complex made of tara gum and the exopolysaccharides produced by the microorganism Streptococcus thermophilus ST10 to prospectively restore the gut physiological barrier: a pilot study. J Clin Gastroenterol 2014; 48 Suppl 1:S56-61. [PMID: 25291130 DOI: 10.1097/mcg.0000000000000254] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Leaky gut, or intestinal permeability, is the phenomenon of the gut wall exhibiting increased absorbency. It is pretty well recognised that an altered or damaged bowel lining or gut wall may result from unbalanced diet, parasites, infection, or medications and that this allows substances such as toxins, microbes, undigested food, or waste to leak through. As a natural consequence, this prompts the body to initiate an immune reaction leading to potentially severe health conditions. Different strategies may be used to improve, at least temporarily, the physiological intestinal barrier. The use of specific beneficial microorganisms, such as lactobacilli and bifidobacteria, has been suggested as an innovative tool to counteract an improper level of intestinal permeability. The association of bacteria with specific gelling agents, such as gums, may represent an improvement since these molecules are able to form hydrophilic gels that distribute uniformly over the inner intestinal surface. This pilot study was undertaken to evaluate intestinal permeability in subjects treated with a gelling complex, an association of tara gum and the microorganism Streptococcus thermophilus ST10 (DSM 25246), which has a well-demonstrated in vitro ability to synthesise and secrete exopolysaccharides (EPSs). METHODS Twenty-five healthy subjects were enrolled in this human intervention, double-blind, placebo-controlled, pilot trial (age between 21 and 57 y, mean 37.7±11.2). Subjects were then randomised into 2 groups: group A (13 subjects) was given an active formulation containing 250 mg of tara gum and 1 billion viable cells of S. thermophilus ST10, whereas group B (12 subjects) was given a placebo formulation. All the subjects participating in the study were directed to take 1 dose per day for 30 consecutive days. The presence and concentration of exopolysaccharides (EPSs) in the faeces was determined at time 0 (d0), after 30 days of treatment (d30), and at the end of the 2-week follow-up period (d45). The monosaccharide composition of EPSs was used to quantify the possible contribution of tara gum to the amount of polysaccharides detected in the faecal material. Intestinal permeability was evaluated at the same time by means of the lactitol/mannitol ratio (small intestine permeability) and sucralose concentration (colonic permeability) in urine specimens sampled after specified times. A statistical comparison was made between the concentration of EPSs, the lactulose/mannitol ratio, and the amount of excreted sucralose in the 2 groups at d0, d30, and d45. RESULTS In the active group, supplementation with S. thermophilus ST10 and tara gum was able to significantly increase the faecal EPSs concentration compared with placebo (from 0.169 mg/g to 0.633 mg/g after 30 d, P<0.001). An interesting decrease in intestinal permeability, both of the small bowel and in the colon, was also recorded. The L/M ratio diminished from 0.021 in the active group to 0.014 and 0.015 after 30 and 45 days, respectively (P=0.045 and P=0.033 compared with placebo). The sucralose concentration decreased from 35.8 mg to 27.9 mg and 29.1 mg (P=0.038 and P=0.026 compared with placebo) at the end of the supplementation period and after the follow-up, respectively. No significant differences were recorded in the placebo after 30 days or at the end of the follow-up. CONCLUSIONS The association of the EPSs produced by S. thermophilus ST10 and tara gum seems capable of significantly improving the intestinal functional barrier in healthy subjects. A wider study in subjects presenting impaired gut permeability would be useful in the future to confirm the positive results from this pilot trial. In any case, our findings are consistent with the parallel increase in exopolysaccharide concentration in the faecal material, thus suggesting the effective ability of the strain used to secrete EPSs in the gut lumen. An innovative approach of this type may be useful in helping to restore the physiological barrier by means of a merely natural and mechanical action.
Collapse
|
99
|
Spiller R. Editorial: abnormal permeability and altered mucosal serotonin metabolism in the irritable bowel syndrome - is there a link? Aliment Pharmacol Ther 2014; 40:1116-8. [PMID: 25280258 DOI: 10.1111/apt.12952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Affiliation(s)
- R Spiller
- Nottingham Digestive Diseases Biomedical Research Unit, Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK.
| |
Collapse
|
100
|
Ford AC, Quigley EMM, Lacy BE, Lembo AJ, Saito YA, Schiller LR, Soffer EE, Spiegel BMR, Moayyedi P. Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: systematic review and meta-analysis. Am J Gastroenterol 2014; 109:1547-61; quiz 1546, 1562. [PMID: 25070051 DOI: 10.1038/ajg.2014.202] [Citation(s) in RCA: 447] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/30/2014] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Irritable bowel syndrome (IBS) and chronic idiopathic constipation (CIC) are functional bowel disorders. Evidence suggests that disturbance in the gastrointestinal microbiota may be implicated in both conditions. We performed a systematic review and meta-analysis to examine the efficacy of prebiotics, probiotics, and synbiotics in IBS and CIC. METHODS MEDLINE, EMBASE, and the Cochrane Controlled Trials Register were searched (up to December 2013). Randomized controlled trials (RCTs) recruiting adults with IBS or CIC, which compared prebiotics, probiotics, or synbiotics with placebo or no therapy, were eligible. Dichotomous symptom data were pooled to obtain a relative risk (RR) of remaining symptomatic after therapy, with a 95% confidence interval (CI). Continuous data were pooled using a standardized or weighted mean difference with a 95% CI. RESULTS The search strategy identified 3,216 citations. Forty-three RCTs were eligible for inclusion. The RR of IBS symptoms persisting with probiotics vs. placebo was 0.79 (95% CI 0.70-0.89). Probiotics had beneficial effects on global IBS, abdominal pain, bloating, and flatulence scores. Data for prebiotics and synbiotics in IBS were sparse. Probiotics appeared to have beneficial effects in CIC (mean increase in number of stools per week=1.49; 95% CI=1.02-1.96), but there were only two RCTs. Synbiotics also appeared beneficial (RR of failure to respond to therapy=0.78; 95% CI 0.67-0.92). Again, trials for prebiotics were few in number, and no definite conclusions could be drawn. CONCLUSIONS Probiotics are effective treatments for IBS, although which individual species and strains are the most beneficial remains unclear. Further evidence is required before the role of prebiotics or synbiotics in IBS is known. The efficacy of all three therapies in CIC is also uncertain.
Collapse
Affiliation(s)
- Alexander C Ford
- 1] Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK [2] Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Leeds, UK
| | - Eamonn M M Quigley
- Division of Gastroenterology and Hepatology, Department of Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - Brian E Lacy
- Dartmouth-Hitchcock Medical Center, Gastroenterology, One Medical Center Drive, Lebanon, New Hampshire, USA
| | - Anthony J Lembo
- The Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Yuri A Saito
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lawrence R Schiller
- Digestive Health Associates of Texas, Baylor University Medical Center, Dallas, Texas, USA
| | - Edy E Soffer
- Division of Gastroenterology at Cedars-Sinai, University of Southern California, Los Angeles, California, USA
| | - Brennan M R Spiegel
- Department of Gastroenterology, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Paul Moayyedi
- Gastroenterology Division, McMaster University, Health Sciences Center, Hamilton, Ontario, Canada
| |
Collapse
|