1
|
Whelan K, Ford AC, Burton-Murray H, Staudacher HM. Dietary management of irritable bowel syndrome: considerations, challenges, and solutions. Lancet Gastroenterol Hepatol 2024; 9:1147-1161. [PMID: 39521003 DOI: 10.1016/s2468-1253(24)00238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 11/16/2024]
Abstract
Diet is a cornerstone in the management of irritable bowel syndrome (IBS). There is evidence of efficacy across the spectrum of dietary management strategies, including some supplements (eg, specific fibres), foods, and whole diets (eg, a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols [known as the low-FODMAP diet]). Whole-diet interventions, in particular those that restrict intake, can be challenging to deliver effectively and safely. Factors to consider include patient demographics, food cost and availability, and the acceptability of dietary management and its impact on food-related quality of life. There is concern regarding a potential role of restrictive whole-diet interventions in eating disorder risk. Optimal approaches to delivering dietary management in the health-care setting are unclear. The aim of this Review is to summarise the clinical evidence for the dietary management of IBS; to discuss the challenges, burdens, and risks of dietary management; and to propose how these challenges, burdens, and risks should be mitigated and minimised in clinical practice.
Collapse
Affiliation(s)
- Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, UK.
| | - Alexander C Ford
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Helen Burton-Murray
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Heidi M Staudacher
- Food and Mood Centre, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
2
|
Whelan K, Alexander M, Gaiani C, Lunken G, Holmes A, Staudacher HM, Theis S, Marco ML. Design and reporting of prebiotic and probiotic clinical trials in the context of diet and the gut microbiome. Nat Microbiol 2024; 9:2785-2794. [PMID: 39478082 DOI: 10.1038/s41564-024-01831-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 09/12/2024] [Indexed: 11/02/2024]
Abstract
Diet is a major determinant of the gastrointestinal microbiome composition and function, yet our understanding of how it impacts the efficacy of prebiotics and probiotics is limited. Here we examine current evidence of dietary influence on prebiotic and probiotic efficacy in human studies, including potential mechanisms. We propose that habitual diet be included as a variable in prebiotic and probiotic intervention studies. This recommendation is based on the potential mechanisms via which diet can affect study outcomes, either directly or through the gut microbiome. We consider the challenges and opportunities of dietary assessment in this context. Lastly, we provide recommendations for the design, conduct and reporting of human clinical trials of prebiotics and probiotics (and other biotic interventions) to account for any effect of diet and nutrition.
Collapse
Affiliation(s)
- Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, UK.
| | - Margaret Alexander
- Department of Medical Microbiology and Immunology, University of Wisconsin, Madison, WI, USA
| | - Claire Gaiani
- Laboratoire d'Ingenierie des Biomolecules, Université de Lorraine, Nancy, France
- Institut Universitaire de France, Paris, France
| | - Genelle Lunken
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - Andrew Holmes
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Heidi M Staudacher
- Food and Mood Centre, IMPACT Institute, Deakin University, Melbourne, Victoria, Australia
| | | | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
3
|
Cheng X, Ren C, Mei X, Jiang Y, Zhou Y. Gut microbiota and irritable bowel syndrome: status and prospect. Front Med (Lausanne) 2024; 11:1429133. [PMID: 39484201 PMCID: PMC11524842 DOI: 10.3389/fmed.2024.1429133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Irritable bowel syndrome (IBS) is a very common gastrointestinal disease that, although not as aggressive as tumors, affects patients' quality of life in different ways. The cause of IBS is still unclear, but more and more studies have shown that the characteristics of the gut microbiota, such as diversity, abundance, and composition, are altered in patients with IBS, compared to the healthy population, which confirms that the gut microbiota plays a crucial role in the development of IBS. This paper aims to identify the commonalities by reviewing a large body of literature. Changes in the characteristics of gut microbiota in patients with different types of IBS are discussed, relevant mechanisms are described, and the treatment modalities of gut microbiota in IBS are summarized. Although there are more clinical trials that have made good progress, more standardized, more generalized, larger-scale, multi-omics clinical studies are what is missing. Overall, gut microbiota plays a crucial role in the development of IBS, and there is even more potential for treating IBS by modulating gut microbiota.
Collapse
Affiliation(s)
- Xinyu Cheng
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Cheng Ren
- Department of Cardiology, The First people’s Hospital of Zhangjiagang, Affiliated Hospital of Soochow University, Medical Center of Soochow University, Zhangjiagang, Jiangsu, China
| | - Xiaofei Mei
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| | - Yufeng Jiang
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| | - Yafeng Zhou
- Department of Cardiology, The Fourth Affiliated Hospital of Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Institute for Hypertension, Soochow University, Suzhou, China
| |
Collapse
|
4
|
Matsuura N, Kanayama M, Watanabe Y, Yamada H, Lili L, Torii A. Effect of Personalized Prebiotic and Probiotic Supplements on the Symptoms of Irritable Bowel Syndrome: An Open-Label, Single-Arm, Multicenter Clinical Trial. Nutrients 2024; 16:3333. [PMID: 39408300 PMCID: PMC11478705 DOI: 10.3390/nu16193333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Prebiotics and probiotics have been reported to improve symptoms of irritable bowel syndrome (IBS). Nevertheless, the effects of prebiotics/probiotics can vary depending on the IBS subtypes. The purpose of this study was to investigate the effects of personalized prebiotic and probiotic supplements based on intestinal microbiota and IBS subtypes in patients. METHODS Patients with diarrhea-type IBS (IBS-D), constipation-type IBS (IBS-C), and mixed-type IBS (IBS-M) were enrolled (n = 40 per group; total: n = 120). Personalized prebiotic and probiotic supplements were determined according to the IBS subtypes and intestinal microbiota. The patients received supplements for 4 weeks. The primary outcome was the change in the IBS-severity scoring system from baseline to week 4. RESULTS The IBS-severity scoring system significantly decreased in all patients (-38.0 [95% confidence interval (CI): -53.6, -22.4]; p < 0.001), in patients with IBS-D (-44.5 [95% CI: -70.6, -18.5]; p = 0.004) and IBS-C (-51.2 [95% CI: -79.4, -22.9]; p = 0.002), but not in those with IBS-M (-20.0 [95% CI: -48.0, 8.1]; p = 0.47). In this study, no serious adverse events were observed that had a causal relationship with the intervention. CONCLUSIONS In conclusion, personalized prebiotic and probiotic supplements selected according to individual intestinal microbiota and IBS subtype may alleviate the severity of IBS symptoms, particularly in patients with IBS-C and IBS-D.
Collapse
Affiliation(s)
- Nozomi Matsuura
- Institute of Health Science, Health Science Business Division, Kirin Holdings Company, Limited, Fujisawa, Kanagawa 251-8555, Japan; (N.M.); (Y.W.)
| | - Masaya Kanayama
- Institute of Health Science, Health Science Business Division, Kirin Holdings Company, Limited, Fujisawa, Kanagawa 251-8555, Japan; (N.M.); (Y.W.)
| | - Yuta Watanabe
- Institute of Health Science, Health Science Business Division, Kirin Holdings Company, Limited, Fujisawa, Kanagawa 251-8555, Japan; (N.M.); (Y.W.)
| | - Hirokazu Yamada
- Soiken Inc., Chiyoda, Tokyo 101-0052, Japan
- EviPRO Co., Ltd., Chiyoda, Tokyo 101-0032, Japan
| | - Loukia Lili
- Thorne HealthTech, Inc., New York, NY 10019, USA
| | - Akira Torii
- Torii Medical Clinic, Setagaya, Tokyo 157-0066, Japan
| |
Collapse
|
5
|
Wu X, Cao Y, Liu Y, Zheng J. A New Strategy for Dietary Nutrition to Improve Intestinal Homeostasis in Diarrheal Irritable Bowel Syndrome: A Perspective on Intestinal Flora and Intestinal Epithelial Interaction. Nutrients 2024; 16:3192. [PMID: 39339792 PMCID: PMC11435304 DOI: 10.3390/nu16183192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/12/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Although a reasonable diet is essential for promoting human health, precise nutritional regulation presents a challenge for different physiological conditions. Irritable Bowel Syndrome (IBS) is characterized by recurrent abdominal pain and abnormal bowel habits, and diarrheal IBS (IBS-D) is the most common, seriously affecting patients' quality of life. Therefore, the implementation of precise nutritional interventions for IBS-D has become an urgent challenge in the fields of nutrition and food science. IBS-D intestinal homeostatic imbalance involves intestinal flora disorganization and impaired intestinal epithelial barrier function. A familiar interaction is evident between intestinal flora and intestinal epithelial cells (IECs), which together maintain intestinal homeostasis and health. Dietary patterns, such as the Mediterranean diet, have been shown to regulate gut flora, which in turn improves the body's health by influencing the immune system, the hormonal system, and other metabolic pathways. METHODS This review summarized the relationship between intestinal flora, IECs, and IBS-D. It analyzed the mechanism behind IBS-D intestinal homeostatic imbalance by examining the interactions between intestinal flora and IECs, and proposed a precise dietary nutrient intervention strategy. RESULTS AND CONCLUSION This increases the understanding of the IBS-D-targeted regulation pathways and provides guidance for designing related nutritional intervention strategies.
Collapse
Affiliation(s)
- Xinyu Wu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Yilong Cao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China; (X.W.); (Y.C.)
| | - Jie Zheng
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
6
|
Deyaert S, Poppe J, Dai Vu L, Baudot A, Bubeck S, Bayne T, Krishnan K, Giusto M, Moltz S, Van den Abbeele P. Functional Muffins Exert Bifidogenic Effects along with Highly Product-Specific Effects on the Human Gut Microbiota Ex Vivo. Metabolites 2024; 14:497. [PMID: 39330504 PMCID: PMC11433953 DOI: 10.3390/metabo14090497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/07/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
GoodBiome™ Foods are functional foods containing a probiotic (Bacillus subtilis HU58™) and prebiotics (mainly inulin). Their effects on the human gut microbiota were assessed using ex vivo SIFR® technology, which has been validated to provide clinically predictive insights. GoodBiome™ Foods (BBM/LCM/OSM) were subjected to oral, gastric, and small intestinal digestion/absorption, after which their impact on the gut microbiome of four adults was assessed (n = 3). All GoodBiome™ Foods boosted health-related SCFA acetate (+13.1/14.1/13.8 mM for BBM/LCM/OSM), propionate (particularly OSM; +7.4/7.5/8.9 mM for BBM/LCM/OSM) and butyrate (particularly BBM; +2.6/2.1/1.4 mM for BBM/LCM/OSM). This is related to the increase in Bifidobacterium species (B. catenulatum, B. adolescentis, B. pseudocatenulatum), Coprococcus catus and Bacteroidetes members (Bacteroides caccae, Phocaeicola dorei, P. massiliensis), likely mediated via inulin. Further, the potent propionogenic potential of OSM related to increased Bacteroidetes members known to ferment oats (s key ingredient of OSM), while the butyrogenic potential of BBM related to a specific increase in Anaerobutyricum hallii, a butyrate producer specialized in the fermentation of erythritol (key ingredient of BBM). In addition, OSM/BBM suppressed the pathogen Clostridioides difficile, potentially due to inclusion of HU58™ in GoodBiome™ Foods. Finally, all products enhanced a spectrum of metabolites well beyond SCFA, including vitamins (B3/B6), essential amino acids, and health-related metabolites such as indole-3-propionic acid. Overall, the addition of specific ingredients to complex foods was shown to specifically modulate the gut microbiome, potentially contributing to health benefits. Noticeably, our findings contradict a recent in vitro study, underscoring the critical role of employing a physiologically relevant digestion/absorption procedure for a more accurate evaluation of the microbiome-modulating potential of complex foods.
Collapse
Affiliation(s)
- Stef Deyaert
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Jonas Poppe
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Lam Dai Vu
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Aurélien Baudot
- Cryptobiotix, Technologiepark-Zwijnaarde 82, 9052 Ghent, Belgium; (S.D.)
| | - Sarah Bubeck
- Bubeck Scientific Communications, 194 Rainbow Drive #9418, Livingston, TX 77399, USA
| | - Thomas Bayne
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Kiran Krishnan
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Morgan Giusto
- Microbiome Labs, 101 E Town Pl, Saint Augustine, FL 92092, USA
| | - Samuel Moltz
- Novonesis, Biologiens Vej 2, 2800 Lyngby, Denmark
| | | |
Collapse
|
7
|
Noguera-Fernández N, Candela-González J, Orenes-Piñero E. Probiotics, Prebiotics, Fecal Microbiota Transplantation, and Dietary Patterns in Inflammatory Bowel Disease. Mol Nutr Food Res 2024; 68:e2400429. [PMID: 39194379 DOI: 10.1002/mnfr.202400429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/08/2024] [Indexed: 08/29/2024]
Abstract
SCOPE Inflammatory bowel disease (IBD) is one of the most common chronic and debilitating functional bowel disorders affecting around 11% of the population across the world. IBD is associated with 3.6 million physician visits per year, being the most common reason visiting a gastroenterologist and the second most common reason to be absent from work, sharply increasing the health care costs. METHODS AND RESULTS Several treatments seem to be effective in IBD symptoms relief, such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and dietary patterns. Probiotics (living microorganisms that can be supplemented) can protect against pathogenic bacteria due to their antimicrobial qualities. Prebiotics (nondigestible food ingredients) promote the growth of beneficial microbial strains in the gut, giving a health benefit to the host. FMT is supposed to directly change the recipient's microbial composition when a transfer of gastrointestinal microbiota from a healthy donor is carried out. And finally, dietary patterns are in the spotlight, due to the presence of certain nutrients in the gastrointestinal tract affecting gastrointestinal motility, sensitivity, barrier function, and gut microbiota. CONCLUSION It is particularly important to know what treatment options are available and which are the most efficient in relieving IBD symptoms and improving IBD patient's quality of life.
Collapse
Affiliation(s)
- Noah Noguera-Fernández
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Lorca, Murcia, 30800, Spain
| | - Joana Candela-González
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Lorca, Murcia, 30800, Spain
| | - Esteban Orenes-Piñero
- Department of Biochemistry and Molecular Biology-A, University of Murcia, Lorca, Murcia, 30800, Spain
| |
Collapse
|
8
|
Yuan D, Xiao W, Gao A, Lu W, Gao Z, Hu B, Wu Y, Jiang W, Li Y. In vitro colon fermentation behaviors of Ca 2+ cross-linked guluronic acid block from sodium alginate. Food Funct 2024; 15:8128-8142. [PMID: 39011745 DOI: 10.1039/d4fo00934g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
The degradation of sodium alginate by human gut microbiota was found to be retarded via calcium cross-linking in our previous study. We hypothesized that the guluronic acid block (GB) on the alginate molecule might be the key structural region affecting alginate degradation by the gut microbiota when cross-linked with calcium. This study aims to prove this hypothesis by studying the structural features of the cross-linked GB on its in vitro fecal fermentation behaviors concerning the aspects of total carbohydrate contents, monosaccharide contents, short-chain fatty acids production, calcium state variations, and structural variations. Herein, GB isolated from sodium alginate was cross-linked under ranges of molar ratios of [Ca2+]/[-COOH] that further restricted the degradation by gut microbiota similar to the cross-linked alginates. First, total carbohydrate contents, short-chain fatty acids production, monosaccharides contents, and calcium state analyses confirmed that the degradation of GB by gut microbiota was restricted by calcium cross-linking. Furthermore, the tracking analysis of structural variations during in vitro fermentation revealed that the "granules" structure could further restrict degradation by the gut microbiota, leaving more cross-linked GB fragments surviving in comparison to the "networks" structure. In addition, Bacteroides xylanisolvens showed a significant positive correlation to the "cross-linking porosity (R = 0.825, p < 0.001), which supported our previous findings on fermentation behaviors of cross-linked alginate. Together, guluronic acid blocks are the key structural regions that retard the degradation of sodium alginate by the gut microbiota when cross-linked with calcium.
Collapse
Affiliation(s)
- Dan Yuan
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Wenqian Xiao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Ao Gao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Wei Lu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Zhiming Gao
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Bing Hu
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, School of Life Sciences, Dalian Minzu University, Dalian 116600, China
| | - Yuehan Wu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Wenxin Jiang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| | - Yanlei Li
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China.
- Glyn O. Phillips Hydrocolloid Research Centre, School of Life and Health Sciences, Hubei University of Technology, Nanli Road, Wuhan 430068, P. R. China
| |
Collapse
|
9
|
So D, Tuck C. Innovative concepts in diet therapies in disorders of gut-brain interaction. JGH Open 2024; 8:e70001. [PMID: 39027160 PMCID: PMC11255864 DOI: 10.1002/jgh3.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024]
Abstract
Diet therapy in disorders of gut-brain interaction (DGBI) is rapidly advancing, with accumulating evidence to support two innovative therapies-manipulation of dietary fibers and enzyme supplementation-that target specific DGBI pathophysiology and modulate digestion. Dietary fibers escape digestion in the upper gastrointestinal tract and can influence gut function by impacting digestion, improving laxation, and interacting with the microbiota. A more nuanced understanding of different fiber types and their ability to impact gut function in highly specific ways has shown that fibers can impact distinct gut symptoms and pathophysiology. By considering their functional characteristics of bulking, gel-forming, and fermentability, restriction or supplementation of specific fibers can offer clinical value in DGBI. Similarly to fiber specificity, emerging evidence suggests that supplemental digestive enzymes may be targeted to known food triggers with consideration that enzymes are substrate specific. Limited evidence supports use of lactase to target lactose, and α-galactosidase to target galacto-oligosaccharides. Application of enzymes during manufacturing of food products may prove to be an additional strategy, although evidence is scant. Both innovative therapies may be utilized in isolation or in combination with other diet and nondiet therapies. Implementation can be guided by the principles that fiber modulation can be targeted to specific symptomology or requirement for alterations to gut function, and digestive enzymes can be targeted to known food triggers. This review aims to summarize recent literature of these two innovative concepts and provide practical suggestions for their implementation in clinical practice.
Collapse
Affiliation(s)
- Daniel So
- Department of GastroenterologyMonash University and Alfred HealthMelbourneAustralia
| | - Caroline Tuck
- Department of Nursing and Allied HealthSwinburne UniversityHawthornAustralia
| |
Collapse
|
10
|
Biruete A, Shin A, Kistler BM, Moe SM. Feeling gutted in chronic kidney disease (CKD): Gastrointestinal disorders and therapies to improve gastrointestinal health in individuals CKD, including those undergoing dialysis. Semin Dial 2024; 37:334-349. [PMID: 34708456 PMCID: PMC9043041 DOI: 10.1111/sdi.13030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/21/2021] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) affects 9.1% of the population worldwide. CKD may lead to structural and functional gastrointestinal alterations, including impairment in the intestinal barrier, digestion and absorption of nutrients, motility, and changes to the gut microbiome. These changes can lead to increased gastrointestinal symptoms in people with CKD, even in early grades of kidney dysfunction. Gastrointestinal symptoms have been associated with lower quality of life and reduced nutritional status. Therefore, there has been considerable interest in improving gastrointestinal health in this clinical population. Gastrointestinal health can be influenced by lifestyle and medications, particularly in advanced grades of kidney dysfunction. Therapies focused on gastrointestinal health have been studied, including the use of probiotics, prebiotics, and synbiotics, yielding limited and conflicting results. This review summarizes the alterations in the gastrointestinal tract structure and function and provides an overview of potential nutritional interventions that kidney disease professionals can provide to improve gastrointestinal health in individuals with CKD.
Collapse
Affiliation(s)
- Annabel Biruete
- Department of Nutrition and Dietetics, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Andrea Shin
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Brandon M. Kistler
- Department of Nutrition and Health Science, Ball State University, Muncie, Indiana, USA
| | - Sharon M. Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Anatomy, Cell Biology, and Anatomy, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
11
|
Li Q, Gu Y, Liang J, Yang Z, Qin J. A long journey to treat epilepsy with the gut microbiota. Front Cell Neurosci 2024; 18:1386205. [PMID: 38988662 PMCID: PMC11233807 DOI: 10.3389/fncel.2024.1386205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Epilepsy is a common neurological disorder that affects approximately 10.5 million children worldwide. Approximately 33% of affected patients exhibit resistance to all available antiseizure medications, but the underlying mechanisms are unknown and there is no effective treatment. Increasing evidence has shown that an abnormal gut microbiota may be associated with epilepsy. The gut microbiota can influence the function of the brain through multiple pathways, including the neuroendocrine, neuroimmune, and autonomic nervous systems. This review discusses the interactions between the central nervous system and the gastrointestinal tract (the brain-gut axis) and the role of the gut microbiota in the pathogenesis of epilepsy. However, the exact gut microbiota involved in epileptogenesis is unknown, and no consistent results have been obtained based on current research. Moreover, the target that should be further explored to identify a novel antiseizure drug is unclear. The role of the gut microbiota in epilepsy will most likely be uncovered with the development of genomics technology.
Collapse
Affiliation(s)
- Qinrui Li
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Youyu Gu
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Jingjing Liang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Zhixian Yang
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, China
- Epilepsy Center, Peking University People's Hospital, Beijing, China
| |
Collapse
|
12
|
Ribeiro MC, Levi Y, Moraschini V, Messora MR, Furlaneto FAC. Effects of Prebiotic Therapy on Gastrointestinal Microbiome of Individuals with Different Inflammatory Conditions: A Systematic Review of Randomized Controlled Trials. Probiotics Antimicrob Proteins 2024; 16:673-695. [PMID: 37093515 DOI: 10.1007/s12602-023-10075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2023] [Indexed: 04/25/2023]
Abstract
Prebiotics are substrates selectively utilized by host microorganisms conferring a health benefit. The effects of prebiotics on the gut microbiome of individuals with inflammatory processes need further investigations. The purpose of this study was to evaluate the effects of prebiotics on the gastrointestinal microbiome of individuals with some types of inflammatory conditions. Randomized controlled clinical trials (RCTs) evaluating the effects of different prebiotics on the gut microbiome were included. A systematic review of the literature including searches in PubMed/MEDLINE, EMBASE, Cochrane Library, Web of Science, and Scopus databases was performed until 23 March 2023. The risk of bias was assessed using the Cochrane Collaboration's criteria. Qualitative data was tabulated to facilitate comparisons and represented in the form of descriptive statistics and summary tables. Thirty trials, ranging from 12 to 135 patients, were included. The most commonly used prebiotic type was inulin-type fructans, and the treatment duration ranged from 1 to 36 weeks. The majority of the trials investigated the gut microbiome using 16 s rRNA gene sequencing on the Illumina Miseq platform. In general, prebiotic therapy exerted positive effects on inflammatory conditions. An increase in Bifidobacterium genus was the most common shift in bacterial composition observed. Within the limits of this systematic review, it can be suggested that prebiotic therapy presents the potential to favorably modulate the gastrointestinal microbiome of individuals with different types of inflammatory conditions.
Collapse
Affiliation(s)
- M C Ribeiro
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - Ylas Levi
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - V Moraschini
- Department of Periodontology, Dental Research Division, School of Dentistry, Veiga de Almeida University, Rio de Janeiro, Brazil
| | - M R Messora
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil
| | - F A C Furlaneto
- Department of Oral Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo - USP, Av. Café S/N, 14020-150, Ribeirao Preto, São Paulo, Brazil.
| |
Collapse
|
13
|
Sinopoulou V, Gordon M, Gregory V, Saadeh A, Akobeng AK. Prebiotics for induction and maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 2024; 3:CD015084. [PMID: 38501688 PMCID: PMC10949417 DOI: 10.1002/14651858.cd015084.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
BACKGROUND People affected by ulcerative colitis (UC) are interested in dietary therapies as treatments that can improve their health and quality of life. Prebiotics are a category of food ingredients theorised to have health benefits for the gastrointestinal system through their effect on the growth and activity of intestinal bacteria and probiotics. OBJECTIVES To assess the efficacy and safety of prebiotics for the induction and maintenance of remission in people with active UC. SEARCH METHODS We searched CENTRAL, MEDLINE, Embase, ClinicalTrials.gov, and WHO ICTRP on 24 June 2023. SELECTION CRITERIA We included randomised controlled trials (RCTs) on people with UC. We considered any type of standalone or combination prebiotic intervention, except those prebiotics combined with probiotics (known as synbiotics), compared to any control intervention. We considered interventions of any dose and duration. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. MAIN RESULTS We included 9 RCTs involving a total of 445 participants. Study duration ranged from 14 days to 2 to 3 months for induction and 1 to 6 months for maintenance of remission. All studies were on adults. Five studies were on people with mild to moderate active disease, three in remission or mild activity, and one did not mention. We judged only one study as at low risk of bias in all areas. Two studies compared prebiotics with placebo for induction of remission. We cannot draw any conclusions about clinical remission (70% versus 67%; risk ratio (RR) 1.05, 95% confidence interval (CI) 0.57 to 1.94); clinical improvement (mean Rachmilewitz score on day 14 of 4.1 versus 4.5; mean difference (MD) -0.40, 95% CI -2.67 to 1.87); faecal calprotectin levels (mean faecal calprotectin on day 14 of 1211 μg/mL versus 3740 μg/mL; MD -2529.00, 95% CI -6925.38 to 1867.38); interleukin-8 (IL-8) levels (mean IL-8 on day 7 of 2.9 pg/mL versus 5.0 pg/mL; MD -2.10, 95% CI -4.93 to 0.73); prostaglandin E2 (PGE-2) levels (mean PGE-2 on day 7 of 7.1 ng/mL versus 11.5 ng/mL; MD -4.40, 95% CI -20.25 to 11.45); or withdrawals due to adverse events (21% versus 8%; RR 2.73, 95% CI 0.51 to 14.55). All evidence was of very low certainty. No other outcomes were reported. Two studies compared inulin and oligofructose 15 g with inulin and oligofructose 7.5 g for induction of remission. We cannot draw any conclusions about clinical remission (53% versus 12.5%; RR 4.27, 95% CI 1.07 to 16.96); clinical improvement (67% versus 25%; RR 2.67, 95% CI 1.06 to 6.70); total adverse events (53.5% versus 31%; RR 1.71, 95% CI 0.72 to 4.06); or withdrawals due to adverse events (13% versus 25%; RR 0.53, 95% CI 0.11 to 2.50). All evidence was of very low certainty. No other outcomes were reported. One study compared prebiotics and anti-inflammatory therapy with anti-inflammatory therapy alone for induction of remission. We cannot draw any conclusions about clinical improvement (mean Lichtiger score at 4 weeks of 6.2 versus 10.3; MD -4.10, 95% CI -8.14 to -0.06) or serum C-reactive protein (CRP) levels (mean CRP levels at 4 weeks 0.55 ng/mL versus 0.50 ng/mL; MD 0.05, 95% CI -0.37 to 0.47). All evidence was of very low certainty. No other outcomes were reported. Three studies compared prebiotics with placebo for maintenance of remission. There may be no difference between groups in rate of clinical relapse (44% versus 33%; RR 1.36, 95% CI 0.79 to 2.31), and prebiotics may lead to more total adverse events than placebo (77% versus 46%; RR 1.68, 95% CI 1.18 to 2.40). The evidence was of low certainty. We cannot draw any conclusions about clinical improvement (mean partial Mayo score at day 60 of 0.428 versus 1.625; MD -1.20, 95% CI -2.17 to -0.22); faecal calprotectin levels (mean faecal calprotectin level at day 60 of 214 μg/mL versus 304 μg/mL; MD -89.79, 95% CI -221.30 to 41.72); quality of life (mean Inflammatory Bowel Disease Questionnaire (IBDQ) score at day 60 of 193.5 versus 188.0; MD 5.50, 95% CI -8.94 to 19.94); or withdrawals due to adverse events (28.5% versus 11%; RR 2.57, 95% CI 1.15 to 5.73). The evidence for these outcomes was of very low certainty. No other outcomes were reported. One study compared prebiotics with synbiotics for maintenance of remission. We cannot draw any conclusions about quality of life (mean IBDQ score at 4 weeks 182.4 versus 176.1; MD 6.30, 95% CI -6.61 to 19.21) or withdrawals due to adverse events (23% versus 20%; RR 1.13, 95% CI 0.48 to 2.62). All evidence was of very low certainty. No other outcomes were reported. One study compared prebiotics with probiotics for maintenance of remission. We cannot draw any conclusions about quality of life (mean IBDQ score at 4 weeks 182.4 versus 168.6; MD 13.60, 95% CI 1.22 to 25.98) or withdrawals due to adverse events (22.5% versus 22.5%; RR 1.00, 95% CI 0.44 to 2.26). All evidence was of very low certainty. No other outcomes were reported. AUTHORS' CONCLUSIONS There may be no difference in occurrence of clinical relapse when adjuvant treatment with prebiotics is compared with adjuvant treatment with placebo for maintenance of remission in UC. Adjuvant treatment with prebiotics may result in more total adverse events when compared to adjuvant treatment with placebo for maintenance of remission. We could draw no conclusions for any of the other outcomes in this comparison due to the very low certainty of the evidence. The evidence for all other comparisons and outcomes was also of very low certainty, precluding any conclusions. It is difficult to make any clear recommendations for future research based on the findings of this review given the clinical and methodological heterogeneity among studies. It is recommended that a consensus is reached on these issues prior to any further research.
Collapse
Affiliation(s)
| | - Morris Gordon
- School of Medicine, University of Central Lancashire, Preston, UK
| | | | - Anas Saadeh
- School of Medicine, University of Central Lancashire, Preston, UK
| | | |
Collapse
|
14
|
Lau RI, Su Q, Lau ISF, Ching JYL, Wong MCS, Lau LHS, Tun HM, Mok CKP, Chau SWH, Tse YK, Cheung CP, Li MKT, Yeung GTY, Cheong PK, Chan FKL, Ng SC. A synbiotic preparation (SIM01) for post-acute COVID-19 syndrome in Hong Kong (RECOVERY): a randomised, double-blind, placebo-controlled trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:256-265. [PMID: 38071990 DOI: 10.1016/s1473-3099(23)00685-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/11/2023] [Accepted: 10/26/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND Post-acute COVID-19 syndrome (PACS) affects over 65 million individuals worldwide but treatment options are scarce. We aimed to assess a synbiotic preparation (SIM01) for the alleviation of PACS symptoms. METHODS In this randomised, double-blind, placebo-controlled trial at a tertiary referral centre in Hong Kong, patients with PACS according to the US Centers for Disease Control and Prevention criteria were randomly assigned (1:1) by random permuted blocks to receive SIM01 (10 billion colony-forming units in sachets twice daily) or placebo orally for 6 months. Inclusion criterion was the presence of at least one of 14 PACS symptoms for 4 weeks or more after confirmed SARS-CoV-2 infection, including fatigue, memory loss, difficulty in concentration, insomnia, mood disturbance, hair loss, shortness of breath, coughing, inability to exercise, chest pain, muscle pain, joint pain, gastrointestinal upset, or general unwellness. Individuals were excluded if they were immunocompromised, were pregnant or breastfeeding, were unable to receive oral fluids, or if they had received gastrointestinal surgery in the 30 days before randomisation. Participants, care providers, and investigators were masked to group assignment. The primary outcome was alleviation of PACS symptoms by 6 months, assessed by an interviewer-administered 14-item questionnaire in the intention-to-treat population. Forward stepwise multivariable logistical regression was performed to identify predictors of symptom alleviation. The trial is registered with ClinicalTrials.gov, NCT04950803. FINDINGS Between June 25, 2021, and Aug 12, 2022, 463 patients were randomly assigned to receive SIM01 (n=232) or placebo (n=231). At 6 months, significantly higher proportions of the SIM01 group had alleviation of fatigue (OR 2·273, 95% CI 1·520-3·397, p=0·0001), memory loss (1·967, 1·271-3·044, p=0·0024), difficulty in concentration (2·644, 1·687-4·143, p<0·0001), gastrointestinal upset (1·995, 1·304-3·051, p=0·0014), and general unwellness (2·360, 1·428-3·900, p=0·0008) compared with the placebo group. Adverse event rates were similar between groups during treatment (SIM01 22 [10%] of 232 vs placebo 25 [11%] of 231; p=0·63). Treatment with SIM01, infection with omicron variants, vaccination before COVID-19, and mild acute COVID-19, were predictors of symptom alleviation (p<0·0036). INTERPRETATION Treatment with SIM01 alleviates multiple symptoms of PACS. Our findings have implications on the management of PACS through gut microbiome modulation. Further studies are warranted to explore the beneficial effects of SIM01 in other chronic or post-infection conditions. FUNDING Health and Medical Research Fund of Hong Kong, Hui Hoy and Chow Sin Lan Charity Fund, and InnoHK of the HKSAR Government. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Raphaela I Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China
| | - Qi Su
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China
| | - Ivan S F Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jessica Y L Ching
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China
| | - Martin C S Wong
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Louis H S Lau
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hein M Tun
- Microbiota I-Center, Hong Kong Special Administrative Region, China; The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chris K P Mok
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Steven W H Chau
- Department of Psychiatry, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yee Kit Tse
- Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Medical Data Analytics Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chun Pan Cheung
- Microbiota I-Center, Hong Kong Special Administrative Region, China
| | - Moses K T Li
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China
| | - Giann T Y Yeung
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Pui Kuan Cheong
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Francis K L Chan
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China; Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Siew C Ng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Microbiota I-Center, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
15
|
Ni Lochlainn M, Bowyer RCE, Moll JM, García MP, Wadge S, Baleanu AF, Nessa A, Sheedy A, Akdag G, Hart D, Raffaele G, Seed PT, Murphy C, Harridge SDR, Welch AA, Greig C, Whelan K, Steves CJ. Effect of gut microbiome modulation on muscle function and cognition: the PROMOTe randomised controlled trial. Nat Commun 2024; 15:1859. [PMID: 38424099 PMCID: PMC10904794 DOI: 10.1038/s41467-024-46116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Studies suggest that inducing gut microbiota changes may alter both muscle physiology and cognitive behaviour. Gut microbiota may play a role in both anabolic resistance of older muscle, and cognition. In this placebo controlled double blinded randomised controlled trial of 36 twin pairs (72 individuals), aged ≥60, each twin pair are block randomised to receive either placebo or prebiotic daily for 12 weeks. Resistance exercise and branched chain amino acid (BCAA) supplementation is prescribed to all participants. Outcomes are physical function and cognition. The trial is carried out remotely using video visits, online questionnaires and cognitive testing, and posting of equipment and biological samples. The prebiotic supplement is well tolerated and results in a changed gut microbiome [e.g., increased relative Bifidobacterium abundance]. There is no significant difference between prebiotic and placebo for the primary outcome of chair rise time (β = 0.579; 95% CI -1.080-2.239 p = 0.494). The prebiotic improves cognition (factor score versus placebo (β = -0.482; 95% CI,-0.813, -0.141; p = 0.014)). Our results demonstrate that cheap and readily available gut microbiome interventions may improve cognition in our ageing population. We illustrate the feasibility of remotely delivered trials for older people, which could reduce under-representation of older people in clinical trials. ClinicalTrials.gov registration: NCT04309292.
Collapse
Affiliation(s)
- Mary Ni Lochlainn
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK.
| | - Ruth C E Bowyer
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
- The Alan Turing Institute, London, NW1 2DB, UK
| | | | - María Paz García
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Samuel Wadge
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Andrei-Florin Baleanu
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Ayrun Nessa
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Alyce Sheedy
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Gulsah Akdag
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Deborah Hart
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK
| | - Giulia Raffaele
- GKT School of Medical Education, King's College London, London, UK
| | - Paul T Seed
- Unit for Medical Statistics/Department for Women and Children's Health, School of Life Course and Population Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Caroline Murphy
- King's Clinical Trials Unit, Research Management and Innovation Directorate, King's College London, London, UK
| | - Stephen D R Harridge
- Centre for Human & Applied Physiological Sciences, King's College London, London, UK
| | - Ailsa A Welch
- Department of Epidemiology and Public Health, Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, UK
| | - Carolyn Greig
- School of Sport, Exercise, and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
- MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Kevin Whelan
- King's College London, Department of Nutritional Sciences, Franklin Wilkins Building, SE1 9NH, London, UK
| | - Claire J Steves
- King's College London, Department of Twin Research and Genetic Epidemiology, London, SE1 7EH, UK.
| |
Collapse
|
16
|
Bevilacqua A, Campaniello D, Speranza B, Racioppo A, Sinigaglia M, Corbo MR. An Update on Prebiotics and on Their Health Effects. Foods 2024; 13:446. [PMID: 38338581 PMCID: PMC10855651 DOI: 10.3390/foods13030446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/20/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Prebiotic compounds were originally defined as "a nondigestible food ingredient that beneficially affects the host by selectively stimulating the growth and/or activity of one or a limited number of bacteria in the colon, and thus improves host health"; however, a significant modulation of the definition was carried out in the consensus panel of The International Scientific Association for Probiotics and Prebiotics (ISAPP), and the last definition states that "prebiotics are substrates that are selectively utilized by host microorganisms conferring a health benefit". Health effects of prebiotics compounds attracted the interest of researchers, food companies and Regulatory Agencies, as inferred by the number of articles on Scopus for the keywords "prebiotic" and "health effects", that is ca. 2000, for the period January 2021-January 2024. Therefore, the aim of this paper is to contribute to the debate on these topics by offering an overview of existing knowledge and advances in this field. A literature search was performed for the period 2012-2023 and after the selection of the most relevant items, the attention was focused on seven conditions for which at least 8-10 different studies were found, namely colorectal cancer, neurological or psychiatric conditions, intestinal diseases, obesity, diabetes, metabolic syndrome, and immune system disorders. In addition, the analysis of the most recent articles through the software VosViewer version 1.6.20 pointed out the existence of five clusters or macro-categories, namely: (i) pathologies; (ii) metabolic condvitions; (iii) structure and use in food; (iv) immunomodulation; (v) effect on gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Rosaria Corbo
- Department of the Science of Agriculture, Food, Natural Resources and Engineering, University of Foggia, 71122 Foggia, Italy; (A.B.); (D.C.); (B.S.); (A.R.); (M.S.)
| |
Collapse
|
17
|
Tuohy K, Vaughan EE, Harthoorn LF, Blaak EE, Burnet PWJ, Busetti A, Chakrabarti A, Delzenne N, de Vos P, Dye L, Guillemet D, Houghton LA, Kardinaal AFM, Mersh C, Musa-Veloso K, Nielsen A, Palasinska J, Salminen S, Walton G, Venlet N, Hubermont C, Calder PC. Prebiotics in food and dietary supplements: a roadmap to EU health claims. Gut Microbes 2024; 16:2428848. [PMID: 39544074 DOI: 10.1080/19490976.2024.2428848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 11/17/2024] Open
Abstract
Numerous studies have established that prebiotic ingredients in foods and dietary supplements may play a role in supporting human health. Over the three decades that have passed since prebiotics were first defined as a concept, research has revealed a complex universe of prebiotic-induced changes to the human microbiota. There are strong indications of a direct link between these prebiotic-induced changes and specific health benefits. However, at the present time, the EU has not permitted use of the term 'prebiotic' in connection with an approved health claim. This paper is the outcome of a workshop organized on the 25th October 2023 by the European branch of the International Life Science Institute (ILSI). It provides an overview of the regulatory requirements for authorized health claims in the EU, key areas of prebiotic research, and findings to date in relation to prebiotics and digestive, immune, metabolic and cognitive health. Research gaps and documentation challenges are then explored and a roadmap proposed for achieving authorization of 'prebiotic' in the wording of future EU health claims.
Collapse
Affiliation(s)
- Kieran Tuohy
- School of Food Science & Nutrition, University of Leeds, Leeds, UK
| | | | | | - Ellen E Blaak
- Department of Human Biology, NUTRIM, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | | | | | - Nathalie Delzenne
- Woluwe Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Paul de Vos
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Louise Dye
- Institute for Sustainable Food, University of Sheffield, Sheffield, UK
| | | | | | | | - Cath Mersh
- Cath Mersh Communications, Aarhus, Denmark
| | | | | | | | - Seppo Salminen
- Center for Food and Nutrition Research, Faculty of Medicine, University of Turku, Turku, Finland
| | - Gemma Walton
- Food and Nutritional Sciences, University of Reading, Reading, UK
| | | | | | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, United Kingdom and NIHR Southampton Biomedical, Research Centre, University Hospital NHS Foundation Trust and University of Southampton, Southampton, UK
| |
Collapse
|
18
|
Weingarden AR, Ko CW. Non-prescription Therapeutics. Am J Gastroenterol 2024; 119:S7-S15. [PMID: 38153220 DOI: 10.14309/ajg.0000000000002578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/31/2023] [Indexed: 12/29/2023]
Affiliation(s)
- Alexa R Weingarden
- Division of Gastroenterology & Hepatology, Department of Medicine, Stanford University, Redwood City, California, USA
| | - Cynthia W Ko
- Division of Gastroenterology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
19
|
Guo J, Zhou B, Niu Y, Liu L, Yang L. Engineered probiotics introduced to improve intestinal microecology for the treatment of chronic diseases: present state and perspectives. J Diabetes Metab Disord 2023; 22:1029-1038. [PMID: 37975092 PMCID: PMC10638336 DOI: 10.1007/s40200-023-01279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/05/2023] [Indexed: 11/19/2023]
Abstract
Purpose Correcting intestinal microecological imbalance has become one of the core strategies to treat chronic diseases. Some traditional microecology-based therapies targeting intestine, such as prebiotic therapy, probiotic therapy and fecal microbiota transplantation therapy, have been used in the prevention and treatment of clinical chronic diseases, which still facing low safety and poor controllability problems. The development of synthetic biology technology has promoted the development of intestinal microecology-based therapeutics for chronic diseases, which exhibiting higher robustness and controllability, and become an important part of the next generation of microecological therapy. The purpose of this review is to summarize the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases. Methods The available literatures were searched to find out experimental studies and relevant review articles on the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases from year 1990 to 2023. Results Evidence proposed that synthetic biology has been applied in the intestinal microecology-based therapeutics for chronic diseases, covering metabolic diseases (e.g. diabetes, obesity, nonalcoholic fatty liver disease and phenylketonuria), digestive diseases (e.g. inflammatory bowel disease and colorectal cancer), and neurodegenerative diseases (e.g. Alzheimer's disease and Parkinson's disease). Conclusion This review summarizes the application of synthetic biology in intestinal microecology-based therapeutics for major chronic diseases and discusses the opportunities and challenges in the above process, providing clinical possibilities of synthetic biology technology applied in microecological therapies.
Collapse
Affiliation(s)
- Jianquan Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, (Shanxi Medical University), Ministry of Education, Taiyuan, PR China
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Bangyuan Zhou
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Yali Niu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liyang Yang
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine, 030619 Jinzhong, PR China
| |
Collapse
|
20
|
Nagy DU, Sándor-Bajusz KA, Bódy B, Decsi T, Van Harsselaar J, Theis S, Lohner S. Effect of chicory-derived inulin-type fructans on abundance of Bifidobacterium and on bowel function: a systematic review with meta-analyses. Crit Rev Food Sci Nutr 2023; 63:12018-12035. [PMID: 35833477 DOI: 10.1080/10408398.2022.2098246] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Inulin-type fructans are considered to stimulate the growth of beneficial microorganisms, like Bifidobacterium in the gut and support health. However, both the fructan source and chemical structure may modify these effects. A systematic review was conducted to assess the effects of chicory-derived inulin-type fructans consumed either in specific foods or as dietary supplements on abundance of Bifidobacterium in the gut and on health-related outcomes. Three electronic databases and two clinical trial registries were systematically searched until January 2021. Two authors independently selected randomized controlled trials that investigated with a protocol of minimum seven days supplementation the effect of chicory-derived inulin-type fructans on Bifidobacterium abundance in any population. Meta-analyses with random-effects model were conducted on Bifidobacterium abundance and bowel function parameters. We evaluated risk of bias using Cochrane RoB tool. Chicory-derived inulin-type fructans at a dose of 3-20 g/day significantly increased Bifidobacterium abundance in participants with an age range from 0 to 83 years (standardized mean difference: 0.83, 95% CI: 0.58-1.08; p < 0.01; 50 studies; 2525 participants). Significant bifidogenic effects were observed in healthy individuals and in populations with health impairments, except gastrointestinal disorders. Significant beneficial effects on bowel function parameters were observed in healthy subjects. Chicory-derived inulin-type fructans may have significant bifidogenic effects and may beneficially influence bowel function in healthy individuals. PROSPERO registration number CRD42020162892.
Collapse
Affiliation(s)
- Dávid U Nagy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Halle (Saale), Germany
| | - Kinga Amália Sándor-Bajusz
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Blanka Bódy
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Decsi
- Department of Paediatrics, Clinical Center of the University of Pécs, Medical School, University of Pécs, Pécs, Hungary
| | | | - Stephan Theis
- BENEO-Institute, c/o BENEO GmbH, Obrigheim, (Germany)
| | - Szimonetta Lohner
- Department of Public Health Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
21
|
Garcia-Mazcorro JF, Amieva-Balmori M, Triana-Romero A, Wilson B, Smith L, Reyes-Huerta J, Rossi M, Whelan K, Remes-Troche JM. Fecal Microbial Composition and Predicted Functional Profile in Irritable Bowel Syndrome Differ between Subtypes and Geographical Locations. Microorganisms 2023; 11:2493. [PMID: 37894151 PMCID: PMC10608977 DOI: 10.3390/microorganisms11102493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Increasing evidence suggests a microbial pathogenesis in irritable bowel syndrome (IBS) but the relationship remains elusive. Fecal DNA samples from 120 patients with IBS, 82 Mexican (IBS-C: n = 33, IBS-D: n = 24, IBS-M: n = 25) and 38 British (IBS-C: n = 6, IBS-D: n = 27, IBS-M: n = 5), were available for analysis using 16S rRNA gene sequencing. Firmicutes (mean: 82.1%), Actinobacteria (10.2%), and Bacteroidetes (4.4%) were the most abundant taxa. The analysis of all samples (n = 120), and females (n = 94) only, showed no significant differences in bacterial microbiota, but the analysis of Mexican patients (n = 82) showed several differences in key taxa (e.g., Faecalibacterium) among the different IBS subtypes. In IBS-D there were significantly higher Bacteroidetes in British patients (n = 27) than in Mexican patients (n = 24), suggesting unique fecal microbiota signatures within the same IBS subtype. These differences in IBS-D were also observed at lower phylogenetic levels (e.g., higher Enterobacteriaceae and Streptococcus in Mexican patients) and were accompanied by differences in several alpha diversity metrics. Beta diversity was not different among IBS subtypes when using all samples, but the analysis of IBS-D patients revealed consistent differences between Mexican and British patients. This study suggests that fecal microbiota is different between IBS subtypes and also within each subtype depending on geographical location.
Collapse
Affiliation(s)
| | - Mercedes Amieva-Balmori
- Instituto de Investigaciones Médico Biológicas, Universidad Veracruzana, Veracruz 91700, Mexico
| | - Arturo Triana-Romero
- Instituto de Investigaciones Médico Biológicas, Universidad Veracruzana, Veracruz 91700, Mexico
| | - Bridgette Wilson
- Department of Nutritional Sciences, King’s College London, London WC2R 2LS, UK
| | - Leanne Smith
- Department of Nutritional Sciences, King’s College London, London WC2R 2LS, UK
| | - Job Reyes-Huerta
- Instituto de Investigaciones Médico Biológicas, Universidad Veracruzana, Veracruz 91700, Mexico
| | - Megan Rossi
- Department of Nutritional Sciences, King’s College London, London WC2R 2LS, UK
| | - Kevin Whelan
- Department of Nutritional Sciences, King’s College London, London WC2R 2LS, UK
| | - Jose M. Remes-Troche
- Instituto de Investigaciones Médico Biológicas, Universidad Veracruzana, Veracruz 91700, Mexico
| |
Collapse
|
22
|
Andersen S, Henden A, Staudacher H, Kennedy G, Gavin N. Fibre intake and supplementation during treatment for haematological malignancies: A scoping review. J Hum Nutr Diet 2023; 36:1982-1991. [PMID: 37403340 DOI: 10.1111/jhn.13209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/03/2023] [Indexed: 07/06/2023]
Abstract
BACKGROUND Gastrointestinal microbiome diversity decreases rapidly during haematological cancer treatment with low diversity associated with poorer clinical outcomes. Therefore, factors that may benefit the microbiome require evaluation. This scoping review aimed to identify and describe the available research on fibre intake and supplementation during haematological cancer treatment. METHODS This scoping review included observational studies of usual fibre intake and intervention fibre supplementation trials with patients undergoing chemotherapy, immunotherapy or stem cell transplantation for haematological malignancy. Comprehensive searching of four databases plus grey literature was conducted. Study design, type of fibre (for fibre supplementation trials) and evaluated outcomes were recorded. The review was registered on Open Science Framework and completed in three stages. There were no date restrictions in the search and only studies in English were included. RESULTS Five studies met the inclusion criteria for the review including two observational studies and three supplementation trials. No randomised control trials were identified. The interventional studies provided either a single fibre supplement (fructo-oligosaccharide) or a combination of fibres (polydextrose, lactosucrose, resistant starch or oligosaccharides plus fibre) during stem cell transplantation. The most frequently evaluated outcomes included tolerability of the fibre supplement, clinical outcomes (infection, graft versus host disease, survival) and the impact on the gastrointestinal microbiome. CONCLUSIONS Further research, including randomised controlled trials, is needed to investigate the role of fibre during haematological cancer treatment, including the pathways in which it might improve disease outcome.
Collapse
Affiliation(s)
- Sarah Andersen
- Department of Dietetics and Foodservices, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- School of Human Movement and Nutrition Sciences, University of Queensland, St Lucia, QLD, Australia
| | - Andrea Henden
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Translational Cancer Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Heidi Staudacher
- Food & Mood Centre, IMPACT Institute, School of Medicine, Geelong, Australia
| | - Glen Kennedy
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Nicole Gavin
- Cancer Care Services, Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Centre for Healthcare Transformation, Kelvin Grove, QLD, Australia
| |
Collapse
|
23
|
Napolitano M, Fasulo E, Ungaro F, Massimino L, Sinagra E, Danese S, Mandarino FV. Gut Dysbiosis in Irritable Bowel Syndrome: A Narrative Review on Correlation with Disease Subtypes and Novel Therapeutic Implications. Microorganisms 2023; 11:2369. [PMID: 37894027 PMCID: PMC10609453 DOI: 10.3390/microorganisms11102369] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder characterized by chronic abdominal pain and altered bowel habits. It can be subclassified in different subtypes according to the main clinical manifestation: constipation, diarrhea, mixed, and unclassified. Over the past decade, the role of gut microbiota in IBS has garnered significant attention in the scientific community. Emerging research spotlights the intricate involvement of microbiota dysbiosis in IBS pathogenesis. Studies have demonstrated reduced microbial diversity and stability and specific microbial alterations for each disease subgroup. Microbiota-targeted treatments, such as antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and even diet, offer exciting prospects for managing IBS. However, definitive conclusions are hindered by the heterogeneity of these studies. Further research should focus on elucidating the mechanisms, developing microbiome-based diagnostics, and enabling personalized therapies tailored to an individual's microbiome profile. This review takes a deep dive into the microscopic world inhabiting our guts, and its implications for IBS. Our aim is to elucidate the complex interplay between gut microbiota and each IBS subtype, exploring novel microbiota-targeted treatments and providing a comprehensive overview of the current state of knowledge.
Collapse
Affiliation(s)
- Maria Napolitano
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Ernesto Fasulo
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Federica Ungaro
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Luca Massimino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy;
| | - Silvio Danese
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Francesco Vito Mandarino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| |
Collapse
|
24
|
Qu Y, Park SH, Dallas DC. The Role of Bovine Kappa-Casein Glycomacropeptide in Modulating the Microbiome and Inflammatory Responses of Irritable Bowel Syndrome. Nutrients 2023; 15:3991. [PMID: 37764775 PMCID: PMC10538225 DOI: 10.3390/nu15183991] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/10/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal disorder marked by chronic abdominal pain, bloating, and irregular bowel habits. Effective treatments are still actively sought. Kappa-casein glycomacropeptide (GMP), a milk-derived peptide, holds promise because it can modulate the gut microbiome, immune responses, gut motility, and barrier functions, as well as binding toxins. These properties align with the recognized pathophysiological aspects of IBS, including gut microbiota imbalances, immune system dysregulation, and altered gut barrier functions. This review delves into GMP's role in regulating the gut microbiome, accentuating its influence on bacterial populations and its potential to promote beneficial bacteria while inhibiting pathogenic varieties. It further investigates the gut microbial shifts observed in IBS patients and contemplates GMP's potential for restoring microbial equilibrium and overall gut health. The anti-inflammatory attributes of GMP, especially its impact on vital inflammatory markers and capacity to temper the low-grade inflammation present in IBS are also discussed. In addition, this review delves into current research on GMP's effects on gut motility and barrier integrity and examines the changes in gut motility and barrier function observed in IBS sufferers. The overarching goal is to assess the potential clinical utility of GMP in IBS management.
Collapse
Affiliation(s)
- Yunyao Qu
- Department of Food Science & Technology, Oregon State University, Corvallis, OR 97331, USA; (Y.Q.); (S.H.P.)
- Nutrition Program, College of Health, Oregon State University, Corvallis, OR 97331, USA
| | - Si Hong Park
- Department of Food Science & Technology, Oregon State University, Corvallis, OR 97331, USA; (Y.Q.); (S.H.P.)
| | - David C. Dallas
- Department of Food Science & Technology, Oregon State University, Corvallis, OR 97331, USA; (Y.Q.); (S.H.P.)
- Nutrition Program, College of Health, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
25
|
Oh CK, Park JK, Kim YJ, Kim JB. Efficacy and safety of human gut-derived multi-strain probiotics in patients with irritable bowel syndrome: A prospective open-label observation study. Medicine (Baltimore) 2023; 102:e34899. [PMID: 37653742 PMCID: PMC10470732 DOI: 10.1097/md.0000000000034899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/25/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
This study aimed to investigate the efficacy and safety of human gut-derived multi-strain probiotics in patients with irritable bowel syndrome (IBS). This was an open-label, prospective, observational study. Patients with IBS were administered human gut-derived multi-strain probiotics for 4 weeks. The primary and secondary outcomes were based on the overall responder rate of the total IBS severity scoring system (IBS-SSS) score (>50-point decrease) and the IBS quality of life (IBS-QOL) score and IBS-SSS1 subscore (>10-point decrease in both scores), respectively. The estimated response rate is 55%. Of 44 patients, the total IBS-SSS score responder rate was 18.2% and 63.6% of patients at 2 and 4 weeks, respectively (P = .018). Compared with baseline, a significant improvement in the IBS-QOL score was observed in 27.3% and 63.6% of patients at 2 and 4 weeks, respectively (P = .001). Overall improvement rates in the IBS-SSS1 subscore were observed in 29.5% and 61.4% of patients at 2 and 4 weeks, respectively (P < .001). Primary and secondary outcomes were higher at 4 weeks (total IBS-SSS score, 63.6%; IBS-QOL score, 63.6%; IBS-SSS1 subscore, 61.4%) than the estimated responder rate (55%). Human gut-derived multi-strain probiotics have the potential to become an effective and safe treatment option for IBS patients.
Collapse
Affiliation(s)
- Chang Kyo Oh
- Division of Gastroenterology, Department of Internal Medicine, Kangnam Sacred Heart Hospital, College of Medicine, The Hallym University of Korea, Yeoungdeungpo-gu, Seoul, Korea
| | - Jae Keun Park
- Division of Gastroenterology, Department of Internal Medicine, Kangnam Sacred Heart Hospital, College of Medicine, The Hallym University of Korea, Yeoungdeungpo-gu, Seoul, Korea
| | - Yu Jin Kim
- Division of Gastroenterology, Department of Internal Medicine, Kangnam Sacred Heart Hospital, College of Medicine, The Hallym University of Korea, Yeoungdeungpo-gu, Seoul, Korea
| | - Jin Bae Kim
- Division of Gastroenterology, Department of Internal Medicine, Kangnam Sacred Heart Hospital, College of Medicine, The Hallym University of Korea, Yeoungdeungpo-gu, Seoul, Korea
| |
Collapse
|
26
|
Camacho-Díaz BH, Arenas-Ocampo ML, Osorio-Díaz P, Jiménez-Aparicio AR, Alvarado-Jasso GM, Saavedra-Briones EV, Valdovinos-Díaz MÁ, Gómez-Reyes E. The Effects of Agave Fructans in a Functional Food Consumed by Patients with Irritable Bowel Syndrome with Constipation: A Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2023; 15:3526. [PMID: 37630717 PMCID: PMC10460012 DOI: 10.3390/nu15163526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Irritable bowel syndrome displays three different subtypes: constipation (IBS-C), diarrhea (IBS-D), and mixed (IBS-M). Treatment with dietary fiber is used, with consideration given both to the chemical composition of the fiber and to the different subtypes of IBS. The IBS-D subtype is usually treated with a low-FODMAPs diet, whereas the IBS-C subtype suggests prebiotics and probiotics to promote microbiota restoration. The aim of this study was to assess the effects of employing agave fructans as the soluble fiber of a jelly (Gelyfun®gastro) containing 8 g per serving in the IBS-C group (n = 50), using a randomized, double-blind, time-limited trial for four weeks. We evaluated changes in the frequency and types of bowel movements through the Bristol scale, and the improvement of the condition was evaluated using quality of life (IBS-QOL) and anxiety-depression (HADS) scales. The main results were that the number of bowel movements increased by more than 80%, with at least one stool per day from fifteen days onwards, without a laxative effect for the group treated. Finally, the quality of life with the prebiotic jelly was significantly improved compared to the placebo in all specific domains, in addition to significantly reducing anxiety and depression.
Collapse
Affiliation(s)
- Brenda Hildeliza Camacho-Díaz
- Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional, San Isidro, Yautepec 62731, Morelos, Mexico; (M.L.A.-O.); (P.O.-D.); (A.R.J.-A.)
| | - Martha Lucía Arenas-Ocampo
- Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional, San Isidro, Yautepec 62731, Morelos, Mexico; (M.L.A.-O.); (P.O.-D.); (A.R.J.-A.)
| | - Perla Osorio-Díaz
- Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional, San Isidro, Yautepec 62731, Morelos, Mexico; (M.L.A.-O.); (P.O.-D.); (A.R.J.-A.)
| | - Antonio Ruperto Jiménez-Aparicio
- Centro de Desarrollo de Productos Bióticos, Instituto Politécnico Nacional, San Isidro, Yautepec 62731, Morelos, Mexico; (M.L.A.-O.); (P.O.-D.); (A.R.J.-A.)
| | | | - Edén Valfré Saavedra-Briones
- Facultad de Nutrición, Universidad Autónoma del Estado de Morelos, Vista Hermosa, Cuernavaca 62290, Morelos, Mexico;
| | - Miguel Ángel Valdovinos-Díaz
- Laboratorio de Motilidad Gastrointestinal del Departamento de Gastroenterología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Tlalpan, Ciudad de México 14080, Mexico;
| | - Elisa Gómez-Reyes
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Campus Ciudad de México, Calle del Puente 222, Col Ejidos de Huipulco, Tlalpan, Ciudad de México 14380, Mexico
| |
Collapse
|
27
|
Jayasinghe M, Damianos JA, Prathiraja O, Oorloff MD, Nagalmulla K GM, Nadella A, Caldera D, Mohtashim A. Irritable Bowel Syndrome: Treating the Gut and Brain/Mind at the Same Time. Cureus 2023; 15:e43404. [PMID: 37706135 PMCID: PMC10496425 DOI: 10.7759/cureus.43404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2023] [Indexed: 09/15/2023] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal (GI) disorders in the world. Although IBS does not affect a person's life span, it can significantly influence their quality of life. The treatment of IBS should be tailored to each patient's specific symptomatology because it can often be difficult to manage. Given that the pathogenesis of IBS is not well understood, it places a tremendous load on healthcare resources. Over the years, IBS has been described as either a simple GI disorder or a more complex multi-symptomatic gut-brain axis disorder. Many persons with IBS have psychological issues in addition to gastrointestinal symptoms, offering the door to non-pharmacological therapies such as cognitive behavioral therapy, gut-directed hypnosis, or psychodynamic interpersonal therapy. Non-pharmacological therapies with no side effects should be used as first-line therapy. Diet, exercise, microbiota-targeted therapies, and psychological treatments are among the most significant interventions. This review goes into the details of all the non-pharmacological interventions that can be used to treat IBS.
Collapse
Affiliation(s)
| | - John A Damianos
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Duncanson K, Tikhe D, Williams GM, Talley NJ. Irritable bowel syndrome - controversies in diagnosis and management. Expert Rev Gastroenterol Hepatol 2023; 17:649-663. [PMID: 37317843 DOI: 10.1080/17474124.2023.2223975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
INTRODUCTION The irritable bowel syndrome (IBS) is the best-recognized disorder of gut brain interactions (DGBI). However, it is controversial if the Rome IV criteria iteration for IBS diagnosis is fit for purpose. AREAS COVERED This review critically evaluates Rome IV criteria for diagnosis of IBS and addresses clinical considerations in IBS treatment and management, including dietary factors, biomarkers, disease mimics, symptom severity, and subtypes. The role of diet in IBS is critically reviewed along with the influence of the microbiota, including small intestinal bacterial overgrowth. EXPERT OPINION Emerging data suggest the Rome IV criteria are more suitable for identifying severe IBS and least useful for sub-diagnostic patients who are still likely to benefit from IBS treatment. Despite convincing evidence that IBS symptoms are diet-driven and often postprandial, a relationship to eating is not a Rome IV diagnostic criterion. Few IBS biomarkers have been identified, suggesting the syndrome is too heterogeneous to be measured by a single marker, and combined biomarker, clinical, dietary, and microbial profiling may be needed for objective characterization. With many organic diseases mimicking and overlapping with IBS, it's important clinicians are knowledgable about this to mitigate the risk of missing comorbid organic intestinal disease and to optimally treat IBS symptoms.
Collapse
Affiliation(s)
- Kerith Duncanson
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Centre of Research Excellence in Digestive Health, The University of Newcastle, New Lambton Heights, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Dhanashree Tikhe
- Centre of Research Excellence in Digestive Health, The University of Newcastle, New Lambton Heights, NSW, Australia
- Department of Gastroenterology, John Hunter Hospital, Hunter New England Local Health District, New Lambton Heights, NSW, Australia
| | - Georgina M Williams
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Centre of Research Excellence in Digestive Health, The University of Newcastle, New Lambton Heights, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nicholas J Talley
- School of Medicine and Public Health, The University of Newcastle, Callaghan, NSW, Australia
- Centre of Research Excellence in Digestive Health, The University of Newcastle, New Lambton Heights, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Gastroenterology, John Hunter Hospital, Hunter New England Local Health District, New Lambton Heights, NSW, Australia
| |
Collapse
|
29
|
Corrêa RO, Castro PR, Fachi JL, Nirello VD, El-Sahhar S, Imada S, Pereira GV, Pral LP, Araújo NVP, Fernandes MF, Matheus VA, de Souza Felipe J, Dos Santos Pereira Gomes AB, de Oliveira S, de Rezende Rodovalho V, de Oliveira SRM, de Assis HC, Oliveira SC, Dos Santos Martins F, Martens E, Colonna M, Varga-Weisz P, Vinolo MAR. Inulin diet uncovers complex diet-microbiota-immune cell interactions remodeling the gut epithelium. MICROBIOME 2023; 11:90. [PMID: 37101209 PMCID: PMC10131329 DOI: 10.1186/s40168-023-01520-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/16/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND The continuous proliferation of intestinal stem cells followed by their tightly regulated differentiation to epithelial cells is essential for the maintenance of the gut epithelial barrier and its functions. How these processes are tuned by diet and gut microbiome is an important, but poorly understood question. Dietary soluble fibers, such as inulin, are known for their ability to impact the gut bacterial community and gut epithelium, and their consumption has been usually associated with health improvement in mice and humans. In this study, we tested the hypothesis that inulin consumption modifies the composition of colonic bacteria and this impacts intestinal stem cells functions, thus affecting the epithelial structure. METHODS Mice were fed with a diet containing 5% of the insoluble fiber cellulose or the same diet enriched with an additional 10% of inulin. Using a combination of histochemistry, host cell transcriptomics, 16S microbiome analysis, germ-free, gnotobiotic, and genetically modified mouse models, we analyzed the impact of inulin intake on the colonic epithelium, intestinal bacteria, and the local immune compartment. RESULTS We show that the consumption of inulin diet alters the colon epithelium by increasing the proliferation of intestinal stem cells, leading to deeper crypts and longer colons. This effect was dependent on the inulin-altered gut microbiota, as no modulations were observed in animals deprived of microbiota, nor in mice fed cellulose-enriched diets. We also describe the pivotal role of γδ T lymphocytes and IL-22 in this microenvironment, as the inulin diet failed to induce epithelium remodeling in mice lacking this T cell population or cytokine, highlighting their importance in the diet-microbiota-epithelium-immune system crosstalk. CONCLUSION This study indicates that the intake of inulin affects the activity of intestinal stem cells and drives a homeostatic remodeling of the colon epithelium, an effect that requires the gut microbiota, γδ T cells, and the presence of IL-22. Our study indicates complex cross kingdom and cross cell type interactions involved in the adaptation of the colon epithelium to the luminal environment in steady state. Video Abstract.
Collapse
Affiliation(s)
- Renan Oliveira Corrêa
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil.
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, 02139, USA.
| | - Pollyana Ribeiro Castro
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - José Luís Fachi
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Vinícius Dias Nirello
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Salma El-Sahhar
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, UK
| | - Shinya Imada
- Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA, 02139, USA
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, 734-8551, Japan
| | - Gabriel Vasconcelos Pereira
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
- University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Laís Passariello Pral
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Nathália Vitoria Pereira Araújo
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Mariane Font Fernandes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Valquíria Aparecida Matheus
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Jaqueline de Souza Felipe
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Arilson Bernardo Dos Santos Pereira Gomes
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Sarah de Oliveira
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Vinícius de Rezende Rodovalho
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Samantha Roberta Machado de Oliveira
- Laboratory of Biotherapeutics Agents, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Helder Carvalho de Assis
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Sergio Costa Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Flaviano Dos Santos Martins
- Laboratory of Biotherapeutics Agents, Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Eric Martens
- University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Patrick Varga-Weisz
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
- School of Life Sciences, University of Essex, Colchester, CO4 3SQ, UK
- São Paulo Excellence Chair, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil
| | - Marco Aurélio Ramirez Vinolo
- Laboratory of Immunoinflammation, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil.
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP, 13083-862, Brazil.
- Experimental Medicine Research Cluster, Campinas, SP, 13083-862, Brazil.
- Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, SP, 13083-864, Brazil.
| |
Collapse
|
30
|
Bischoff SC, Ockenga J, Eshraghian A, Barazzoni R, Busetto L, Campmans-Kuijpers M, Cardinale V, Chermesh I, Kani HT, Khannoussi W, Lacaze L, Léon-Sanz M, Mendive JM, Müller MW, Tacke F, Thorell A, Vranesic Bender D, Weimann A, Cuerda C. Practical guideline on obesity care in patients with gastrointestinal and liver diseases - Joint ESPEN/UEG guideline. Clin Nutr 2023; 42:987-1024. [PMID: 37146466 DOI: 10.1016/j.clnu.2023.03.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Patients with chronic gastrointestinal disease such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), celiac disease, gastroesophageal reflux disease (GERD), pancreatitis, and chronic liver disease (CLD) often suffer from obesity because of coincidence (IBD, IBS, celiac disease) or related pathophysiology (GERD, pancreatitis and CLD). It is unclear if such patients need a particular diagnostic and treatment that differs from the needs of lean gastrointestinal patients. The present guideline addresses this question according to current knowledge and evidence. OBJECTIVE The present practical guideline is intended for clinicians and practitioners in general medicine, gastroenterology, surgery and other obesity management, including dietitians and focuses on obesity care in patients with chronic gastrointestinal diseases. METHODS The present practical guideline is the shortened version of a previously published scientific guideline developed according to the standard operating procedure for ESPEN guidelines. The content has been re-structured and transformed into flow-charts that allow a quick navigation through the text. RESULTS In 100 recommendations (3× A, 33× B, 24 × 0, 40× GPP, all with a consensus grade of 90% or more) care of gastrointestinal patients with obesity - including sarcopenic obesity - is addressed in a multidisciplinary way. A particular emphasis is on CLD, especially metabolic associated liver disease, since such diseases are closely related to obesity, whereas liver cirrhosis is rather associated with sarcopenic obesity. A special chapter is dedicated to obesity care in patients undergoing bariatric surgery. The guideline focuses on adults, not on children, for whom data are scarce. Whether some of the recommendations apply to children must be left to the judgment of the experienced pediatrician. CONCLUSION The present practical guideline offers in a condensed way evidence-based advice how to care for patients with chronic gastrointestinal diseases and concomitant obesity, an increasingly frequent constellation in clinical practice.
Collapse
Affiliation(s)
- Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Johann Ockenga
- Medizinische Klinik II, Klinikum Bremen-Mitte, Bremen FRG, Bremen, Germany.
| | - Ahad Eshraghian
- Department of Gastroenterology and Hepatology, Avicenna Hospital, Shiraz, Iran.
| | - Rocco Barazzoni
- Department of Medical, Technological and Translational Sciences, University of Trieste, Ospedale di Cattinara, Trieste, Italy.
| | - Luca Busetto
- Department of Medicine, University of Padova, Padova, Italy.
| | - Marjo Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, the Netherlands.
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Irit Chermesh
- Department of Gastroenterology, Rambam Health Care Campus, Affiliated with Technion-Israel Institute of Technology, Haifa, Israel.
| | - Haluk Tarik Kani
- Department of Gastroenterology, Marmara University, School of Medicine, Istanbul, Turkey.
| | - Wafaa Khannoussi
- Hepato-Gastroenterology Department, Mohammed VI University Hospital, Oujda, Morocco; and Laboratoire de Recherche des Maladies Digestives (LARMAD), Mohammed the First University, Oujda, Morocco.
| | - Laurence Lacaze
- Department of General Surgery, Mantes-la-Jolie Hospital, Mantes-la-Jolie, France.
| | - Miguel Léon-Sanz
- Department of Endocrinology and Nutrition, University Hospital Doce de Octubre, Medical School, University Complutense, Madrid, Spain.
| | - Juan M Mendive
- La Mina Primary Care Academic Health Centre, Catalan Institute of Health (ICS), University of Barcelona, Barcelona, Spain.
| | - Michael W Müller
- Department of General and Visceral Surgery, Regionale Kliniken Holding, Kliniken Ludwigsburg-Bietigheim gGmbH, Krankenhaus Bietigheim, Bietigheim-Bissingen, Germany.
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet & Department of Surgery, Ersta Hospital, Stockholm, Sweden.
| | - Darija Vranesic Bender
- Unit of Clinical Nutrition, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Arved Weimann
- Department of General, Visceral and Oncological Surgery, St. George Hospital, Leipzig, Germany.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
31
|
Dhopatkar N, Keeler JL, Mutwalli H, Whelan K, Treasure J, Himmerich H. Gastrointestinal symptoms, gut microbiome, probiotics and prebiotics in anorexia nervosa: A review of mechanistic rationale and clinical evidence. Psychoneuroendocrinology 2023; 147:105959. [PMID: 36327759 DOI: 10.1016/j.psyneuen.2022.105959] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/22/2022] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Recent research has revealed the pivotal role that the gut microbiota might play in psychiatric disorders. In anorexia nervosa (AN), the gut microbiota may be involved in pathophysiology as well as in the gastrointestinal (GI) symptoms commonly experienced. This review collates evidence for the potential role of gut microbiota in AN, including modulation of the immune system, the gut-brain axis and GI function. We examined studies comparing gut microbiota in AN with healthy controls as well as those looking at modifications in gut microbiota with nutritional treatment. Changes in energy intake and nutritional composition influence gut microbiota and may play a role in the evolution of the gut microbial picture in AN. Additionally, some evidence indicates that pre-morbid gut microbiota may influence risk of developing AN. There appear to be similarities in gut microbial composition, mechanisms of interaction and GI symptoms experienced in AN and other GI disorders such as inflammatory bowel disease and functional GI disorders. Probiotics and prebiotics have been studied in these disorders showing therapeutic effects of probiotics in some cases. Additionally, some evidence exists for the therapeutic benefits of probiotics in depression and anxiety, commonly seen as co-morbidities in AN. Moreover, preliminary evidence for the use of probiotics in AN has shown positive effects on immune modulation. Based on these findings, we discuss the potential therapeutic role for probiotics in ameliorating symptoms in AN.
Collapse
Affiliation(s)
- Namrata Dhopatkar
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham BR3 3BX, UK.
| | - Johanna Louise Keeler
- Section of Eating Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK.
| | - Hiba Mutwalli
- Section of Eating Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK.
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London SE1 9NH, UK.
| | - Janet Treasure
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham BR3 3BX, UK; Section of Eating Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK.
| | - Hubertus Himmerich
- South London and Maudsley NHS Foundation Trust, Bethlem Royal Hospital, Monks Orchard Road, Beckenham BR3 3BX, UK; Section of Eating Disorders, Department of Psychological Medicine, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK.
| |
Collapse
|
32
|
Nordin E, Brunius C, Landberg R, Hellström PM. FODMAPs-Do they really affect IBS symptoms? Front Med (Lausanne) 2023; 10:1123576. [PMID: 36936224 PMCID: PMC10017764 DOI: 10.3389/fmed.2023.1123576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Affiliation(s)
- Elise Nordin
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
- *Correspondence: Elise Nordin
| | - Carl Brunius
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Rikard Landberg
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Per M. Hellström
- Department of Medical Sciences, Gastroenterology/Hepatology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
33
|
Mysonhimer AR, Holscher HD. Gastrointestinal Effects and Tolerance of Nondigestible Carbohydrate Consumption. Adv Nutr 2022; 13:2237-2276. [PMID: 36041173 PMCID: PMC9776669 DOI: 10.1093/advances/nmac094] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/11/2022] [Accepted: 08/25/2022] [Indexed: 01/29/2023] Open
Abstract
Nondigestible carbohydrates (NDCs) are food components, including nonstarch polysaccharides and resistant starches. Many NDCs are classified as dietary fibers by the US FDA. Because of their beneficial effects on human health and product development, NDCs are widely used in the food supply. Although there are dietary intake recommendations for total dietary fiber, there are no such recommendations for individual NDCs. NDCs are heterogeneous in their chemical composition and physicochemical properties-characteristics that contribute to their tolerable intake levels. Guidance on tolerable intake levels of different NDCs is needed because overconsumption can lead to undesirable gastrointestinal side effects, further widening the gap between actual and suggested fiber intake levels. In this review, we synthesize the literature on gastrointestinal effects of NDCs that the FDA accepts as dietary fibers (β-glucan, pectin, arabinoxylan, guar gum, alginate, psyllium husk, inulin, fructooligosaccharides and oligofructose, galactooligosaccharides, polydextrose, cellulose, soy fiber, resistant maltodextrin/dextrin) and present tolerable intake dose recommendations for their consumption. We summarized the findings from 103 clinical trials in adults without gastrointestinal disease who reported gastrointestinal effects, including tolerance (e.g., bloating, flatulence, borborygmi/rumbling) and function (e.g., transit time, stool frequency, stool consistency). These studies provided doses ranging from 0.75-160 g/d and lasted for durations ranging from a single-meal tolerance test to 28 wk. Tolerance was NDC specific; thus, recommendations ranged from 3.75 g/d for alginate to 25 g/d for soy fiber. Future studies should address gaps in the literature by testing a wider range of NDC doses and consumption forms (solid compared with liquid). Furthermore, future investigations should also adopt a standard protocol to examine tolerance and functional outcomes across studies consistently.
Collapse
|
34
|
Algera JP, Törnblom H, Simrén M. Treatments targeting the luminal gut microbiota in patients with irritable bowel syndrome. Curr Opin Pharmacol 2022; 66:102284. [PMID: 36067685 DOI: 10.1016/j.coph.2022.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/07/2022] [Accepted: 08/03/2022] [Indexed: 11/03/2022]
Abstract
Irritable bowel syndrome (IBS) is a common disorder of gut-brain interaction affecting 4% of the world's population. Patients with IBS experience chronic or recurrent abdominal pain in combination with altered bowel habits (diarrhea and/or constipation), and have reduced quality of life. Despite the high prevalence and substantial burden of IBS, its pathophysiology is incompletely understood and remains to be elucidated. The importance of the gut microenvironment has been highlighted in IBS, as there are signs that the gut microbiota of patients differs from healthy controls. Recent studies have aimed to alter the gut microbiota and thereby, attempted to alleviate gastrointestinal symptoms in IBS patients. We highlighted recent advances in common treatments that are targeting the luminal gut microbiota in IBS.
Collapse
Affiliation(s)
- Joost P Algera
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Hans Törnblom
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Simrén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Center for Functional GI & Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
35
|
Chang C, Yuan X, Zhang X, Chen X, Li K. Gastrointestinal Microbiome and Multiple Health Outcomes: Umbrella Review. Nutrients 2022; 14:3726. [PMID: 36145102 PMCID: PMC9505003 DOI: 10.3390/nu14183726] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, there has been growing concern about the impact of the gastrointestinal microbiome on human health outcomes. To clarify the evidence for a link between the gastrointestinal microbiome and a variety of health outcomes in humans, we conducted an all-encompassing review of meta-analyses and systematic reviews that included 195 meta-analyses containing 950 unique health outcomes. The gastrointestinal microbiome is related to mortality, gastrointestinal disease, immune and metabolic outcomes, neurological and psychiatric outcomes, maternal and infant outcomes, and other outcomes. Existing interventions for intestinal microbiota (such as probiotics, fecal microbiota transplant, etc.) are generally safe and beneficial to a variety of human health outcomes, but the quality of evidence is not high, and more detailed and well-designed randomized controlled trials are necessary.
Collapse
Affiliation(s)
- Chengting Chang
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingzhu Yuan
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingxia Zhang
- Department of Organization, West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xinrong Chen
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Ka Li
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| |
Collapse
|
36
|
Thomas A, Thomas A, Butler-Sanchez M. Dietary Modification for the Restoration of Gut Microbiome and Management of Symptoms in Irritable Bowel Syndrome. Am J Lifestyle Med 2022; 16:608-621. [PMID: 36072680 PMCID: PMC9442469 DOI: 10.1177/15598276211012968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/13/2021] [Accepted: 04/08/2021] [Indexed: 08/27/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder leading to chronic debilitating issues. A healthy diet plays an integral role in maintaining the gut microbiota equilibrium, thus promoting digestive health. The structure and function of gut microbiota are affected by genetics and environmental factors, such as altered dietary habits, gastroenteritis, stress, increased use of alcohol and drugs, and medication use. Whereas there are various management approaches cited in the literature to manage symptoms of IBS, the purpose of this article is to focus on dietary options that will restore the gut microbiome and help in managing IBS symptoms. Some of the diets that are discussed in this article include a low-FODMAP (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) diet, gluten-free/wheat-free diet, high-fiber diet, dietary and herbal supplements (psyllium, peppermint oil), and probiotics/prebiotics/synbiotics. The clinical practice guidelines recommended by the American College of Gastroenterology outlines evidence-based dietary recommendations for patients with IBS to manage symptoms. Recent advancements in the dietary management of IBS highlighting the use of a patient-centered, personalized nutrition approach along with lifestyle changes, pharmacological therapies, and psychosocial and behavioral interventions are also reviewed and discussed.
Collapse
Affiliation(s)
- Andrew Thomas
- Bharati Vidyapeeth Medical College, Pune,
India, and University of Illinois Health Sciences System, Chicago,
Illinois
| | - Annie Thomas
- Marcella Niehoff School of Nursing, Loyola
University Chicago, Illinois
| | | |
Collapse
|
37
|
Bischoff SC, Barazzoni R, Busetto L, Campmans‐Kuijpers M, Cardinale V, Chermesh I, Eshraghian A, Kani HT, Khannoussi W, Lacaze L, Léon‐Sanz M, Mendive JM, Müller MW, Ockenga J, Tacke F, Thorell A, Vranesic Bender D, Weimann A, Cuerda C. European guideline on obesity care in patients with gastrointestinal and liver diseases - Joint European Society for Clinical Nutrition and Metabolism / United European Gastroenterology guideline. United European Gastroenterol J 2022; 10:663-720. [PMID: 35959597 PMCID: PMC9486502 DOI: 10.1002/ueg2.12280] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Patients with chronic gastrointestinal (GI) disease such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), celiac disease, gastroesophageal reflux disease (GERD), pancreatitis, and chronic liver disease (CLD) often suffer from obesity because of coincidence (IBD, IBS, celiac disease) or related pathophysiology (GERD, pancreatitis and CLD). It is unclear if such patients need a particular diagnostic and treatment that differs from the needs of lean GI patients. The present guideline addresses this question according to current knowledge and evidence. OBJECTIVE The objective of the guideline is to give advice to all professionals working in the field of gastroenterology care including physicians, surgeons, dietitians and others how to handle patients with GI disease and obesity. METHODS The present guideline was developed according to the standard operating procedure for European Society for Clinical Nutrition and Metabolism guidelines, following the Scottish Intercollegiate Guidelines Network grading system (A, B, 0, and good practice point [GPP]). The procedure included an online voting (Delphi) and a final consensus conference. RESULTS In 100 recommendations (3x A, 33x B, 24x 0, 40x GPP, all with a consensus grade of 90% or more) care of GI patients with obesity - including sarcopenic obesity - is addressed in a multidisciplinary way. A particular emphasis is on CLD, especially fatty liver disease, since such diseases are closely related to obesity, whereas liver cirrhosis is rather associated with sarcopenic obesity. A special chapter is dedicated to obesity care in patients undergoing bariatric surgery. The guideline focuses on adults, not on children, for whom data are scarce. Whether some of the recommendations apply to children must be left to the judgment of the experienced pediatrician. CONCLUSION The present guideline offers for the first time evidence-based advice how to care for patients with chronic GI diseases and concomitant obesity, an increasingly frequent constellation in clinical practice.
Collapse
Affiliation(s)
| | - Rocco Barazzoni
- Department of Medical, Technological and Translational SciencesUniversity of TriesteTriesteItaly
| | - Luca Busetto
- Department of MedicineUniversity of PadovaPadovaItaly
| | - Marjo Campmans‐Kuijpers
- Department of Gastroenterology and HepatologyUniversity Medical Centre GroningenGroningenThe Netherlands
| | - Vincenzo Cardinale
- Department of Medico‐Surgical Sciences and BiotechnologiesSapienza University of RomeRomeItaly
| | - Irit Chermesh
- Department of GastroenterologyRambam Health Care CampusAffiliated with Technion‐Israel Institute of TechnologyHaifaIsrael
| | - Ahad Eshraghian
- Department of Gastroenterology and HepatologyAvicenna HospitalShirazIran
| | - Haluk Tarik Kani
- Department of GastroenterologyMarmara UniversitySchool of MedicineIstanbulTurkey
| | - Wafaa Khannoussi
- Hepato‐Gastroenterology DepartmentMohammed VI University HospitalOujdaMorocco
- Laboratoire de Recherche des Maladies Digestives (LARMAD)Mohammed the First UniversityOujdaMorocco
| | - Laurence Lacaze
- Department of NutritionRennes HospitalRennesFrance
- Department of general surgeryMantes‐la‐Jolie HospitalFrance
- Department of clinical nutritionPaul Brousse‐Hospital, VillejuifFrance
| | - Miguel Léon‐Sanz
- Department of Endocrinology and NutritionUniversity Hospital Doce de OctubreMedical SchoolUniversity ComplutenseMadridSpain
| | - Juan M. Mendive
- La Mina Primary Care Academic Health Centre. Catalan Institute of Health (ICS)University of BarcelonaBarcelonaSpain
| | - Michael W. Müller
- Department of General and Visceral SurgeryRegionale Kliniken HoldingKliniken Ludwigsburg‐Bietigheim gGmbHBietigheim‐BissingenGermany
| | - Johann Ockenga
- Medizinische Klinik IIKlinikum Bremen‐MitteBremenGermany
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCharité Universitätsmedizin BerlinCampus Virchow‐Klinikum and Campus Charité MitteBerlinGermany
| | - Anders Thorell
- Department of Clinical ScienceDanderyds HospitalKarolinska InstitutetStockholmSweden
- Department of SurgeryErsta HospitalStockholmSweden
| | - Darija Vranesic Bender
- Department of Internal MedicineUnit of Clinical NutritionUniversity Hospital Centre ZagrebZagrebCroatia
| | - Arved Weimann
- Department of General, Visceral and Oncological SurgerySt. George HospitalLeipzigGermany
| | - Cristina Cuerda
- Departamento de MedicinaUniversidad Complutense de MadridNutrition UnitHospital General Universitario Gregorio MarañónMadridSpain
| |
Collapse
|
38
|
Gerasimidis K, Gkikas K, Stewart C, Neelis E, Svolos V. Microbiome and paediatric gut diseases. Arch Dis Child 2022; 107:784-789. [PMID: 34716173 DOI: 10.1136/archdischild-2020-320875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/16/2021] [Indexed: 11/04/2022]
Abstract
In the human gut resides a vast community of microorganisms which perform critical functions for the maintenance of whole body homeostasis. Changes in the composition and function of this community, termed microbiome, are believed to provoke disease onset, including non-communicable diseases. In this review, we debate the current evidence on the role of the gut microbiome in the pathogenesis, outcomes and management of paediatric gut disease. We conclude that even though the gut microbiome is altered in paediatric inflammatory bowel disease, coeliac disease, intestinal failure, necrotising enterocolitis and irritable bowel syndrome, there are currently very few implications for unravelling disease pathogenesis or guiding clinical practice. In the future, the gut microbiome may aid in disease differential diagnosis and prediction of clinical outcomes, and comprise a target for therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Christopher Stewart
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Esther Neelis
- Paediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Vaios Svolos
- Human Nutrition, University of Glasgow, Glasgow, UK
| |
Collapse
|
39
|
Iribarren C, Maasfeh L, Öhman L, Simrén M. Modulating the gut microenvironment as a treatment strategy for irritable bowel syndrome: a narrative review. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e7. [PMID: 39295774 PMCID: PMC11406401 DOI: 10.1017/gmb.2022.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/02/2022] [Accepted: 07/26/2022] [Indexed: 09/21/2024]
Abstract
Irritable bowel syndrome (IBS) is a disorder of gut-brain interaction with a complex pathophysiology. Growing evidence suggests that alterations of the gut microenvironment, including microbiota composition and function, may be involved in symptom generation. Therefore, attempts to modulate the gut microenvironment have provided promising results as an indirect approach for IBS management. Antibiotics, probiotics, prebiotics, food and faecal microbiota transplantation are the main strategies for alleviating IBS symptom severity by modulating gut microbiota composition and function (eg. metabolism), gut barrier integrity and immune activity, although with varying efficacy. In this narrative review, we aim to provide an overview of the current approaches targeting the gut microenvironment in order to indirectly manage IBS symptoms.
Collapse
Affiliation(s)
- Cristina Iribarren
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lujain Maasfeh
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Öhman
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Simrén
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Center for Functional GI and Motility Disorders, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
40
|
Bischoff SC, Barazzoni R, Busetto L, Campmans-Kuijpers M, Cardinale V, Chermesh I, Eshraghian A, Kani HT, Khannoussi W, Lacaze L, Léon-Sanz M, Mendive JM, Müller MW, Ockenga J, Tacke F, Thorell A, Vranesic Bender D, Weimann A, Cuerda C. European guideline on obesity care in patients with gastrointestinal and liver diseases - Joint ESPEN/UEG guideline. Clin Nutr 2022; 41:2364-2405. [PMID: 35970666 DOI: 10.1016/j.clnu.2022.07.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Patients with chronic gastrointestinal (GI) disease such as inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), celiac disease, gastroesophageal reflux disease (GERD), pancreatitis, and chronic liver disease (CLD) often suffer from obesity because of coincidence (IBD, IBS, celiac disease) or related pathophysiology (GERD, pancreatitis and CLD). It is unclear if such patients need a particular diagnostic and treatment that differs from the needs of lean GI patients. The present guideline addresses this question according to current knowledge and evidence. OBJECTIVE The objective of the guideline is to give advice to all professionals working in the field of gastroenterology care including physicians, surgeons, dietitians and others how to handle patients with GI disease and obesity. METHODS The present guideline was developed according to the standard operating procedure for ESPEN guidelines, following the Scottish Intercollegiate Guidelines Network (SIGN) grading system (A, B, 0, and good practice point (GPP)). The procedure included an online voting (Delphi) and a final consensus conference. RESULTS In 100 recommendations (3x A, 33x B, 24x 0, 40x GPP, all with a consensus grade of 90% or more) care of GI patients with obesity - including sarcopenic obesity - is addressed in a multidisciplinary way. A particular emphasis is on CLD, especially fatty liver disease, since such diseases are closely related to obesity, whereas liver cirrhosis is rather associated with sarcopenic obesity. A special chapter is dedicated to obesity care in patients undergoing bariatric surgery. The guideline focuses on adults, not on children, for whom data are scarce. Whether some of the recommendations apply to children must be left to the judgment of the experienced pediatrician. CONCLUSION The present guideline offers for the first time evidence-based advice how to care for patients with chronic GI diseases and concomitant obesity, an increasingly frequent constellation in clinical practice.
Collapse
Affiliation(s)
- Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Rocco Barazzoni
- Department of Medical, Technological and Translational Sciences, University of Trieste, Ospedale di Cattinara, Trieste, Italy.
| | - Luca Busetto
- Department of Medicine, University of Padova, Padova, Italy.
| | - Marjo Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, Groningen, the Netherlands.
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Irit Chermesh
- Department of Gastroenterology, Rambam Health Care Campus, Affiliated with Technion-Israel Institute of Technology, Haifa, Israel.
| | - Ahad Eshraghian
- Department of Gastroenterology and Hepatology, Avicenna Hospital, Shiraz, Iran.
| | - Haluk Tarik Kani
- Department of Gastroenterology, Marmara University, School of Medicine, Istanbul, Turkey.
| | - Wafaa Khannoussi
- Hepato-Gastroenterology Department, Mohammed VI University Hospital, Oujda, Morocco; Laboratoire de Recherche des Maladies Digestives (LARMAD), Mohammed the First University, Oujda, Morocco.
| | - Laurence Lacaze
- Department of General Surgery, Mantes-la-Jolie Hospital, Mantes-la-Jolie, France; Department of Clinical Nutrition, Paul-Brousse-Hospital, Villejuif, France.
| | - Miguel Léon-Sanz
- Department of Endocrinology and Nutrition, University Hospital Doce de Octubre, Medical School, University Complutense, Madrid, Spain.
| | - Juan M Mendive
- La Mina Primary Care Academic Health Centre, Catalan Institute of Health (ICS), University of Barcelona, Barcelona, Spain.
| | - Michael W Müller
- Department of General and Visceral Surgery, Regionale Kliniken Holding, Kliniken Ludwigsburg-Bietigheim GGmbH, Krankenhaus Bietigheim, Bietigheim-Bissingen, Germany.
| | - Johann Ockenga
- Medizinische Klinik II, Klinikum Bremen-Mitte, Bremen FRG, Bremen, Germany.
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Berlin, Germany.
| | - Anders Thorell
- Department of Clinical Science, Danderyds Hospital, Karolinska Institutet & Department of Surgery, Ersta Hospital, Stockholm, Sweden.
| | - Darija Vranesic Bender
- Unit of Clinical Nutrition, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Arved Weimann
- Department of General, Visceral and Oncological Surgery, St. George Hospital, Leipzig, Germany.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
41
|
Singh P, Tuck C, Gibson PR, Chey WD. The Role of Food in the Treatment of Bowel Disorders: Focus on Irritable Bowel Syndrome and Functional Constipation. Am J Gastroenterol 2022; 117:947-957. [PMID: 35435179 PMCID: PMC9169760 DOI: 10.14309/ajg.0000000000001767] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022]
Abstract
Irritable bowel syndrome (IBS) and functional constipation (FC) are among the most common disorders of gut-brain interaction, affecting millions of individuals worldwide. Most patients with disorders of gut-brain interaction perceive food as a trigger for their gastrointestinal symptoms, and specific dietary manipulations/advice have now been recognized as a cornerstone therapeutic option for IBS and FC. We discuss in detail the 2 most common dietary interventions used for the management of IBS-general dietary advice based on the National Institute for Health and Care Excellence guidelines and a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs). We summarize the literature around the possible mechanisms of FODMAP-mediated IBS pathophysiology, the current 3-step, top-down approach of administering a low FODMAP diet (LFD) (restriction phase, followed by reintroduction and personalization), the efficacy data of its restriction and personalization phases, and possible biomarkers for response to an LFD. We also summarize the limitations and challenges of an LFD along with the alternative approach to administering an LFD (e.g., bottom-up). Finally, we discuss the available efficacy data for fiber, other dietary interventions (e.g., Mediterranean diet, gluten-free diet, and holistic dietary interventions), and functional foods (e.g., kiwifruit, rhubarb, aloe, and prunes) in the management of IBS and FC.
Collapse
Affiliation(s)
- Prashant Singh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Caroline Tuck
- Department of Dietetics, Nutrition and Sport, La Trobe University, Bundoora, Australia;
| | - Peter R. Gibson
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne Victoria, Australia.
| | - William D. Chey
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
42
|
Abstract
INTRODUCTION Patients with irritable bowel syndrome (IBS) frequently resort to natural products, or request doctors to prescribe them, to relieve their symptoms, due to the poor efficacy and tolerability of several traditional drugs. Products containing fiber are among the most used and their clinical efficacy is discussed here based on the most recent scientific evidence. AREAS COVERED A literature search was carried out to identify the most significant publications in order to deal with the topics of the general characteristics of fibers and the scientific evidence underlying their therapeutic use, the properties of ispaghula husk and the mechanisms by which this product carries out its therapeutic actions. EXPERT OPINION The most recent clinical guidelines on the management of IBS consider ispaghula husk, a product containing soluble fiber, as a reasonable first line therapy for IBS patients with symptoms. In contrast, products containing insoluble fibers, particularly wheat bran, do not appear to be useful in treating IBS symptoms. The clinical data on the use of prebiotics in IBS are still inconclusive. However, low daily amounts of fructo-oligosaccharides or β-galacto-oligosaccharides (also known as trans-galacto-oligosaccharides) may be effective in improving IBS symptoms; further trials are needed to definitively establish their clinical usefulness.
Collapse
Affiliation(s)
- Diego Currò
- Dipartimento Di Sicurezza E Bioetica, Sezione Di Farmacologia, Università Cattolica Del Sacro Cuore - Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| |
Collapse
|
43
|
Gunn D, Abbas Z, Harris HC, Major G, Hoad C, Gowland P, Marciani L, Gill SK, Warren FJ, Rossi M, Remes-Troche JM, Whelan K, Spiller RC. Psyllium reduces inulin-induced colonic gas production in IBS: MRI and in vitro fermentation studies. Gut 2022; 71:919-927. [PMID: 34353864 PMCID: PMC8995815 DOI: 10.1136/gutjnl-2021-324784] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Health-promoting dietary fibre including inulin often triggers gastrointestinal symptoms in patients with IBS, limiting their intake. Our aim was to test if coadministering psyllium with inulin would reduce gas production. DESIGN A randomised, four-period, four-treatment, placebo-controlled, crossover trial in 19 patients with IBS. Subjects ingested a 500 mL test drink containing either inulin 20 g, psyllium 20 g, inulin 20 g+ psyllium 20 g or dextrose 20 g (placebo). Breath hydrogen was measured every 30 min with MRI scans hourly for 6 hours. Faecal samples from a subset of the patients with IBS were tested using an in vitro fermentation model. Primary endpoint was colonic gas assessed by MRI. RESULTS Colonic gas rose steadily from 0 to 6 hours, with inulin causing the greatest rise, median (IQR) AUC(0-360 min) 3145 (848-6502) mL·min. This was significantly reduced with inulin and psyllium coadministration to 618 (62-2345) mL·min (p=0.02), not significantly different from placebo. Colonic volumes AUC(0-360 min) were significantly larger than placebo for both inulin (p=0.002) and inulin and psyllium coadministration (p=0.005). Breath hydrogen rose significantly from 120 min after inulin but not psyllium; coadministration of psyllium with inulin delayed and reduced the maximum increase, AUC(0-360 min) from 7230 (3255-17910) ppm·hour to 1035 (360-4320) ppm·hour, p=0.007.Fermentation in vitro produced more gas with inulin than psyllium. Combining psyllium with inulin did not reduce gas production. CONCLUSIONS Psyllium reduced inulin-related gas production in patients with IBS but does not directly inhibit fermentation. Whether coadministration with psyllium increases the tolerability of prebiotics in IBS warrants further study. TRIAL REGISTRATION NUMBER NCT03265002.
Collapse
Affiliation(s)
- David Gunn
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| | - Zainab Abbas
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| | - Hannah C Harris
- Food, Innovation and Health, Quadram Institute Bioscience, Norwich, UK
| | - Giles Major
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| | - Caroline Hoad
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Penny Gowland
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Sir Peter Mansfield Imaging Centre, University of Nottingham, Nottingham, UK
| | - Luca Marciani
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK.,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| | - Samantha K Gill
- Department of Nutritional Sciences, King's College London, London, UK
| | - Fred J Warren
- Food, Innovation and Health, Quadram Institute Bioscience, Norwich, UK
| | - Megan Rossi
- Department of Nutritional Sciences, King's College London, London, UK
| | | | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, UK
| | - Robin C Spiller
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham, UK .,Nottingham Digestive Diseases Centre, University of Nottingham, Nottingham, UK
| |
Collapse
|
44
|
Kindt S, Louis H, De Schepper H, Arts J, Caenepeel P, De Looze D, Gerkens A, Holvoet T, Latour P, Mahler T, Mokaddem F, Nullens S, Piessevaux H, Poortmans P, Rasschaert G, Surmont M, Vafa H, Van Malderen K, Vanuytsel T, Wuestenberghs F, Tack J. Belgian consensus on irritable bowel syndrome. Acta Gastroenterol Belg 2022; 85:360-382. [PMID: 35709780 DOI: 10.51821/85.2.10100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is characterised by recurrent abdominal pain related to defaecation or associated with altered stool frequency or consistency. Despite its prevalence, major uncertainties in the diagnostic and therapeutic management persist in clinical practice. METHODS A Delphi consensus was conducted by 20 experts from Belgium, and consisted of literature review and voting process on 78 statements. Grading of recommendations, assessment, development and evaluation criteria were applied to evaluate the quality of evidence. Consensus was defined as > 80 % agreement. RESULTS Consensus was reached for 50 statements. The Belgian consensus agreed as to the multifactorial aetiology of IBS. According to the consensus abdominal discomfort also represents a cardinal symptom, while bloating and abdominal distension often coexist. IBS needs subtyping based on stool pattern. The importance of a positive diagnosis, relying on history and clinical examination is underlined, while additional testing should remain limited, except when alarm features are present. Explanation of IBS represents a crucial part of patient management. Lifestyle modification, spasmolytics and water-solube fibres are considered first-line agents. The low FODMAP diet, selected probiotics, cognitive behavioural therapy and specific treatments targeting diarrhoea and constipation are considered appropriate. There is a consensus to restrict faecal microbiota transplantation and gluten-free diet, while other treatments are strongly discouraged. CONCLUSIONS A panel of Belgian gastroenterologists summarised the current evidence on the aetiology, symptoms, diagnosis and treatment of IBS with attention for the specificities of the Belgian healthcare system.
Collapse
Affiliation(s)
- S Kindt
- Department of gastroenterology and Hepatology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussel, Belgium
| | - H Louis
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - H De Schepper
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Antwerp, Belgium
| | - J Arts
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
- Department of Gastroenterology, AZ Sint-Lucas, Brugge, Belgium
| | - P Caenepeel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
- Department of Gastroenterology, Ziekenhuis Oost-Limburg, Campus Sint-Jan, Genk, Belgium
- UHasselt, Hasselt, Belgium
| | - D De Looze
- Department of Gastroenterology and Hepatology, University Hospital Ghent, Gent, Belgium
| | - A Gerkens
- Boitsfort Medical Center, Brussels, Belgium
| | - T Holvoet
- Department of Gastroenterology and Hepatology, University Hospital Ghent, Gent, Belgium
- Department of Gastroenterology, AZ Nikolaas, Sint Niklaas, Belgium
| | - P Latour
- Department of Gastroenterology, Hepatology and Digestive Oncology, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - T Mahler
- Department of Pediatrics, Universitair Ziekenuis Brussel, Brussel, Belgium
| | - F Mokaddem
- Department of Gastroenterology and Hepatology, Vivalia-Centre Sud Luxembourg, Arlon, Belgium
| | - S Nullens
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Antwerp, Belgium
| | - H Piessevaux
- Department of Hepato-gastroenterology, Cliniques universitaires St-Luc, Université catholique de Louvain, Brussels, Belgium
| | - P Poortmans
- Department of gastroenterology and Hepatology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussel, Belgium
| | - G Rasschaert
- Department of gastroenterology and Hepatology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussel, Belgium
| | - M Surmont
- Department of gastroenterology and Hepatology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussel, Belgium
| | - H Vafa
- Department of Gastroenterology and Hepatology, Chirec-Site Delta, Brussels, Belgium
| | - K Van Malderen
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, Antwerp, Belgium
| | - T Vanuytsel
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - F Wuestenberghs
- Department of Gastroenterology and Hepatology, CHU UCL Namur, Université catholique de Louvain, Yvoir, Belgium
| | - J Tack
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
45
|
Sinopoulou V, Gordon M, Limketkai BN, Mullin G, Aali G, Akobeng AK. Prebiotics for induction of remission in ulcerative colitis. Hippokratia 2022. [DOI: 10.1002/14651858.cd015084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Morris Gordon
- School of Medicine; University of Central Lancashire; Preston UK
| | - Berkeley N Limketkai
- Division of Digestive Diseases; University of California Los Angeles; Los Angeles California USA
| | - Gerard Mullin
- Division of Gastroenterology & Hepatology, Department of Medicine; Johns Hopkins University; Baltimore MD USA
| | - Ghazaleh Aali
- School of Medicine; University of Central Lancashire; Preston UK
| | | |
Collapse
|
46
|
Evaluating tolerability of resistant starch 2, alone and in combination with minimally fermented fibre for patients with irritable bowel syndrome: a pilot randomised controlled cross-over trial. J Nutr Sci 2022; 11:e15. [PMID: 35291272 PMCID: PMC8889220 DOI: 10.1017/jns.2022.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Resistant starch 2 (RS2) may offer therapeutic value to irritable bowel syndrome (IBS) patients particularly in combination with minimally fermented fibre, but tolerability data are lacking. The present study evaluated the tolerability of RS2, sugarcane bagasse and their combination in IBS patients and healthy controls. Following baseline, participants consumed the fibres in escalating doses lasting 3 d each: RS2 (10, 15 and 20 g/d); sugarcane bagasse (5, 10 and 15 g/d); and their combination (20, 25 and 30 g/d). Gastrointestinal symptoms were assessed daily. Six IBS patients and five controls were recruited. No differences in overall symptoms from baseline were found across the fibre doses (IBS, P = 0⋅586; controls, P = 0⋅687). For IBS patients, all RS2 doses led to increased bloating. One IBS patient did not tolerate the low combination dose and another the high sugarcane bagasse dose. Supplementation of RS2 ≤ 20 g/d caused mild symptoms and was generally tolerated in IBS patients even when combined with minimally fermented fibre.
Collapse
|
47
|
Prebiotic Galacto-Oligosaccharides Impact Stool Frequency and Fecal Microbiota in Self-Reported Constipated Adults: A Randomized Clinical Trial. Nutrients 2022; 14:nu14020309. [PMID: 35057489 PMCID: PMC8780623 DOI: 10.3390/nu14020309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Constipation is a major issue for 10-20% of the global population. In a double-blind randomized placebo-controlled clinical trial, we aimed to determine a dose-response effect of galacto-oligosaccharides (GOS) on stool characteristics and fecal microbiota in 132 adults with self-reported constipation according to Rome IV criteria (including less than three bowel movements per week). Subjects (94% females, aged: 18-59 years) received either 11 g or 5.5 g of BiotisTM GOS, or a control product, once daily for three weeks. Validated questionnaires were conducted weekly to study primarily stool frequency and secondary stool consistency. At base- and endline, stool samples were taken to study fecal microbiota. A trend towards an increased stool frequency was observed after the intervention with 11 g of GOS compared to control. While during screening everybody was considered constipated, not all subjects (n = 78) had less than three bowel movements per week at baseline. In total, 11 g of GOS increased stool frequency compared to control in subjects with a low stool frequency at baseline (≤3 bowel movements per week) and in self-reported constipated adults 35 years of age or older. A clear dose-response of GOS was seen on fecal Bifidobacterium, and 11 g of GOS significantly increased Anaerostipes hadrus. In conclusion, GOS seems to be a solution to benefit adults with a low stool frequency and middle-aged adults with self-reported constipation.
Collapse
|
48
|
Rastgoo S, Ebrahimi-Daryani N, Agah S, Karimi S, Taher M, Rashidkhani B, Hejazi E, Mohseni F, Ahmadzadeh M, Sadeghi A, Hekmatdoost A. Glutamine Supplementation Enhances the Effects of a Low FODMAP Diet in Irritable Bowel Syndrome Management. Front Nutr 2022; 8:746703. [PMID: 34977110 PMCID: PMC8716871 DOI: 10.3389/fnut.2021.746703] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Although irritable bowel syndrome is one of the most common gastrointestinal disorders presented to gastroenterologists, therapeutic strategies are not yet well-established. Accordingly, we conducted a randomized, double-blind, placebo-controlled, clinical trial to evaluate the possible superiority of adding glutamine supplement to low fermentable oligo- di- monosaccharides and polyols (FODMAP) diet in patients with irritable bowel syndrome (IBS). Methods: Eligible adults were randomized to receive a low FODMAP diet either with glutamine (15 g/day) or a placebo for 6 weeks. The primary endpoint was a significant reduction in IBS-symptom severity score (IBS-SSS). Secondary endpoints were changes in IBS symptoms, stool frequency, consistency, and quality of life. Results: The study group enrolled 50 patients, among which 22 participants from each group completed the study protocol. The glutamine group had significant changes in total IBS-severity score, dissatisfaction of bowel habit and interference with community function (58% reduction; P < 0.001, 57% reduction; P < 0.001, 51% reduction; P = 0.043, respectively). Improvement in IBS-severity score of more than 45% was observed in 22 of 25 participants (88%) in the glutamine group, while it was only 15 of 25 participants (60%) in the control group (p = 0.015). No serious adverse events were observed. Conclusions: Our findings indicated the superiority of adding glutamine supplementation to a low FODMAP diet in amelioration of IBS symptoms while confirming the beneficial effects of a low FODMAP diet in IBS management.
Collapse
Affiliation(s)
- Samira Rastgoo
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasser Ebrahimi-Daryani
- Department of Gastroenterology and Hepatology, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Agah
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Karimi
- Department of Gastroenterology and Hepatology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taher
- Department of Gastroenterology and Hepatology, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Rashidkhani
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Hejazi
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mohseni
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Ahmadzadeh
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azita Hekmatdoost
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, National Nutrition and Food Technology, Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Abstract
Milk proteins are known for their high nutritional quality, based on their essential amino acid composition, and they exhibit a wide range of bioactivities, including satiety, antimicrobial, mineral-binding, and anti-lipidemic properties. Because of their unique water solubility, milk proteins are readily separated into casein and whey fractions, which can be further fractionated into many individual proteins, including alpha-S1- and alpha-S2-caseins, beta-casein, and kappa-casein, and the whey proteins alpha-lactalbumin, lactoferrin, beta-lactoglobulin, and glycomacropeptide. Many of these proteins have unique bioactivities. Further, over the past 30 years, peptides that are encrypted in the primary amino acid sequences of proteins and released along with amino acids during digestion are increasingly recognized as biologically active protein metabolites that may have beneficial effects on human health. This review examines the current state of the science on the contribution of dairy proteins and their unique peptides and amino acids to human health.
Collapse
Affiliation(s)
| | - Donald K Layman
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
50
|
So D, Gibson PR, Muir JG, Yao CK. Dietary fibres and IBS: translating functional characteristics to clinical value in the era of personalised medicine. Gut 2021; 70:2383-2394. [PMID: 34417199 DOI: 10.1136/gutjnl-2021-324891] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022]
Abstract
Clinical guidelines in the use of fibre supplementation for patients with IBS provide one-size-fits-all advice, which has limited value. This narrative review addresses data and concepts around the functional characteristics of fibre and subsequent physiological responses induced in patients with IBS with a view to exploring the application of such knowledge to the precision use of fibre supplements. The key findings are that first, individual fibres elicit highly distinct physiological responses that are associated with their functional characteristics rather than solubility. Second, the current evidence has focused on the use of fibres as a monotherapy for IBS symptoms overall without attempting to exploit these functional characteristics to elicit specific, symptom-targeted effects, or to use fibre types as adjunctive therapies. Personalisation of fibre therapies can therefore target several therapeutic goals. Proposed goals include achieving normalisation of bowel habit, modulation of gut microbiota function towards health and correction of microbial effects of other dietary therapies. To put into perspective, bulking fibres that are minimally fermented can offer utility in modulating indices of bowel habit; slowly fermented fibres may enhance the activities of the gut microbiota; and the combination of both fibres may potentially offer both benefits while optimising the activities of the microbiota throughout the different regions of the colon. In conclusion, understanding the GI responses to specific fibres, particularly in relation to the physiology of the individual, will be the future for personalising fibre therapy for enhancing the personalised management of patients with IBS.
Collapse
Affiliation(s)
- Daniel So
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Jane G Muir
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Chu K Yao
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|