1
|
Gerasimidis K. Nutrition and dietary therapy in paediatric inflammatory bowel disease. Clin Nutr ESPEN 2025; 67:233-241. [PMID: 40064235 DOI: 10.1016/j.clnesp.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Affiliation(s)
- Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, G31 2ER, Glasgow, UK.
| |
Collapse
|
2
|
Bargas A, Palmela C, Glória L. Enteral Nutrition in Crohn's Disease: A Comprehensive Review of Its Role in Induction and Maintenance of Remission and Perioperative Management in Adult Patients. Nutrients 2025; 17:1481. [PMID: 40362790 PMCID: PMC12073377 DOI: 10.3390/nu17091481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/23/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disorder frequently associated with significant nutritional deficiencies. Enteral nutrition (EN), particularly exclusive enteral nutrition (EEN), has gained recognition not only for its nutritional support but also for its therapeutic potential in reducing intestinal inflammation. While EEN is well established in pediatric populations, its application in adults remains limited due to lower adherence and palatability challenges. Nonetheless, emerging evidence supports its efficacy in various clinical settings, including as an adjunct to pharmacologic therapies and in mitigating pre- and postoperative disease burden. The heterogeneity of study designs, formula compositions, and clinical protocols underscores the need for standardized guidelines and personalized approaches. This narrative review synthesizes the current evidence on the role of EN in adult patients with CD, with a focus on its use for induction and maintenance of remission, as well as perioperative optimization.
Collapse
Affiliation(s)
- André Bargas
- Gastroenterology Service, Hospital Beatriz Ângelo, 2670-433 Loures, Portugal
| | | | | |
Collapse
|
3
|
Day AS, Ballard TM, Yao CK, Gibson PR, Bryant RV. Food-Based Interventions as Therapy for Inflammatory Bowel Disease: Important Steps in Diet Trial Design and Reporting of Outcomes. Inflamm Bowel Dis 2025; 31:1121-1137. [PMID: 39177975 DOI: 10.1093/ibd/izae185] [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: 01/16/2024] [Indexed: 08/24/2024]
Abstract
Diet therapy for inflammatory bowel disease (IBD) is an international research priority but guidance for IBD-specific diet trial design is lacking. This review critically evaluates key elements of prospective IBD food-based intervention trials and identifies gaps. Electronic databases were searched for interventional IBD diet studies. Prospective primary studies/trials were included if used food-based dietary strategies. Forty studies/trials evaluating 29 food-based strategies as therapy for IBD were identified. Considerable heterogeneity in diets, trial design, and methodology exists. Thirty-one trials (78%) intended the diet to modulate inflammation but 14/31 (46%) did not have a primary endpoint measuring an objective change in inflammatory activity and 20/31 (65%) controlled for medication stability prior to application of diet at baseline. Higher-quality IBD diet trials used symptom-based assessment tools coupled with an objective evaluation of inflammatory activity. Dietary advice trials are the most common. One-third of trials developed and administered diet education without a dietitian. Evaluation and reporting on adherence to diet therapy occurred in <60% of trials. Failure to include or report on key elements of trial design reduced the interpretability and validity of the results. This is a considerable limitation to advancing scientific knowledge in this area. Diet therapy trials should adhere to similar rigorous quality standards used to develop other IBD therapies. Therefore, a set of practical recommendations was generated to provide the authors' perspective to help inform the future design of high-quality IBD diet trials.
Collapse
Affiliation(s)
- Alice S Day
- Department of Gastroenterology and Hepatology, Inflammatory Bowel Disease Services, The Queen Elizabeth Hospital, 28 Woodville Road, Woodville South 5011, South Australia, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Frome Road, Adelaide 5000, South Australia, Australia
- Inflammatory Bowel Disease Research Group, Basil Hetzel Institute, 33 Woodville Road, Woodville South 5011, South Australia, Australia
| | - Tessa M Ballard
- Department of Gastroenterology and Hepatology, Inflammatory Bowel Disease Services, The Queen Elizabeth Hospital, 28 Woodville Road, Woodville South 5011, South Australia, Australia
- Discipline of Nutrition and Dietetics, College of Nursing and Health Sciences, Flinders University, GPO Box 2100, Adelaide 5001, South Australia, Australia
| | - Chu K Yao
- Department of Gastroenterology, Central Clinical School, Monash University & Alfred Hospital, 99 Commercial Road, Melbourne 3004, Victoria, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Central Clinical School, Monash University & Alfred Hospital, 99 Commercial Road, Melbourne 3004, Victoria, Australia
| | - Robert V Bryant
- Department of Gastroenterology and Hepatology, Inflammatory Bowel Disease Services, The Queen Elizabeth Hospital, 28 Woodville Road, Woodville South 5011, South Australia, Australia
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Frome Road, Adelaide 5000, South Australia, Australia
- Inflammatory Bowel Disease Research Group, Basil Hetzel Institute, 33 Woodville Road, Woodville South 5011, South Australia, Australia
| |
Collapse
|
4
|
Fitzpatrick JA, Gibson PR, Taylor KM, Anderson EJ, Friedman AB, Ardalan ZS, Smith RL, Halmos EP. Clinical Trial: The Effects of Emulsifiers in the Food Supply on Disease Activity in Crohn's Disease: An Exploratory Double-Blinded Randomised Feeding Trial. Aliment Pharmacol Ther 2025; 61:1276-1289. [PMID: 39967287 PMCID: PMC11950802 DOI: 10.1111/apt.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/29/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Advice to avoid dietary emulsifiers in Crohn's disease (CD) is based on preclinical data. AIMS To examine the effect of diets high (HED) and low (LED) in emulsifiers in the food supply on disease activity in CD. METHODS In a double-blinded, randomised feeding study, we randomised adults with symptomatic, sonographically active CD with ileal involvement on ≥ 2 months' stable medical therapy to 4 weeks of a HED or LED modelled on Australian healthy eating guidelines. We measured the Harvey-Bradshaw Index (HBI), sonographic indices (IBUS-SAS, bowel wall thickness), quality of life (QOL) and fatigue at baseline and study completion. RESULTS We randomised 24 patients, mean age 37 (95% CI 32, 41) years, 12 male, HBI 6 (6, 8), bowel wall thickness 6.0 (5.5-6.6) mm. Adherence was > 95%. Clinical remission (HBI < 5) occurred in 9/12 on HED and 7/12 on LED; 2 and 3, respectively, withdrew early with increasing gastrointestinal symptoms. IBUS-SAS fell from 51 (35, 68) to 33 (15, 51) on HED (p = 0.014) and from 57 (38, 76) to 44 (29, 59) on LED (p = 0.01). Bowel wall thickness reduced by 34% on HED and 15% on LED in those who completed the study. QOL and fatigue improved on both diets (p ≤ 0.05). There were no statistically significant differences in outcomes between diets. CONCLUSIONS In the context of a healthy diet, the emulsifier content had no influence over disease activity over 4 weeks in patients with CD. Recommendations to avoid emulsifiers in patients with active CD are not supported. Australian New Zealand Clinical Trials Registry (ACTRN12619001099112).
Collapse
Affiliation(s)
- Jessica A. Fitzpatrick
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Peter R. Gibson
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Kirstin M. Taylor
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Ellen J. Anderson
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Antony B. Friedman
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Zaid S. Ardalan
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Rebecca L. Smith
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| | - Emma P. Halmos
- Department of GastroenterologySchool of Translational Medicine, Monash University and Alfred HealthMelbourneVictoriaAustralia
| |
Collapse
|
5
|
Russell RK, Fagbemi A, Benyacoub J, Capobianco ME, Wells LE, Shergill-Bonner R, Sharma P, Patel M. Specialized and standard nutritional formulas for the dietary management of pediatric patients with Crohn's disease: a systematic literature review. Expert Rev Gastroenterol Hepatol 2025; 19:455-465. [PMID: 40198155 DOI: 10.1080/17474124.2025.2488887] [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: 11/15/2024] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION This systematic literature review (SLR) aims to compare the clinical, humanistic, and economic outcomes associated with specialized and standard nutritional formulas for the treatment of mild-to-moderate pediatric Crohn's disease. METHODS Search strategies were applied across MEDLINE, Cochrane and Web of Science (January 2000-October 2023) and recent congress proceedings (January 2021-October 2023). PRISMA-P guidelines were followed. Quality assessment evaluated risk of bias. RESULTS Twenty-three unique studies met the inclusion criteria. Nineteen studies (754 patients) evaluated specialized formula, 10 assessed standard formula (246 patients). Mucosal healing (7 studies), induction (20 studies) and maintenance of remission (9 studies) were reported over various timeframes. High proportions of patients who received specialized formula achieved mucosal healing (63-89% 8 weeks; 25-74% 10 weeks), and remission (50-100% 8 weeks). Specialized formula sustained remission (34-62.5% 6 months and 24-87.5% 1 year). Results were not directly comparable with standard formula due to significant heterogeneity in study methodology, patient populations, and remission definition. CONCLUSIONS The evidence predominantly supports the benefits of specialized formula in inducing mucosal healing, remission, and sustaining positive outcomes across multiple timepoints. Direct comparison of nutritional interventions is required to further support the findings of this SLR.Protocol registration: PROSPERO CRD42023472370.
Collapse
Affiliation(s)
- Richard K Russell
- Department of Paediatric Gastroenterology, Clinical Staff Offices, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Andrew Fagbemi
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jalil Benyacoub
- Medical Affairs, Pediatric Medical Nutrition Nestle Health Science, Vevey, Vaud, Switzerland
| | - Maria E Capobianco
- Greenway House Larkwood Way, Valid Insight Ltd, Bioscript Group Ltd, Macclesfield, UK
| | - Laura E Wells
- Greenway House Larkwood Way, Valid Insight Ltd, Bioscript Group Ltd, Macclesfield, UK
| | - Rita Shergill-Bonner
- Department of Paediatric Gastroenterology, Evelina London Children's Hospital, London, UK
| | - Preeti Sharma
- Medical Affairs, Pediatric Medical Nutrition Nestle Health Science, Vevey, Vaud, Switzerland
| | - Minal Patel
- Department Nutrition and Dietetics, Bart's Health NHS Trust, London, UK
| |
Collapse
|
6
|
Zhu W, Zhang Y, Wang LDL, Li J, Hou S. Factors influencing food-related quality of life in patients with inflammatory bowel disease: A systematic review. J Eval Clin Pract 2025; 31:e14133. [PMID: 39234630 DOI: 10.1111/jep.14133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/23/2024] [Accepted: 08/19/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND People diagnosed with Inflammatory bowel disease (IBD) often have severe dietary restrictions and avoidance due to the uncertainty of intestinal symptoms. Inadequate dietary intake may increase the risk of malnutrition and result in impaired food-related quality of life (FRQoL). Few studies investigated factors influencing FRQoL in patients with IBD. This study aimed to synthesize the existing evidence regarding FRQoL among patients with IBD, including the current situation, measurement instruments, and related influencing factors. METHOD The comprehensive literature search was conducted in databases including PubMed, Embase, CINAHL, PsycInfo, Cochrane Library, as well as the most commonly used Chinese databases (CNKI, Wanfang and CBM). Studies published between January 2015 and December 2023 on FRQoL in patients with IBD were included. The Joanna Briggs Institute (JBI) critical appraisal checklist was utilized to evaluate the methodological quality of the selected studies. RESULTS Finally, only five studies met the inclusion criteria were reviewed, including three cross-sectional studies and two case-control studies. The Food-related quality of life-29 Scale (FR-QoL-29) with a total core of 145 was the most used instrument measuring FRQoL in patients with IBD. The mean scores of FRQoL ranged from 69.9 to 102.3 in adult patients with IBD and 94.3 in children. A diverse range of factors were associated with FRQoL, including socio-demographic, clinical, psychological, diet-related, and nutrient intake factors. CONCLUSIONS The main influencing factors of FRQoL among patients with IBD included disease activity status, severe symptoms, history of IBD surgery, negative emotion reaction, and restrictive eating behaviour. Clinicians should work in concert with dietitians, keeping an eye on the disease status, nutrition intake, and restrictive eating habits, assessing FRQoL and providing personalized dietary recommendation for the patients with IBD.
Collapse
Affiliation(s)
- Wenli Zhu
- School of Public Health, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Yan Zhang
- School of Nursing, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Linda Dong-Ling Wang
- Institute of Translational Medicine, Medical College of Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jiajia Li
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Sicong Hou
- Institute of Translational Medicine, Medical College of Yangzhou University, Yangzhou, Jiangsu Province, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Sigall Boneh R, van der Kruk N, Wine E, Verburgt CM, de Meij TGJ, Löwenberg M, Gecse KB, Wierdsma N, Derikx JPM, de Jonge WJ, D’Haens G, Ghiboub M, Van Limbergen JE. Tryptophan metabolites profile predict remission with dietary therapy in pediatric Crohn's disease. Therap Adv Gastroenterol 2025; 18:17562848251323004. [PMID: 40012837 PMCID: PMC11863242 DOI: 10.1177/17562848251323004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Background Crohn's disease (CD) exclusion diet combined with partial enteral nutrition (CDED + PEN) or exclusive enteral nutrition (EEN) is effective in inducing remission in mild-to-moderate pediatric CD. Although CDED + PEN is better tolerated and has higher compliance compared to EEN, a subset of patients does not achieve remission. Diet-induced remission is shown to be positively associated with specific changes in tryptophan-metabolites. Objectives To investigate whether the abundance of baseline fecal tryptophan-metabolites predicts dietary therapy outcomes in pediatric CD. Design Diagnostic accuracy study and secondary analysis of previously conducted Randomized Controlled Trial (RCT). Methods Twenty-six patients from previously performed RCT of mild-to-moderate pediatric CD were included. The patients were classified as having clinical remission (R) (n = 19 in total; CDED + PEN = 10 and to EEN = 9) or No-Remission (NR) (n = 7 in total; CDED + PEN = 3 and EEN = 4) following 6 weeks of therapy, based on the Pediatric Crohn's Disease Activity Index score (⩽10 = remission). We performed a targeted quantitative analysis of 21 tryptophan-metabolites in baseline (t = 0) fecal samples from both groups, utilizing liquid chromatography coupled with quadrupole mass spectrometry. Receiver operator characteristic curve (ROC) and random forest analysis (RFA) were used to assess the predictive power of fecal tryptophan-metabolites for dietary outcomes at baseline. Ratios of tryptophan-metabolites were compared to investigate different downstream tryptophan pathways. Results Baseline fecal kynurenine level was significantly higher in NR compared to R for CDED + PEN (p = 0.02) and EEN (p = 0.04). ROC analysis highlighted the robust predictive power of kynurenine for CDED + PEN (area under the curve (AUC = 0.97)) and EEN (AUC = 0.88)-induced remission. RFA corroborated these observations. The ratio serotonin/kynurenine was the strongest predictor of CDED + PEN-induced remission (AUC = 1). The ratio 5-hydroxytryptophan/kynurenine (AUC = 0.88) predicted EEN-induced remission. By combining data from CDED + PEN and EEN, kynurenine (AUC = 0.91) and ratios of quinolinic acid/kynurenine (AUC = 0.93) and kynurenine/indole-3-acetic acid (AUC = 0.88) demonstrated strong predictive performance for dietary therapy-induced remission. Conclusion Baseline tryptophan metabolites have the potential to serve as a biomarker for dietary remission in pediatric CD. Some tryptophan metabolite ratios showed the most promising predictive capabilities. If confirmed in validation studies, baseline fecal tryptophan markers may be able to provide much-needed guidance to personalize dietary intervention within the management of pediatric CD. Trial registration NCT01728870.
Collapse
Affiliation(s)
- Rotem Sigall Boneh
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center, Holon, Israel
| | - Nikki van der Kruk
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Eytan Wine
- Division of Pediatric Gastroenterology, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Charlotte M. Verburgt
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Tim G. J. de Meij
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Krisztina B. Gecse
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Nicolette Wierdsma
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Wouter J. de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Department of Surgery, University of Bonn, Bonn, Germany
| | - Geert D’Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Mohammed Ghiboub
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 69, Amsterdam 1105 BK, The Netherlands
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
| | - Johan E. Van Limbergen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, Amsterdam University, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
- Department of Pediatric Gastroenterology and Nutrition, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam University, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Ananthakrishnan AN, Whelan K, Allegretti JR, Sokol H. Diet and Microbiome-Directed Therapy 2.0 for IBD. Clin Gastroenterol Hepatol 2025; 23:406-418. [PMID: 38992408 DOI: 10.1016/j.cgh.2024.05.049] [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: 03/08/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic and relapsing disorders of the gastrointestinal tract, characterized by dysregulated immune responses to the gut microbiome. The gut microbiome and diet are key environmental factors that influence the onset and progression of IBD and can be leveraged for treatment. In this review, we summarize the current evidence on the role of the gut microbiome and diet in IBD pathogenesis, and the potential of microbiome-directed therapies and dietary interventions to improve IBD outcomes. We discuss available data and the advantages and drawbacks of the different approaches to manipulate the gut microbiome, such as fecal microbiota transplantation, next-generation and conventional probiotics, and postbiotics. We also review the use of diet as a therapeutic tool in IBD, including the effects in induction and maintenance, special diets, and exclusive enteral nutrition. Finally, we highlight the challenges and opportunities for the translation of diet and microbiome interventions into clinical practice, such as the need for personalization, manufacturing and regulatory hurdles, and the specificity to take into account for clinical trial design.
Collapse
Affiliation(s)
- Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts.
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, United Kingdom
| | - Jessica R Allegretti
- Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Harry Sokol
- Gastroenterology Department, Centre de Recherche Saint-Antoine, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Institut National de la Santé et de la Recherche Médicale, Sorbonne Université, Paris, France; Paris Center for Microbiome Medicine, Fédération Hospitalo-Univeresitaire, Paris, France; Micalis Institute, AgroParisTech, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
9
|
Deleu S, Becherucci G, Godny L, Mentella MC, Petito V, Scaldaferri F. The Key Nutrients in the Mediterranean Diet and Their Effects in Inflammatory Bowel Disease: A Narrative Review. Nutrients 2024; 16:4201. [PMID: 39683595 DOI: 10.3390/nu16234201] [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: 11/14/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The gut microbiome, a collection of gut microorganisms, is crucial in the development and progression of inflammatory bowel diseases (IBD). Therefore, diet and dietary interventions are promising strategies to shape the gut microbiota for IBD management. Of all the diets studied in the IBD field, the Mediterranean diet has the least restrictive nature, promoting long-term adherence. The Mediterranean diet is rich in plants, with a high daily intake of fruits and vegetables (high in fiber, antioxidants, and vitamins), olive oil, whole grains, legumes, and nuts. It includes the moderate consumption of animal products such as oily fish (rich in mono- and polyunsaturated fatty acids), dairy products, and poultry, with a limited intake of red meat and processed foods. This diet is associated with a decreased risk of chronic diseases, including IBD. However, the mechanisms of specific nutrients behind these effects in the Mediterranean diet remain under investigation. Therefore, in this review, we aim to provide an overview of the nutrients that are abundant in the Mediterranean diet and their effects on IBD, with a main focus on preclinical evidence. While several nutrients like fructo-oligosaccharide, chitosan, plant-derived protein, polyphenols, omega-3 polyunsaturated fatty acids, and resveratrol have shown potential beneficial effects in preclinical models, clinical evidence is often limited. However, understanding the complex interactions between specific nutrients and IBD is essential to developing a tailored, multidisciplinary, and personalized approach for disease management; therefore, further research is required.
Collapse
Affiliation(s)
- Sara Deleu
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Guia Becherucci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lihi Godny
- Division of Gastroenterology and Nutrition Unit, Rabin Medical Center, Petah-Tikva 49100, Israel
| | - Maria Chiara Mentella
- UOC di Nutrizione Clinica, Dipartimento Scienze Mediche e Chirurgiche Addominali ed Endocrino-Metaboliche, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Valentina Petito
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- CEMAD Translational Research Laboratories, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
10
|
Häcker D, Siebert K, Smith BJ, Köhler N, Riva A, Mahapatra A, Heimes H, Nie J, Metwaly A, Hölz H, Manz Q, De Zen F, Heetmeyer J, Socas K, Le Thi G, Meng C, Kleigrewe K, Pauling JK, Neuhaus K, List M, Pollard KS, Schwerd T, Haller D. Exclusive enteral nutrition initiates individual protective microbiome changes to induce remission in pediatric Crohn's disease. Cell Host Microbe 2024; 32:2019-2034.e8. [PMID: 39461337 PMCID: PMC12017801 DOI: 10.1016/j.chom.2024.10.001] [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: 03/28/2024] [Revised: 08/14/2024] [Accepted: 10/01/2024] [Indexed: 10/29/2024]
Abstract
Exclusive enteral nutrition (EEN) is a first-line therapy for pediatric Crohn's disease (CD), but protective mechanisms remain unknown. We established a prospective pediatric cohort to characterize the function of fecal microbiota and metabolite changes of treatment-naive CD patients in response to EEN (German Clinical Trials DRKS00013306). Integrated multi-omics analysis identified network clusters from individually variable microbiome profiles, with Lachnospiraceae and medium-chain fatty acids as protective features. Bioorthogonal non-canonical amino acid tagging selectively identified bacterial species in response to medium-chain fatty acids. Metagenomic analysis identified high strain-level dynamics in response to EEN. Functional changes in diet-exposed fecal microbiota were further validated using gut chemostat cultures and microbiota transfer into germ-free Il10-deficient mice. Dietary model conditions induced individual patient-specific strain signatures to prevent or cause inflammatory bowel disease (IBD)-like inflammation in gnotobiotic mice. Hence, we provide evidence that EEN therapy operates through explicit functional changes of temporally and individually variable microbiome profiles.
Collapse
Affiliation(s)
- Deborah Häcker
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany; TUMCREATE, 1 CREATE way, #10-02 CREATE Tower, Singapore 138602, Singapore
| | - Kolja Siebert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | | | - Nikolai Köhler
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Alessandra Riva
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Aritra Mahapatra
- ZIEL Institute for Food & Health, Technische Universität München, 85354 Freising, Germany
| | - Helena Heimes
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Jiatong Nie
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Amira Metwaly
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Hannes Hölz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | - Quirin Manz
- Data Science in Systems Biology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Federica De Zen
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | - Jeannine Heetmeyer
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | - Katharina Socas
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | - Giang Le Thi
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany
| | - Josch K Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Klaus Neuhaus
- ZIEL Institute for Food & Health, Technische Universität München, 85354 Freising, Germany
| | - Markus List
- Data Science in Systems Biology, School of Life Sciences, Technische Universität München, 85354 Freising, Germany; Munich Data Science Institute (MDSI), Technical University of Munich, 85748 Garching, Germany
| | - Katherine S Pollard
- Gladstone Institutes, San Francisco, CA 94158, USA; University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Tobias Schwerd
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital LMU, 80337 Munich, Germany.
| | - Dirk Haller
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technische Universität München, 85354 Freising, Germany; TUMCREATE, 1 CREATE way, #10-02 CREATE Tower, Singapore 138602, Singapore; ZIEL Institute for Food & Health, Technische Universität München, 85354 Freising, Germany.
| |
Collapse
|
11
|
Magen-Rimon R, Day AS, Shaoul R. An Overview of Nutritional Interventions in Inflammatory Bowel Diseases. Nutrients 2024; 16:3055. [PMID: 39339655 PMCID: PMC11435346 DOI: 10.3390/nu16183055] [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: 08/01/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Food is an important environmental factor in the development of inflammatory bowel diseases, chronic immune-mediated diseases of the gastrointestinal tract. Consequently, there is significant focus on the role that dietary approaches might have in the management of these diseases. The introduction of exclusive enteral nutrition (EEN) as a treatment option for induction of remission in Crohn's disease was a breakthrough in disease pathophysiology understanding and has paved the way for dietary options based on this understanding. This review aims to summarize the current data on the effect of different available diets on disease symptoms and the inflammatory process.
Collapse
Affiliation(s)
- Ramit Magen-Rimon
- Pediatric Gastroenterology & Nutrition Institute, Ruth Children’s Hospital of Haifa, Rambam Health Care Campus, Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525408, Israel;
| | - Andrew S. Day
- Department of Paediatrics, University of Otago Christchurch, Christchurch 8011, New Zealand;
| | - Ron Shaoul
- Pediatric Gastroenterology & Nutrition Institute, Ruth Children’s Hospital of Haifa, Rambam Health Care Campus, Faculty of Medicine, Technion—Israel Institute of Technology, Haifa 3525408, Israel;
| |
Collapse
|
12
|
Harvey A, Mannette J, Sigall-Boneh R, Macintyre B, Parrott M, Cahill L, Connors J, Otley A, Haskett J, van Limbergen J, Grant S. Co-Development of Three Dietary Indices to Facilitate Dietary Intake Assessment of Pediatric Crohn's Disease Patients. CAN J DIET PRACT RES 2024; 85:161-168. [PMID: 38634640 DOI: 10.3148/cjdpr-2024-005] [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] [Indexed: 04/19/2024]
Abstract
Literature on dietary behaviours of the pediatric Crohn's Disease (CD) population and the relationship between dietary intake and CD activity is limited. Three dietary indices were developed and tested to conduct dietary pattern analysis in pediatric patients with CD consuming a free diet following remission induction via exclusive enteral nutrition (n = 11). Index scores underwent descriptive and inferential analysis. The mean adjusted scores (out of 100) for the Pediatric Western Diet Index, Pediatric Prudent Diet Index, and Pediatric-Adapted 2010 Alternate Healthy Eating Index (PA2010-AHEI) were 29.82 ± 15.22, 34.25 ± 15.18, and 51.50 ± 11.69, respectively. The mean Western-to-Prudent ratio was 0.94 ± 0.55. A significant correlation (r = -0.71) and relationship (F[1, 9] = 9.04, P < 0.05, R2 = 0.501) between the Western-to-Prudent ratio and PA2010-AHEI was found. The results suggest participants were not following a Western or Prudent diet, and were consuming foods not captured by the indices. More research is needed to describe dietary intake of individuals with CD, validate dietary indices in diverse samples, and explore the utility of these indices in CD assessment and treatment. The co-authors hope this work will stimulate/inspire subsequent interprofessional, dietitian-led research on this topic.
Collapse
Affiliation(s)
| | | | - Rotem Sigall-Boneh
- The E. Wolfson Medical Center, Pediatric Gastroenterology and Nutrition Unit, Holon, Israel
| | | | | | - Leah Cahill
- Dalhousie University, Halifax, NS
- Queen Elizabeth II Health Sciences Centre, Halifax, NS
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Anthony Otley
- Dalhousie University, Halifax, NS
- The E. Wolfson Medical Center, Pediatric Gastroenterology and Nutrition Unit, Holon, Israel
| | | | - Johan van Limbergen
- IWK Health Centre, Halifax, NS
- Dalhousie University, Halifax, NS
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Shannan Grant
- Mount Saint Vincent University, Halifax, NS
- IWK Health Centre, Halifax, NS
- Dalhousie University, Halifax, NS
| |
Collapse
|
13
|
Jauregui-Amezaga A, Smet A. The Microbiome in Inflammatory Bowel Disease. J Clin Med 2024; 13:4622. [PMID: 39200765 PMCID: PMC11354561 DOI: 10.3390/jcm13164622] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
The management of patients with inflammatory bowel disease (IBD) aims to control inflammation through the use of immunosuppressive treatments that target various points in the inflammatory cascade. However, the efficacy of these therapies in the long term is limited, and they often are associated with severe side effects. Although the pathophysiology of the disease is not completely understood, IBD is regarded as a multifactorial disease that occurs due to an inappropriate immune response in genetically susceptible individuals. The gut microbiome is considered one of the main actors in the development of IBD. Gut dysbiosis, characterised by significant changes in the composition and functionality of the gut microbiota, often leads to a reduction in bacterial diversity and anti-inflammatory anaerobic bacteria. At the same time, bacteria with pro-inflammatory potential increase. Although changes in microbiome composition upon biological agent usage have been observed, their role as biomarkers is still unclear. While most studies on IBD focus on the intestinal bacterial population, recent studies have highlighted the importance of other microbial populations, such as viruses and fungi, in gut dysbiosis. In order to modulate the aberrant immune response in patients with IBD, researchers have developed therapies that target different players in the gut microbiome. These innovative approaches hold promise for the future of IBD treatment, although safety concerns are the main limitations, as their effects on humans remain unknown.
Collapse
Affiliation(s)
- Aranzazu Jauregui-Amezaga
- Department of Gastroenterology and Hepatology, University Hospital Antwerp, 2650 Edegem, Belgium
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Annemieke Smet
- Laboratory of Experimental Medicine and Pediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
14
|
Wands DIF, Gianolio L, Wilson DC, Hansen R, Chalmers I, Henderson P, Gerasimidis K, Russell RK. Nationwide Real-World Exclusive Enteral Nutrition Practice Over Time: Persistence of Use as Induction for Pediatric Crohn's Disease and Emerging Combination Strategy With Biologics. Inflamm Bowel Dis 2024; 30:1258-1263. [PMID: 37619221 DOI: 10.1093/ibd/izad167] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Exclusive enteral nutrition (EEN) is the recommended first-line induction treatment in pediatric patients with active luminal Crohn's disease (CD). We aimed to provide a nationwide overview of evolving EEN practices during an era of increasing biologic use. METHODS We analyzed a prospectively identified nationwide cohort of newly diagnosed pediatric patients with CD in Scotland between January 1, 2015, and June 30, 2022. Patients who received EEN for any indication were divided into 6-monthly epochs and examined over time. Differences during the COVID-19 pandemic (March 16, 2020, to July 19, 2021) were examined. Data were retrospectively collected from electronic medical records: demographics, anthropometrics, concomitant treatments, aspects of EEN administration, and remission/response rates. Descriptive statistics and linear regression were used for analyses. RESULTS A total of 649 patients with CD were identified (63% male; median age 12.6 [interquartile range, 10.8-14.8] years); 497 (77%) of 649 received EEN as postdiagnosis induction therapy with a median course length of 7.7 (interquartile range, 5.9-8.0) weeks. Including repeat courses, 547 EEN courses were examined. An increasing incidence of CD was observed over time with no significant changes in EEN usage, remission or response rates, nasogastric tube usage, or course completion (all P > .05). Increasing use of EEN combined with biologics (combination induction) as first-line induction was observed over time (P < .001). Considering COVID-19, lower rates of EEN usage were observed (P = .008) with no differences in remission, oral administration, and course completion rates (all P > .05). CONCLUSIONS Over the past 7.5 years, except during the COVID-19 pandemic, EEN usage rates have not changed despite an increase in biologic use, although combination induction is an emerging trend.
Collapse
Affiliation(s)
- David I F Wands
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, United Kingdom
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, United Kingdom
| | - Laura Gianolio
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - David C Wilson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, United Kingdom
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, United Kingdom
- Child Health, Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Iain Chalmers
- Department of Paediatric Gastroenterology, Hepatology and Clinical Nutrition, Royal Aberdeen Children's Hospital, Aberdeen, United Kingdom
| | - Paul Henderson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, United Kingdom
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Richard K Russell
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young People, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Fitzpatrick JA, Gibson PR, Taylor KM, Halmos EP. Development of Novel High and Low Emulsifier Diets Based upon Emulsifier Distribution in the Australian Food Supply for Intervention Studies in Crohn's Disease. Nutrients 2024; 16:1922. [PMID: 38931276 PMCID: PMC11206755 DOI: 10.3390/nu16121922] [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: 05/26/2024] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The aims of this study were to develop and evaluate a high/low-emulsifier diet and compare emulsifier content with preclinical studies that have associated Crohn's disease with emulsifiers. METHODS Supermarkets were audited with a seven-day high- (HED) and low-emulsifier diet (LED) meal plan developed. The emulsifier content of food was sought from food manufacturers, compared to acceptable daily intake (ADI), and doses were provided in trials. Nutritional composition analysis was completed. Healthy adults ate these diets for seven days in a randomized single-blinded cross-over feeding study to assess palatability, tolerability, satiety, food variety, dietary adherence, blinding and the ease of following the meal plan via visual analogue scale. RESULTS A database of 1680 foods was created. There was no difference in nutritional content between the HED and LED, except HED had a higher ultra-processed food content (p < 0.001). The HED contained 41 emulsifiers, with 53% of the products able to be quantified for emulsifiers (2.8 g/d), which did not exceed the ADI, was similar to that in observational studies, and was exceeded by doses used in experimental studies. In ten participants, diets were rated similarly in palatability-HED mean 62 (5% CI 37-86) mm vs. LED 68 (54-82) mm-in tolerability-HED 41 (20-61) mm vs. LED 55 (37-73) mm-and in satiety HED 57 (32-81) mm vs. LED 49 (24-73) mm. The combined diets were easy to follow (82 (67-97) mm) with good variety (65 (47-81)) and excellent adherence. CONCLUSION Nutritionally well-matched HED and LED were successfully developed, palatable and well tolerated.
Collapse
Affiliation(s)
- Jessica A. Fitzpatrick
- Department of Gastroenterology, School of Translational Medicine, Monash University, Melbourne 3004, Australia; (P.R.G.); (E.P.H.)
| | - Peter R. Gibson
- Department of Gastroenterology, School of Translational Medicine, Monash University, Melbourne 3004, Australia; (P.R.G.); (E.P.H.)
| | - Kirstin M. Taylor
- Department of Gastroenterology, Alfred Health, Melbourne 3004, Australia
| | - Emma P. Halmos
- Department of Gastroenterology, School of Translational Medicine, Monash University, Melbourne 3004, Australia; (P.R.G.); (E.P.H.)
| |
Collapse
|
16
|
Whelan K, Bancil AS, Lindsay JO, Chassaing B. Ultra-processed foods and food additives in gut health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:406-427. [PMID: 38388570 DOI: 10.1038/s41575-024-00893-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/05/2024] [Indexed: 02/24/2024]
Abstract
Ultra-processed foods (UPFs) and food additives have become ubiquitous components of the modern human diet. There is increasing evidence of an association between diets rich in UPFs and gut disease, including inflammatory bowel disease, colorectal cancer and irritable bowel syndrome. Food additives are added to many UPFs and have themselves been shown to affect gut health. For example, evidence shows that some emulsifiers, sweeteners, colours, and microparticles and nanoparticles have effects on a range of outcomes, including the gut microbiome, intestinal permeability and intestinal inflammation. Broadly speaking, evidence for the effect of UPFs on gut disease comes from observational epidemiological studies, whereas, by contrast, evidence for the effect of food additives comes largely from preclinical studies conducted in vitro or in animal models. Fewer studies have investigated the effect of UPFs or food additives on gut health and disease in human intervention studies. Hence, the aim of this article is to critically review the evidence for the effects of UPF and food additives on gut health and disease and to discuss the clinical application of these findings.
Collapse
Affiliation(s)
- Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, UK.
| | - Aaron S Bancil
- Department of Nutritional Sciences, King's College London, London, UK
| | - James O Lindsay
- Blizard Institute, Queen Mary University of London, Barts and the London School of Medicine, London, UK
| | | |
Collapse
|
17
|
Jatkowska A, Gkikas K, Nichols B, Short B, Rizou VK, Kapranos P, Gunnewiek JK, Christina E, Svolos V, Quince C, Gerasimidis K. Dose-dependent effects of enteral nutrition on the faecal microbiota and short chain fatty acids. Clin Nutr 2024; 43:1200-1207. [PMID: 38615449 DOI: 10.1016/j.clnu.2024.04.010] [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: 01/17/2024] [Revised: 03/20/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Enteral nutrition (EN) involves replacing all or part of a person's habitual diet with a nutritional formula. The impact of varying doses of EN on the gut microbiome remains understudied. METHODS Healthy adults replaced all (100% EN) or part (85% EN, 50% EN and 20% EN) of their energy requirements with EN for 7 days. Faecal samples were collected before and on day 7 of interventions. Faecal pH, short chain fatty acids (SCFAs), branched-chain fatty acids (BCFAs) and 16S rRNA sequencing were performed. Dietary assessment was performed with 7-day food diaries. RESULTS Sixty-one participants (31 females; median (IQR) age: 24.7 (23.0-27.8) years) were recruited. A dose-dependent impact of EN on faecal microbiota, SCFAs, BCFAs) and pH was observed, with changes detectable at EN intakes of at least 50% of energy requirements. 100% and 85% EN reduced the abundance of fibre-fermenting taxa such as Agathobacter, Faecalibaterium, Succinivibrio and Acidaminococcus. In parallel, potentially harmful organisms like Eubacterium, Actinomyces, and Klebsiella increased. In the 50% EN group, adherence to a diet high in fish, vegetables, potatoes, non-alcoholic beverages, and fat spreads, and low in cereal products, milk, and meat negatively correlated with changes in microbiota structure (r = -0.75, P = 0.025). This signal was not observed when using compositional tools for microbiota analysis. CONCLUSIONS EN detrimentally influences the faecal microbiota and diet-related bacterial metabolites in a dose-dependent manner, particularly at doses of at least 50%. The findings of this study have implications for the dietary management and counselling of patients receiving high volume EN.
Collapse
Affiliation(s)
- Aleksandra Jatkowska
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Konstantinos Gkikas
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Ben Nichols
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Bryn Short
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | | | - Panagiotis Kapranos
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | | | - Edelyn Christina
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | - Vaios Svolos
- Human Nutrition, School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
18
|
Halmos EP, Godny L, Vanderstappen J, Sarbagili-Shabat C, Svolos V. Role of diet in prevention versus treatment of Crohn's disease and ulcerative colitis. Frontline Gastroenterol 2024; 15:247-257. [PMID: 38665795 PMCID: PMC11042448 DOI: 10.1136/flgastro-2023-102417] [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: 10/04/2023] [Accepted: 12/10/2023] [Indexed: 04/28/2024] Open
Abstract
Diet is a modifiable risk factor for disease course and data over the past decade have emerged to indicate its role in Crohn's disease (CD) and ulcerative colitis (UC). However, literature is riddled with misinterpretation of data, often leading to unexpected or conflicting results. The key understanding is that causative factors in disease development do not always proceed to an opportunity to change disease course, once established. Here, we discuss the data on dietary influences in three distinct disease states for CD and UC-predisease, active disease and quiescent disease. We appraise the literature for how our dietary recommendations should be shaped to prevent disease development and if or how that differs for CD and UC induction therapy and maintenance therapy. In UC, principles of healthy eating are likely to play a role in all states of disease. Conversely, data linking dietary factors to CD prevention and treatment are paradoxical with the highest quality evidence for CD treatment being exclusive enteral nutrition, a lactose, gluten and fibre-free diet comprising solely of ultraprocessed food-all dietary factors that are not associated or inversely associated with CD prevention. High-quality evidence from dietary trials is much awaited to expand our understanding and ultimately lead our dietary recommendations for targeted patient populations.
Collapse
Affiliation(s)
- Emma P Halmos
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Lihi Godny
- Division of Gastroenterology and Nutrition Unit, Rabin Medical Center, Petah Tikva, Israel
| | - Julie Vanderstappen
- Department of Gastroenterology and Hepatology, University Hospitals of Leuven, Leuven, Belgium
| | - Chen Sarbagili-Shabat
- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center, Holon, Israel
- The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Vaios Svolos
- School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Laboratory of Clinical Nutrition and Dietetics, Department of Nutrition and Dietetics, School of Physical Education, Sports Science and Dietetics, University of Thessaly, Trikala, Greece
| |
Collapse
|
19
|
Gkikas K, Svolos V, White B, Gerasimidis K. An update on dietary therapies in paediatric Crohn's disease. Curr Opin Clin Nutr Metab Care 2024; 27:304-312. [PMID: 38456807 DOI: 10.1097/mco.0000000000001024] [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] [Indexed: 03/09/2024]
Abstract
PURPOSE OF REVIEW This article provides a literature update on original articles published in the past 18 months (May 2022-November 2023) in the dietary management of paediatric Crohn's disease. RECENT FINDINGS There is more data to support the use of exclusive enteral nutrition in the management of active Crohn's disease in children. Several food-based dietary therapies have been proposed for the management of Crohn's disease. There is an interest in precision nutritional therapy in Crohn's disease, but current data are scarce. SUMMARY Exclusive enteral nutrition is an effective treatment for paediatric Crohn's disease. Predictors of response to exclusive enteral nutrition include mild disease phenotype and ileal disease involvement, although data remain inconclusive. Adherence to exclusive enteral nutrition is cornerstone to its efficacy. Treatment with exclusive enteral nutrition modifies the gut microbiome, modulates bile acid metabolism and has significant effects on host immune responses. More studies are expected in which drugs need to be combined with dietary therapies and microbial therapeutics. The efficacy of Crohn's disease exclusion diet coupled with partial enteral nutrition is supported by independent studies, but tolerance remains an issue, particularly for long-term disease management. More research is anticipated in precision nutritional therapy in paediatric Crohn's disease, but currently no recommendations can be made.
Collapse
Affiliation(s)
- Konstantinos Gkikas
- Human Nutrition, School of Medicine, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, Glasgow, UK
| | | | | | | |
Collapse
|
20
|
Lee D, Braly K, Nuding M, Braly I, Hopp C, Twible H, Pope C, Hayden HS, Hoffman L, Zheng H, Wahbeh G, Suskind DL. Reverse-engineered exclusive enteral nutrition in pediatric Crohn's disease: A pilot trial. J Pediatr Gastroenterol Nutr 2024; 78:1135-1142. [PMID: 38558411 DOI: 10.1002/jpn3.12196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/18/2024] [Accepted: 03/02/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND In pediatric Crohn's disease (CD), commercial formulas used as exclusive enteral nutrition (EEN) are effective at inducing remission. This study aims to assess the impact of a whole-food blended smoothie as EEN on CD activity and the intestinal microbiome. METHODS A 4-week prospective trial assessed the impact of EEN with a whole-food smoothie on newly diagnosed mild-to-moderate active pediatric CD. The smoothie with a multivitamin were developed to meet age-appropriate nutritional requirements. Assessment over 4 weeks included Pediatric Crohn's Disease Activity Index (PCDAI), serum laboratories, fecal calprotectin (FCP), and stool collection for metagenomic shotgun sequencing and microbiota composition analysis. Clinical remission was defined as PCDAI ≤ 10 at week 4. RESULTS Ten participants were enrolled with median age 14.5 years, and 8 completed the trial. Baseline mean PCDAI was 26.3 ± 9.1 and mean FCP 1149 ± 718 µg/g. At week 4, 80% of participants achieved clinical remission. FCP decreased by over half in 60% of participants, with FCP below 250 µg/g in 60% and below 100 µg/g in 40%. Microbiome analysis showed a significant increase in species richness over 4 weeks (p = 0.01). Compared to baseline, the relative abundance at week 2 and at week 4 was significantly increased for Bifidobacterium and Streptococcus and decreased for Blautia (p < 0.05 for all). CONCLUSION A whole-food blended smoothie was effective for inducing clinical remission and decreasing FCP in pediatric CD similar to commercial EEN formulas. Further research may give insight into data-driven whole-food dietary approaches for CD management. CLINICALTRIALS gov NCT03508193.
Collapse
Affiliation(s)
- Dale Lee
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Kim Braly
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Mason Nuding
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Ian Braly
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA
| | - Courtney Hopp
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Heather Twible
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Christopher Pope
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Hillary S Hayden
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Luke Hoffman
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington
| | - Hengqi Zheng
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - Ghassan Wahbeh
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| | - David L Suskind
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Inflammatory Bowel Disease Center, Seattle Children's Hospital, Seattle, Washington, USA
| |
Collapse
|
21
|
Pereira GV, Boudaud M, Wolter M, Alexander C, De Sciscio A, Grant ET, Trindade BC, Pudlo NA, Singh S, Campbell A, Shan M, Zhang L, Yang Q, Willieme S, Kim K, Denike-Duval T, Fuentes J, Bleich A, Schmidt TM, Kennedy L, Lyssiotis CA, Chen GY, Eaton KA, Desai MS, Martens EC. Opposing diet, microbiome, and metabolite mechanisms regulate inflammatory bowel disease in a genetically susceptible host. Cell Host Microbe 2024; 32:527-542.e9. [PMID: 38513656 PMCID: PMC11064055 DOI: 10.1016/j.chom.2024.03.001] [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: 06/30/2023] [Revised: 12/18/2023] [Accepted: 03/01/2024] [Indexed: 03/23/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic conditions characterized by periods of spontaneous intestinal inflammation and are increasing in industrialized populations. Combined with host genetics, diet and gut bacteria are thought to contribute prominently to IBDs, but mechanisms are still emerging. In mice lacking the IBD-associated cytokine, interleukin-10, we show that a fiber-deprived gut microbiota promotes the deterioration of colonic mucus, leading to lethal colitis. Inflammation starts with the expansion of natural killer cells and altered immunoglobulin-A coating of some bacteria. Lethal colitis is then driven by Th1 immune responses to increased activities of mucin-degrading bacteria that cause inflammation first in regions with thinner mucus. A fiber-free exclusive enteral nutrition diet also induces mucus erosion but inhibits inflammation by simultaneously increasing an anti-inflammatory bacterial metabolite, isobutyrate. Our findings underscore the importance of focusing on microbial functions-not taxa-contributing to IBDs and that some diet-mediated functions can oppose those that promote disease.
Collapse
Affiliation(s)
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Celeste Alexander
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alessandro De Sciscio
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | | | - Nicholas A Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shaleni Singh
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Austin Campbell
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mengrou Shan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Qinnan Yang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stéphanie Willieme
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Kwi Kim
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Trisha Denike-Duval
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Jaime Fuentes
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - André Bleich
- Institute for Laboratory Animal Science, Hanover Medical School, Hanover, Germany
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lucy Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Costas A Lyssiotis
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Kathryn A Eaton
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
22
|
Nichols B, Briola A, Logan M, Havlik J, Mascellani A, Gkikas K, Milling S, Ijaz UZ, Quince C, Svolos V, Russell RK, Hansen R, Gerasimidis K. Gut metabolome and microbiota signatures predict response to treatment with exclusive enteral nutrition in a prospective study in children with active Crohn's disease. Am J Clin Nutr 2024; 119:885-895. [PMID: 38569785 PMCID: PMC11007740 DOI: 10.1016/j.ajcnut.2023.12.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/06/2023] [Accepted: 12/22/2023] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Predicting response to exclusive enteral nutrition (EEN) in active Crohn's disease (CD) could lead to therapy personalization and pretreatment optimization. OBJECTIVES This study aimed to explore the ability of pretreatment parameters to predict fecal calprotectin (FCal) levels at EEN completion in a prospective study in children with CD. METHODS In children with active CD, clinical parameters, dietary intake, cytokines, inflammation-related blood proteomics, and diet-related metabolites, metabolomics and microbiota in feces, were measured before initiation of 8 wk of EEN. Prediction of FCal levels at EEN completion was performed using machine learning. Data are presented with medians (IQR). RESULTS Of 37 patients recruited, 15 responded (FCal < 250 μg/g) to EEN (responders) and 22 did not (nonresponders). Clinical and immunological parameters were not associated with response to EEN. Responders had lesser (μmol/g) butyrate [responders: 13.2 (8.63-18.4) compared with nonresponders: 22.3 (12.0-32.0); P = 0.03], acetate [responders: 49.9 (46.4-68.4) compared with nonresponders: 70.4 (57.0-95.5); P = 0.027], phenylacetate [responders: 0.175 (0.013-0.611) compared with nonresponders: 0.943 (0.438-1.35); P = 0.021], and a higher microbiota richness [315 (269-347) compared with nonresponders: 243 (205-297); P = 0.015] in feces than nonresponders. Responders consumed (portions/1000 kcal/d) more confectionery products [responders: 0.55 (0.38-0.72) compared with nonresponders: 0.19 (0.01-0.38); P = 0.045]. A multicomponent model using fecal parameters, dietary data, and clinical and immunological parameters predicted response to EEN with 78% accuracy (sensitivity: 80%; specificity: 77%; positive predictive value: 71%; negative predictive value: 85%). Higher taxon abundance from Ruminococcaceae, Lachnospiraceae, and Bacteroides and phenylacetate, butyrate, and acetate were the most influential variables in predicting lack of response to EEN. CONCLUSIONS We identify microbial signals and diet-related metabolites in feces, which could comprise targets for pretreatment optimization and personalized nutritional therapy in pediatric CD.
Collapse
Affiliation(s)
- Ben Nichols
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Anny Briola
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Michael Logan
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Jaroslav Havlik
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Anna Mascellani
- Department of Food Science, Czech University of Life Sciences Prague, Prague, Czech Republic
| | - Konstantinos Gkikas
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Simon Milling
- School of Infection and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Umer Zeeshan Ijaz
- Civil Engineering, School of Engineering, University of Glasgow, Glasgow, United Kingdom
| | | | - Vaios Svolos
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Richard K Russell
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People, Edinburgh, United Kingdom
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, United Kingdom; Department of Child Health, Division of Clinical and Molecular Medicine, School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow Royal Infirmary, Glasgow, United Kingdom.
| |
Collapse
|
23
|
Correia I, Oliveira PA, Antunes ML, Raimundo MDG, Moreira AC. Is There Evidence of Crohn's Disease Exclusion Diet (CDED) in Remission of Active Disease in Children and Adults? A Systematic Review. Nutrients 2024; 16:987. [PMID: 38613020 PMCID: PMC11013840 DOI: 10.3390/nu16070987] [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: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Crohn's disease (CD) is an inflammatory bowel disease. Previous research has explored the impact of diet on CD, as specific dietary components can influence gut microbiota and immune responses, contributing to damage in the gastrointestinal tract. The Crohn's Disease Exclusion Diet (CDED) is based on an exclusion diet; it is a recent dietary approach that is often used alongside partial enteral nutrition (PEN) and aims to induce disease remission by excluding certain dietary components. This study assesses the current evidence for the effectiveness of the CDED + PEN in achieving remission in both children and adults with active CD. Our systematic review followed PRISMA recommendations and was registered in PROSPERO with CRD number 42022335076. The searched databases were PubMed/MEDLINE, Cochrane Library, Scopus, and Web of Science. The included studies were analyzed using Rayyan software, and the risk of bias was assessed with Cochrane RevMan 5.0 software. The primary assessed outcome was clinical remission, evaluated with validated questionnaire scores such as PCDAI, CDAI, or HBI. All analyzed papers yielded promising results. Notably, the CDED + PEN demonstrated better tolerance than exclusive enteral nutrition (EEN), resulting in higher adherence rates. Therefore, the CDED + PEN appears to be a viable alternative for induction remission in active disease for both children and adults with CD.
Collapse
Affiliation(s)
- Inês Correia
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; (P.A.O.); (M.L.A.); (M.d.G.R.); (A.C.M.)
- Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
- Hospital do Espírito Santo de Évora, EPE, 7000-811 Évora, Portugal
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal
| | - Patrícia Almeida Oliveira
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; (P.A.O.); (M.L.A.); (M.d.G.R.); (A.C.M.)
| | - Maria Luz Antunes
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; (P.A.O.); (M.L.A.); (M.d.G.R.); (A.C.M.)
- APPsyCI—Applied Psychology Research Center Capabilities & Inclusion, ISPA—Instituto Universitário, 1149-041 Lisboa, Portugal
| | - Maria da Graça Raimundo
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; (P.A.O.); (M.L.A.); (M.d.G.R.); (A.C.M.)
- Hospital do Espírito Santo de Évora, EPE, 7000-811 Évora, Portugal
| | - Ana Catarina Moreira
- ESTeSL—Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal; (P.A.O.); (M.L.A.); (M.d.G.R.); (A.C.M.)
- H&TRC—Health & Technology Research Center, ESTeSL—Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, 1990-096 Lisboa, Portugal
| |
Collapse
|
24
|
Gerasimidis K, Faqerah N, Walker D. Letter: The multifaceted role of Escherichia coli in influencing disorders of the industrial populations-Authors' reply. Aliment Pharmacol Ther 2024; 59:719-720. [PMID: 38349700 DOI: 10.1111/apt.17883] [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: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 02/15/2024]
Abstract
LINKED CONTENTThis article is linked to Faqerah et al papers. To view these articles, visit https://doi.org/10.1111/apt.17720 and https://doi.org/10.1111/apt.17865
Collapse
Affiliation(s)
- Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nojoud Faqerah
- Microbiology, Rabigh Medical College, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Daniel Walker
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
25
|
Gkikas K, Wan M, Svolos V, Nichols B, Hansen R, Russell RK, Gerasimidis K. YouTube as a Source of Information for Food, Diet-Related Items, and Advisory Comments for the Management of Inflammatory Bowel Disease. Inflamm Bowel Dis 2024; 30:347-356. [PMID: 37185900 PMCID: PMC10906362 DOI: 10.1093/ibd/izad076] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Indexed: 05/17/2023]
Abstract
BACKGROUND Patients with inflammatory bowel disease (IBD) often use the Internet to seek information beyond that received from healthcare professionals. This study assessed the perceptions of YouTube presenters on the role of diet in the management of IBD. METHODS Videos discussing dietary aspects (food, diet-related items, and advisory comments [FODRIACs]) in the management of IBD were included. The perceptions of presenters toward each FODRIAC were labeled as positive, negative, or neutral/intermediate, and FODRIACs were classified according to their underlying role in the management of IBD (eg, symptom management, gut inflammation). Subgroup analysis was performed by type of video presenter (patients vs healthcare professionals), type of IBD (Crohn's disease vs ulcerative colitis), and reporting of scientific evidence supporting presenters' perceptions. RESULTS We identified 122 FODRIACs within 160 videos. Patient videos received a higher number of likes (median 85 [interquartile range, 35-156]) than healthcare professional videos (median 44 [interquartile range, 16-1440]) (P = .01). Scientific evidence was cited in 2 (3%) of 76 patient videos compared with 25 (35%) of 71 healthcare professional videos (P < .001). Positive perceptions were expressed about avocadoes, salmon, bananas, white bread, and rice, whereas negative perceptions were reported for processed, high-fat and high-sugar foods and carbonated drinks. Fewer negative perceptions were expressed in videos supported by scientific evidence than in videos that lacked evidence (scientific: 4 positive, 0 negative vs nonscientific: 7 positive, 20 negative; P = .01). CONCLUSIONS We have identified FODRIACs proposed as beneficial or detrimental in the management of IBD. The effect this information has on dietary practice as patients with IBD self-manage their condition needs further exploration.
Collapse
Affiliation(s)
- Konstantinos Gkikas
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Mhairi Wan
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Vaios Svolos
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ben Nichols
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, United Kingdom
| | - Richard K Russell
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children and Young People Edinburgh, Edinburgh, United Kingdom
| | - Konstantinos Gerasimidis
- School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
26
|
Russell EE, Day AS, Dimitroff C, Trakman GL, Silva H, Bryant RV, Purcell L, Yao CK, Landorf E, Fitzpatrick JA. Practical application of the Crohn's disease exclusion diet as therapy in an adult Australian population. J Gastroenterol Hepatol 2024; 39:446-456. [PMID: 38059536 DOI: 10.1111/jgh.16414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 09/27/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023]
Abstract
There is demand from patients and clinicians to use the Crohn's disease exclusion diet (CDED) with or without partial enteral nutrition (PEN). However, the therapeutic efficacy and nutritional adequacy of this therapy are rudimentary in an adult population. This review examines the evidence for the CDED in adults with active luminal Crohn's disease and aims to provide practical guidance on the use of the CDED in Australian adults. A working group of nine inflammatory bowel disease (IBD) dietitians of DECCAN (Dietitians Crohn's and Colitis Australian Network) and an IBD gastroenterologist was established. A literature review was undertaken to examine (1) clinical indications, (2) monitoring, (3) dietary adequacy, (4) guidance for remission phase, and (5) diet reintroduction after therapy. Each diet phase was compared with Australian reference ranges for food groups and micronutrients. CDED with PEN is nutritionally adequate for adults containing sufficient energy and protein and meeting > 80% of the recommended daily intake of key micronutrients. An optimal care pathway for the clinical use of the CDED in an adult population was developed with accompanying consensus statements, clinician toolkit, and patient education brochure. Recommendations for weaning from the CDED to the Australian dietary guidelines were developed. The CDED + PEN provides an alternate partial food-based therapy for remission induction of active luminal Crohn's disease in an adult population. The CDED + PEN should be prioritized over CDED alone and prescribed by a specialist IBD dietitian. DECCAN cautions against using the maintenance diet beyond 12 weeks until further evidence becomes available.
Collapse
Affiliation(s)
- Erin E Russell
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Alice Sarah Day
- Department of Gastroenterology, IBD Service, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Claire Dimitroff
- Department of Nutrition and Dietetics, Austin Health, Melbourne, Victoria, Australia
| | - Gina L Trakman
- Department of Gastroenterology, St Vincent's Hospital Melbourne, Melbourne, Victoria, Australia
- Department of Dietetics, Nutrition and Sport, La Trobe University, Melbourne, Victoria, Australia
| | - Hannah Silva
- Department of Dietetics, Eastern Health, Melbourne, Victoria, Australia
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Robert V Bryant
- Department of Gastroenterology, IBD Service, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Liz Purcell
- Metro South Health, Queensland Health, Brisbane, Queensland, Australia
| | - Chu K Yao
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Emma Landorf
- Department of Nutrition, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Jessica A Fitzpatrick
- Department of Gastroenterology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
27
|
Kuffa P, Pickard JM, Campbell A, Yamashita M, Schaus SR, Martens EC, Schmidt TM, Inohara N, Núñez G, Caruso R. Fiber-deficient diet inhibits colitis through the regulation of the niche and metabolism of a gut pathobiont. Cell Host Microbe 2023; 31:2007-2022.e12. [PMID: 37967555 PMCID: PMC10842462 DOI: 10.1016/j.chom.2023.10.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023]
Abstract
Exclusive enteral nutrition (EEN) with fiber-free diets is an effective steroid-sparing treatment to induce clinical remission in children with Crohn's disease (CD). However, the mechanism underlying the beneficial effects of EEN remains obscure. Using a model of microbiota-dependent colitis with the hallmarks of CD, we find that the administration of a fiber-free diet prevents the development of colitis and inhibits intestinal inflammation in colitic animals. Remarkably, fiber-free diet alters the intestinal localization of Mucispirillum schaedleri, a mucus-dwelling pathobiont, which is required for triggering disease. Mechanistically, the absence of dietary fiber reduces nutrient availability and impairs the dissimilatory nitrate reduction to ammonia (DNRA) metabolic pathway of Mucispirillum, leading to its exclusion from the mucus layer and disease remission. Thus, appropriate localization of the specific pathobiont in the mucus layer is critical for disease development, which is disrupted by fiber exclusion. These results suggest strategies to treat CD by targeting the intestinal niche and metabolism of disease-causing microbes.
Collapse
Affiliation(s)
- Peter Kuffa
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Joseph M Pickard
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Austin Campbell
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Misa Yamashita
- Department of Public Health and Preventive Medicine, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Sadie R Schaus
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Thomas M Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Naohiro Inohara
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Tahiri M, Johnsrud C, Steffensen IL. Evidence and hypotheses on adverse effects of the food additives carrageenan (E 407)/processed Eucheuma seaweed (E 407a) and carboxymethylcellulose (E 466) on the intestines: a scoping review. Crit Rev Toxicol 2023; 53:521-571. [PMID: 38032203 DOI: 10.1080/10408444.2023.2270574] [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: 06/13/2023] [Revised: 10/02/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023]
Abstract
This scoping review provides an overview of publications reporting adverse effects on the intestines of the food additives carrageenan (CGN) (E 407)/processed Eucheuma seaweed (PES) (E 407a) and carboxymethylcellulose (CMC) (E 466). It includes evidence from human, experimental mammal and in vitro research publications, and other evidence. The databases Medline, Embase, Scopus, Web of Science Core Collection, Cochrane Database of Systematic Reviews and Epistemonikos were searched without time limits, in addition to grey literature. The publications retrieved were screened against predefined criteria. From two literature searches, 2572 records were screened, of which 224 records were included, as well as 38 records from grey literature, making a total of 262 included publications, 196 on CGN and 101 on CMC. These publications were coded and analyzed in Eppi-Reviewer and data gaps presented in interactive maps. For CGN, five, 69 and 33 research publications on humans, experimental mammals and in vitro experiments were found, further separated as degraded or native (non-degraded) CGN. For CMC, three human, 20 animal and 14 in vitro research publications were obtained. The most studied adverse effects on the intestines were for both additives inflammation, the gut microbiome, including fermentation, intestinal permeability, and cancer and metabolic effects, and immune effects for CGN. Further studies should focus on native CGN, in the form and molecular weight used as food additive. For both additives, randomized controlled trials of sufficient power and with realistic dietary exposure levels of single additives, performed in persons of all ages, including potentially vulnerable groups, are needed.
Collapse
Affiliation(s)
- Mirlinda Tahiri
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Celine Johnsrud
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Inger-Lise Steffensen
- Department of Food Safety, Division of Climate and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
29
|
Yan D, Ye S, He Y, Wang S, Xiao Y, Xiang X, Deng M, Luo W, Chen X, Wang X. Fatty acids and lipid mediators in inflammatory bowel disease: from mechanism to treatment. Front Immunol 2023; 14:1286667. [PMID: 37868958 PMCID: PMC10585177 DOI: 10.3389/fimmu.2023.1286667] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) is a chronic, relapsing inflammatory disorder of the gastrointestinal tract. Though the pathogenesis of IBD remains unclear, diet is increasingly recognized as a pivotal factor influencing its onset and progression. Fatty acids, essential components of dietary lipids, play diverse roles in IBD, ranging from anti-inflammatory and immune-regulatory functions to gut-microbiota modulation and barrier maintenance. Short-chain fatty acids (SCFAs), products of indigestible dietary fiber fermentation by gut microbiota, have strong anti-inflammatory properties and are seen as key protective factors against IBD. Among long-chain fatty acids, saturated fatty acids, trans fatty acids, and ω-6 polyunsaturated fatty acids exhibit pro-inflammatory effects, while oleic acid and ω-3 polyunsaturated fatty acids display anti-inflammatory actions. Lipid mediators derived from polyunsaturated fatty acids serve as bioactive molecules, influencing immune cell functions and offering both pro-inflammatory and anti-inflammatory benefits. Recent research has also highlighted the potential of medium- and very long-chain fatty acids in modulating inflammation, mucosal barriers, and gut microbiota in IBD. Given these insights, dietary intervention and supplementation with short-chain fatty acids are emerging as potential therapeutic strategies for IBD. This review elucidates the impact of various fatty acids and lipid mediators on IBD and delves into potential therapeutic avenues stemming from these compounds.
Collapse
Affiliation(s)
- Dong Yan
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuyu Ye
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Yue He
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Sidan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Yi Xiao
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xin Xiang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Weiwei Luo
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xuejie Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
30
|
Pereira GV, Boudaud M, Wolter M, Alexander C, De Sciscio A, Grant ET, Trindade BC, Pudlo NA, Singh S, Campbell A, Shan M, Zhang L, Willieme S, Kim K, Denike-Duval T, Bleich A, Schmidt TM, Kennedy L, Lyssiotis CA, Chen GY, Eaton KA, Desai MS, Martens EC. Unravelling specific diet and gut microbial contributions to inflammatory bowel disease. RESEARCH SQUARE 2023:rs.3.rs-2518251. [PMID: 36993463 PMCID: PMC10055531 DOI: 10.21203/rs.3.rs-2518251/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic condition characterized by periods of spontaneous intestinal inflammation and is increasing in industrialized populations. Combined with host genetic predisposition, diet and gut bacteria are thought to be prominent features contributing to IBD, but little is known about the precise mechanisms involved. Here, we show that low dietary fiber promotes bacterial erosion of protective colonic mucus, leading to lethal colitis in mice lacking the IBD-associated cytokine, interleukin-10. Diet-induced inflammation is driven by mucin-degrading bacteria-mediated Th1 immune responses and is preceded by expansion of natural killer T cells and reduced immunoglobulin A coating of some bacteria. Surprisingly, an exclusive enteral nutrition diet, also lacking dietary fiber, reduced disease by increasing bacterial production of isobutyrate, which is dependent on the presence of a specific bacterial species, Eubacterium rectale. Our results illuminate a mechanistic framework using gnotobiotic mice to unravel the complex web of diet, host and microbial factors that influence IBD.
Collapse
Affiliation(s)
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | - Celeste Alexander
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Alessandro De Sciscio
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Erica. T. Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, 4365 Esch-sur-Alzette, Luxembourg
| | | | - Nicholas A. Pudlo
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Shaleni Singh
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Austin Campbell
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mengrou Shan
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Dept. of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Li Zhang
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Dept. of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Stéphanie Willieme
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
| | - Kwi Kim
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Trisha Denike-Duval
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - André Bleich
- Institute for Laboratory Animal Science, Hanover Medical School, Hanover, Germany
| | - Thomas M. Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Dept. of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Lucy Kennedy
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Costas A. Lyssiotis
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Dept. of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Grace Y. Chen
- Dept. of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Kathryn A. Eaton
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mahesh S. Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, 5000 Odense, Denmark
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Bischoff SC, Bager P, Escher J, Forbes A, Hébuterne X, Hvas CL, Joly F, Klek S, Krznaric Z, Ockenga J, Schneider S, Shamir R, Stardelova K, Bender DV, Wierdsma N, Weimann A. ESPEN guideline on Clinical Nutrition in inflammatory bowel disease. Clin Nutr 2023; 42:352-379. [PMID: 36739756 DOI: 10.1016/j.clnu.2022.12.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 01/15/2023]
Abstract
The present guideline is an update and extension of the ESPEN scientific guideline on Clinical Nutrition in Inflammatory Bowel Disease published first in 2017. The guideline has been rearranged according to the ESPEN practical guideline on Clinical Nutrition in Inflammatory Bowel Disease published in 2020. All recommendations have been checked and, if needed, revised based on new literature, before they underwent the ESPEN consensus procedure. Moreover, a new chapter on microbiota modulation as a new option in IBD treatment has been added. The number of recommendations has been increased to 71 recommendations in the guideline update. The guideline is aimed at professionals working in clinical practice, either in hospitals or in outpatient medicine, and treating patients with IBD. General aspects of care in patients with IBD, and specific aspects during active disease and in remission are addressed. All recommendations are equipped with evidence grades, consensus rates, short commentaries and links to cited literature.
Collapse
Affiliation(s)
- Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Palle Bager
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark.
| | - Johanna Escher
- Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, the Netherlands.
| | - Alastair Forbes
- Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.
| | - Xavier Hébuterne
- Department of Gastroenterology and Clinical Nutrition, CHU of Nice, University Côte d'Azur, Nice, France.
| | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark.
| | - Francisca Joly
- Department of Gastroenterology and Nutrition Support, CHU de Beaujon, APHP, University of Paris, Paris, France.
| | - Stansilaw Klek
- Surgical Oncology Clinic, Maria Sklodowska-Curie National Cancer Institute, Krakow, Poland.
| | - Zeljko Krznaric
- Department of Gastroenterology, Hepatology and Nutrition, University Hospital Centre Zagreb, University of Zagreb, Croatia.
| | - Johann Ockenga
- Medizinische Klinik II, Klinikum Bremen-Mitte, Bremen FRG, Bremen, Germany.
| | - Stéphane Schneider
- Department of Gastroenterology and Clinical Nutrition, CHU de Nice, University Côte d'Azur, Nice, France.
| | - Raanan Shamir
- Institute for Gastroenterology, Nutrition and Liver Diseases, Schneider Children's Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Kalina Stardelova
- University Clinic for Gastroenterohepatology, Clinical Campus "Mother Theresa", University St Cyrul and Methodius, Skopje, North Macedonia.
| | - Darija Vranesic Bender
- Unit of Clinical Nutrition, Department of Internal Medicine, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Nicolette Wierdsma
- Department of Nutrition and Dietetics, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| | - Arved Weimann
- Department of General, Visceral and Oncological Surgery, St. George Hospital, Leipzig, Germany.
| |
Collapse
|
32
|
Reznikov EA, Suskind DL. Current Nutritional Therapies in Inflammatory Bowel Disease: Improving Clinical Remission Rates and Sustainability of Long-Term Dietary Therapies. Nutrients 2023; 15:nu15030668. [PMID: 36771373 PMCID: PMC9920576 DOI: 10.3390/nu15030668] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Inflammatory Bowel Disease (IBD) includes a spectrum of chronic immune-mediated intestinal diseases thought to be related to the complex interaction between the host immune system and the intestinal microbiome. Research supports the use of nutritional therapy in IBD; however, it is not routinely used in clinical practice. This literature review seeks to advance the understanding of diet and its effect in IBD with a focus on both Crohn's Disease (CD) and Ulcerative Colitis (UC). The contribution of diet to the development and treatment of IBD cannot be overstated. In both pediatric as well as adult IBD, nutritional interventions have been shown to improve clinical symptoms as well as inflammatory burden. The impact of dietary intervention is best exemplified through the use of Exclusive Enteral Nutrition (EEN) in CD. EEN and clinical research on exclusionary whole food diets-Crohn's Disease Exclusion Diet (CDED), Specific Carbohydrate Diet (SCD), low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet, and Mediterranean Diet-are discussed within this review. Current clinical literature supports the elimination of detrimental components and the incorporation of low processed whole foods in the diet. Additional prospective and longitudinal dietary studies on sustainable and long-term dietary options, along with a deeper understanding of the mechanism, are needed to further advance the role of nutritional interventions in IBD.
Collapse
|
33
|
Lee WS, Arai K, Alex G, Treepongkaruna S, Kim KM, Choong CL, Mercado KS, Darma A, Srivastava A, Aw MM, Huang J, Ni YH, Malik R, Tanpowpong P, Tran HN, Ukarapol N. Medical Management of Pediatric Inflammatory Bowel Disease (PIBD) in the Asia Pacific Region: A Position Paper by the Asian Pan-Pacific Society for Pediatric Gastroenterology, Hepatology, and Nutrition (APPSPGHAN) PIBD Working Group. J Gastroenterol Hepatol 2022; 38:523-538. [PMID: 36574956 DOI: 10.1111/jgh.16097] [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: 07/23/2022] [Revised: 11/08/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022]
Abstract
Pediatric inflammatory bowel disease (PIBD) is rising rapidly in many industrialised and affluent areas in the Asia Pacific region. Current available guidelines, mainly from Europe and North America, may not be completely applicable to clinicians caring for children with PIBD in this region due to differences in disease characteristics and regional resources constraints. This position paper is an initiative from the Asian Pan-Pacific Society for Pediatric Gastroenterology, Hepatology and Nutrition (APPSPGHAN) with the aim of providing an up-to-date, evidence-based approach to PIBD in the Asia Pacific region, taking into consideration the unique disease characteristics and financial resources available in this region. A group of pediatric gastroenterologists with special interest in PIBD performed an extensive literature search covering epidemiology, disease characteristics and natural history, management and monitoring. Gastrointestinal infections, including tuberculosis, need to be excluded before diagnosing IBD. In some populations in Asia, the Nudix Hydrolase 15 (NUD15) gene is a better predictor of leukopenia induced by azathioprine than thiopurine-S-methyltransferase (TPMT). The main considerations in the use of biologics in the Asia Pacific region are high cost, ease of access, and potential infectious risk, especially tuberculosis. Conclusion: This position paper provides a useful guide to clinicians in the medical management of children with PIBD in the Asia Pacific region.
Collapse
Affiliation(s)
- Way Seah Lee
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Katsuhiro Arai
- Center for Pediatric Inflammatory Bowel Disease, National Center for Child Health and Development, Tokyo, Japan
| | - George Alex
- Department of Gastroenterology and Nutrition, Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Suporn Treepongkaruna
- Division of Gastroenterology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Chee Liang Choong
- Department of Paediatrics, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Karen Sc Mercado
- Makati Medical Center and The Medical City, Philippine Society for Pediatric Gastroenterology, Hepatology and Nutrition, Manila, Philippines
| | - Andy Darma
- Department of Child Health, Dr. Soetomo General Hospital, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Anshu Srivastava
- Department of Paediatric Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Marion M Aw
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - James Huang
- Division of Paediatric Gastroenterology, Nutrition, Hepatology and Liver Transplantation, Department of Paediatrics, National University Hospital, Singapore
| | - Yen Hsuan Ni
- National Taiwan University College of Medicine, Taiwan
| | - Rohan Malik
- Department of Pediatrics, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Pornthep Tanpowpong
- Department of Pediatrics, Faculty of Medicine Ramathibodi, Mahidol University, Bangkok, Thailand
| | - Hong Ngoc Tran
- Department of Gastroenterology, Children's Hospital # 1, Ho Chi Minh City, Vietnam
| | - Nuthapong Ukarapol
- Department of Pediatric Gastroenterology and Hepatology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
34
|
Dawson R, Wands DIF, Logan M, Bremner G, Efklides S, Benn L, Henderson P, Grant H, Meredith J, Armstrong K, Wilson DC, Gerasimidis K, Alex G, Russell RK. Comparing Effectiveness of a Generic Oral Nutritional Supplement With Specialized Formula in the Treatment of Active Pediatric Crohn's Disease. Inflamm Bowel Dis 2022; 28:1859-1864. [PMID: 35259266 DOI: 10.1093/ibd/izac039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Exclusive enteral nutrition (EEN) is the recommended induction treatment of mild to moderate active pediatric Crohn's disease (CD). This study compared outcomes of 2 proprietary polymeric formulas. Treatment effectiveness was examined along with practical aspects of formula delivery and differences in estimated treatment costs. METHODS Data were retrospectively collected from patients with CD who received a generic oral nutritional supplement (Fortisip) across 2 centers (RCH, Melbourne and RHSC, Edinburgh). This was compared with a prospective cohort (RHC, Glasgow) that used a specialized formula (Modulen IBD). The data collected included patient demographics, remission rates, biochemical markers, administration method, and anthropometrics. The estimated treatment cost was performed by comparing price per kcal between each formula. RESULTS One hundred seventy-one patients were included (106 Fortisip, 65 Modulen IBD, 70 female; median age 13.3 yrs). No difference was demonstrated in remission rate (Fortisip n = 67 of 106 [63%] vs Modulen IBD n = 41 of 64 [64%], P = .89), nonadherence rate (Fortisip n = 7 of 106 [7%] vs Modulen IBD 3 of 64 [5%], P = .57) or method of administration (NGT Fortisip use n = 16 of 106 [12%] vs Modulen IBD 14 of 65 [22%]; P = .31). There was no difference in reduction of biochemical disease markers between the groups (C-reactive protein , P = .13; erythrocyte sedimentation rate, P = .49; fecal calprotectin, P = .94). However, there was a cost-saving of around £500/patient/course if the generic oral nutritional supplement was used. CONCLUSIONS The generic oral nutritional supplement and specialized formulas both had similar clinical effectiveness in induction of remission in pediatric CD. However, there is considerable cost-saving when using a generic oral nutritional supplement.
Collapse
Affiliation(s)
- R Dawson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
| | - D I F Wands
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
- Department of Gastroenterology, Hepatology and Clinical Nutrition, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - M Logan
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - G Bremner
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
| | - S Efklides
- Department of Nutrition and Food Services, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - L Benn
- Department of Nutrition and Food Services, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - P Henderson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young people, Edinburgh, UK
| | - H Grant
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
| | - J Meredith
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
| | - K Armstrong
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
| | - D C Wilson
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young people, Edinburgh, UK
| | - K Gerasimidis
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - G Alex
- Department of Gastroenterology, Hepatology and Clinical Nutrition, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - R K Russell
- Department of Paediatric Gastroenterology and Nutrition, Royal Hospital for Children & Young people, Edinburgh, UK
- School of Medicine, Dentistry and Nursing, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
- Child Life and Health, University of Edinburgh, Royal Hospital for Children & Young people, Edinburgh, UK
| |
Collapse
|
35
|
Abstract
The diet and gut microbiota have been extensively interrogated as a fuel for gut inflammation in inflammatory bowel diseases (IBDs) in the last few years. Here, we review how specific nutrients, typically enriched in a Western diet, instigate or deteriorate experimental gut inflammation in a genetically susceptible host and we discuss microbiota-dependent and independent mechanisms. We depict the study landscape of nutritional trials in paediatric and adult IBD and delineate common grounds for dietary advice. Conclusively, the diet reflects a critical rheostat of microbial dysbiosis and gut inflammation in IBD. Dietary restriction by exclusive enteral nutrition, with or without a specific exclusion diet, is effectively treating paediatric Crohn's disease, while adult IBD trials are less conclusive. Insights into molecular mechanisms of nutritional therapy will change the perception of IBD and will allow us to enter the era of precision nutrition. To achieve this, we discuss the need for carefully designed nutritional trials with scientific rigour comparable to medical trials, which also requires action from stake holders. Establishing evidence-based dietary therapy for IBD does not only hold promise to avoid long-term immunosuppression, but to provide a widely accessible therapy at low cost. Identification of dietary culprits disturbing gut health also bears the potential to prevent IBD and allows informed decision making in food politics.
Collapse
Affiliation(s)
- Timon E Adolph
- Department of Medicine I, Gastroenterology, Hepatology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Jingwan Zhang
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
36
|
Fitzpatrick JA, Melton SL, Yao CK, Gibson PR, Halmos EP. Dietary management of adults with IBD - the emerging role of dietary therapy. Nat Rev Gastroenterol Hepatol 2022; 19:652-669. [PMID: 35577903 DOI: 10.1038/s41575-022-00619-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2022] [Indexed: 02/08/2023]
Abstract
Historically, dietitians played a minor part in the management of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis. Patients were commonly referred for consequences of uncontrolled disease, such as malnutrition and bowel obstruction risk. Today, dietitians are fundamental members of the multidisciplinary IBD team, from educating on the role of diet at diagnosis and throughout the lifespan of a patient with IBD to guiding primary induction therapy. This aspect is reflected in published guidelines for IBD management, which previously placed diet as only a minor factor, but now have diet-specific publications. This Review describes a four-step approach in a dietitian's assessment and management of diet in patients with IBD: (1) identifying and correcting nutritional gaps and dietary imbalances; (2) considering diet to treat active disease with the use of exclusive enteral nutrition (EEN) or emerging diets that could replace EEN; (3) using therapeutic diets to control existing complications of IBD, such as reduced fibre to prevent bowel obstruction in stricturing disease or a fermentable oligosaccharides, disaccharides, monosaccharides and polyols diet to manage co-existing functional gut symptoms; and (4) considering the role of diet in preventing IBD development in high-risk populations.
Collapse
Affiliation(s)
- Jessica A Fitzpatrick
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Sarah L Melton
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Chu Kion Yao
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Peter R Gibson
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Emma P Halmos
- Department of Gastroenterology, The Alfred Hospital and Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
37
|
Gulliver EL, Young RB, Chonwerawong M, D'Adamo GL, Thomason T, Widdop JT, Rutten EL, Rossetto Marcelino V, Bryant RV, Costello SP, O'Brien CL, Hold GL, Giles EM, Forster SC. Review article: the future of microbiome-based therapeutics. Aliment Pharmacol Ther 2022; 56:192-208. [PMID: 35611465 PMCID: PMC9322325 DOI: 10.1111/apt.17049] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/29/2022] [Accepted: 05/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND From consumption of fermented foods and probiotics to emerging applications of faecal microbiota transplantation, the health benefit of manipulating the human microbiota has been exploited for millennia. Despite this history, recent technological advances are unlocking the capacity for targeted microbial manipulation as a novel therapeutic. AIM This review summarises the current developments in microbiome-based medicines and provides insight into the next steps required for therapeutic development. METHODS Here we review current and emerging approaches and assess the capabilities and weaknesses of these technologies to provide safe and effective clinical interventions. Key literature was identified through Pubmed searches with the following key words, 'microbiome', 'microbiome biomarkers', 'probiotics', 'prebiotics', 'synbiotics', 'faecal microbiota transplant', 'live biotherapeutics', 'microbiome mimetics' and 'postbiotics'. RESULTS Improved understanding of the human microbiome and recent technological advances provide an opportunity to develop a new generation of therapies. These therapies will range from dietary interventions, prebiotic supplementations, single probiotic bacterial strains, human donor-derived faecal microbiota transplants, rationally selected combinations of bacterial strains as live biotherapeutics, and the beneficial products or effects produced by bacterial strains, termed microbiome mimetics. CONCLUSIONS Although methods to identify and refine these therapeutics are continually advancing, the rapid emergence of these new approaches necessitates accepted technological and ethical frameworks for measurement, testing, laboratory practices and clinical translation.
Collapse
Affiliation(s)
- Emily L. Gulliver
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Remy B. Young
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Gemma L. D'Adamo
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Tamblyn Thomason
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - James T. Widdop
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Emily L. Rutten
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Vanessa Rossetto Marcelino
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| | - Robert V. Bryant
- Department of GastroenterologyThe Queen Elizabeth HospitalWoodvilleSouth AustraliaAustralia,School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | - Samuel P. Costello
- Department of GastroenterologyThe Queen Elizabeth HospitalWoodvilleSouth AustraliaAustralia,School of MedicineUniversity of AdelaideAdelaideSouth AustraliaAustralia
| | | | - Georgina L. Hold
- Microbiome Research Centre, St George & Sutherland Clinical SchoolUniversity of New South WalesSydneyNew South WalesAustralia
| | - Edward M. Giles
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia,Department of PaediatricsMonash UniversityClaytonVictoriaAustralia
| | - Samuel C. Forster
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVictoriaAustralia,Department of Molecular and Translational SciencesMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
38
|
Bak MTJ, Ruiterkamp MFE, van Ruler O, Campmans-Kuijpers MJE, Bongers BC, van Meeteren NLU, van der Woude CJ, Stassen LPS, de Vries AC. Prehabilitation prior to intestinal resection in Crohn's disease patients: An opinion review. World J Gastroenterol 2022; 28:2403-2416. [PMID: 35979261 PMCID: PMC9258284 DOI: 10.3748/wjg.v28.i22.2403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/21/2022] [Accepted: 05/22/2022] [Indexed: 02/06/2023] Open
Abstract
Patients with Crohn's disease (CD) are at a considerable risk for intestinal surgery. Approximately 25% of patients with CD will undergo an intestinal resection within 10 years of diagnosis. Postoperative complications after CD surgery have been reported in 20%-47% of the patients. Both general and CD-related risk factors are associated with postoperative complications, and comprise non-modifiable (e.g., age) and potentially modifiable risk factors (e.g., malnutrition). Prehabilitation focuses on the preoperative period with strategies designed to optimize modifiable risk factors concerning the physical and mental condition of the individual patient. The aim of prehabilitation is to enhance postoperative recovery and return to or even improve preoperative functional capacity. Preoperative improvement of nutritional status, physical fitness, cessation of smoking, psychological support, and critical revision of preoperative use of CD medication are important strategies. Studies of the effect on postoperative outcome in CD patients are scarce, and guidelines lack recommendations on tailored management. In this opinion review, we review the current evidence on the impact of screening and management of nutritional status, physical fitness, CD medication and laboratory values on the postoperative course following an intestinal resection in CD patients. In addition, we aim to provide guidance for individualized multimodal prehabilitation in clinical practice concerning these modifiable factors.
Collapse
Affiliation(s)
- Michiel T J Bak
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| | - Marit F E Ruiterkamp
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| | - Oddeke van Ruler
- Department of Surgery, IJsselland Hospital, Capelle aan den IJssel 2906 ZC, Netherlands
- Department of Surgery, Erasmus University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| | - Marjo J E Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University Medical Center Groningen and University of Groningen, Groningen 9713 GZ, Netherlands
| | - Bart C Bongers
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht 6200 MD, Netherlands
- Department of Epidemiology, Care and Public Health Research Institute, Maastricht University, Maastricht 6200 MD, Netherlands
| | - Nico L U van Meeteren
- Department of Anaesthesiology, Erasmus MC University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| | - Laurents P S Stassen
- Department of Surgery, Maastricht University Medical Center, Maastricht 6229 HX, Netherlands
| | - Annemarie C de Vries
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, Rotterdam 3015 GD, Netherlands
| |
Collapse
|
39
|
The Impact of Compliance During Exclusive Enteral Nutrition on Faecal Calprotectin in Children With Crohn Disease. J Pediatr Gastroenterol Nutr 2022; 74:801-804. [PMID: 35192573 DOI: 10.1097/mpg.0000000000003425] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
It remains unclear whether suboptimal response to exclusive enteral nutrition (EEN) in some children with Crohn disease (CD) is explained by poor compliance. The present study measured faecal gluten immunogenic peptides (GIP), a biomarker of gluten intake, in 45 children (3- 17 years) with CD, and explored associations with faecal calprotectin (FC) levels at 33 and 54 days of EEN. FC decreased in patients with undetectable GIP at both 33 and 54 days of EEN (mean decrease, 33 days: -743 mg/kg, 54 days: -1043 mg/kg, P < 0.001) but not in patients who had detectable levels. At EEN completion, patients with undetectable GIP had a lower FC by 717 mg/kg compared with patients with a positive GIP result (P = 0.042) and demonstrated a greater decline from baseline FC (-69% vs +5%, P = 0.011). Poorer response to EEN is explained in part by diminished compliance. Faecal GIP might be useful as proxy biomarker of EEN compliance.
Collapse
|
40
|
Gill PA, Inniss S, Kumagai T, Rahman FZ, Smith AM. The Role of Diet and Gut Microbiota in Regulating Gastrointestinal and Inflammatory Disease. Front Immunol 2022; 13:866059. [PMID: 35450067 PMCID: PMC9016115 DOI: 10.3389/fimmu.2022.866059] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022] Open
Abstract
Diet is an important lifestyle factor that is known to contribute in the development of human disease. It is well established that poor diet plays an active role in exacerbating metabolic diseases, such as obesity, diabetes and hypertension. Our understanding of how the immune system drives chronic inflammation and disease pathogenesis has evolved in recent years. However, the contribution of dietary factors to inflammatory conditions such as inflammatory bowel disease, multiple sclerosis and arthritis remain poorly defined. A western diet has been associated as pro-inflammatory, in contrast to traditional dietary patterns that are associated as being anti-inflammatory. This may be due to direct effects of nutrients on immune cell function. Diet may also affect the composition and function of gut microbiota, which consequently affects immunity. In animal models of inflammatory disease, diet may modulate inflammation in the gastrointestinal tract and in other peripheral sites. Despite limitations of animal models, there is now emerging evidence to show that anti-inflammatory effects of diet may translate to human gastrointestinal and inflammatory diseases. However, appropriately designed, larger clinical studies must be conducted to confirm the therapeutic benefit of dietary therapy.
Collapse
Affiliation(s)
- Paul A Gill
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Saskia Inniss
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Tomoko Kumagai
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| | - Farooq Z Rahman
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom.,Department of Gastroenterology, University College London Hospitals National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Andrew M Smith
- Department of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, United Kingdom
| |
Collapse
|
41
|
Hart L, Verburgt CM, Wine E, Zachos M, Poppen A, Chavannes M, Van Limbergen J, Pai N. Nutritional Therapies and Their Influence on the Intestinal Microbiome in Pediatric Inflammatory Bowel Disease. Nutrients 2021; 14:nu14010004. [PMID: 35010879 PMCID: PMC8746384 DOI: 10.3390/nu14010004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic, autoimmune disorder of the gastrointestinal tract with numerous genetic and environmental risk factors. Patients with Crohn’s disease (CD) or ulcerative colitis (UC) often demonstrate marked disruptions of their gut microbiome. The intestinal microbiota is strongly influenced by diet. The association between the increasing incidence of IBD worldwide and increased consumption of a westernized diet suggests host nutrition may influence the progression or treatment of IBD via the microbiome. Several nutritional therapies have been studied for the treatment of CD and UC. While their mechanisms of action are only partially understood, existing studies do suggest that diet-driven changes in microbial composition and function underlie the diverse mechanisms of nutritional therapy. Despite existing therapies for IBD focusing heavily on immune suppression, nutrition is an important treatment option due to its superior safety profile, potentially low cost, and benefits for growth and development. These benefits are increasingly important to patients. In this review, we will describe the clinical efficacy of the different nutritional therapies that have been described for the treatment of CD and UC. We will also describe the effects of each nutritional therapy on the gut microbiome and summarize the strength of the literature with recommendations for the practicing clinician.
Collapse
Affiliation(s)
- Lara Hart
- Department of Paediatrics, Division of Paediatric Gastroenterology & Nutrition, McMaster University, Hamilton, ON L8N 3Z5, Canada; (L.H.); (M.Z.)
- McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Charlotte M. Verburgt
- Department of Pediatric Gastroenterology and Nutrition, Amsterdam University Medical Centers, Emma Children’s Hospital, 1105 AZ Amsterdam, The Netherlands; (C.M.V.); (J.V.L.)
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Centers, Emma Children’s Hospital, 1105 AZ Amsterdam, The Netherlands
| | - Eytan Wine
- Edmonton Paediatric IBD Clinic, Division of Paediatric Gastroenterology and Nutrition, Departments of Paediatrics & Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Mary Zachos
- Department of Paediatrics, Division of Paediatric Gastroenterology & Nutrition, McMaster University, Hamilton, ON L8N 3Z5, Canada; (L.H.); (M.Z.)
- McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
| | - Alisha Poppen
- College of Medicine and Health, University College Cork, T12 K8AF Cork, Ireland;
| | - Mallory Chavannes
- Department of Paediatrics, Division of Paediatric Gastroenterology and Nutrition, Children’s Hospital of Los Angeles, Los Angeles, CA 90027, USA;
| | - Johan Van Limbergen
- Department of Pediatric Gastroenterology and Nutrition, Amsterdam University Medical Centers, Emma Children’s Hospital, 1105 AZ Amsterdam, The Netherlands; (C.M.V.); (J.V.L.)
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Paediatrics, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Nikhil Pai
- Department of Paediatrics, Division of Paediatric Gastroenterology & Nutrition, McMaster University, Hamilton, ON L8N 3Z5, Canada; (L.H.); (M.Z.)
- McMaster Children’s Hospital, Hamilton, ON L8N 3Z5, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON L8N 3Z5, Canada
- Correspondence: ; Tel.: +905-521-2100 (ext. 73587); Fax: +905-521-2655
| |
Collapse
|
42
|
Shariff S, Moran G, Grimes C, Cooney RM. Current Use of EEN in Pre-Operative Optimisation in Crohn's Disease. Nutrients 2021; 13:4389. [PMID: 34959941 PMCID: PMC8709272 DOI: 10.3390/nu13124389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the increasing array of medications available for the treatment of Crohn's disease and a focus on mucosal healing, approximately 35% of patients with Crohn's disease undergo bowel surgery at some stage. The importance of nutritional optimisation before Crohn's surgery is well-highlighted by surgical, nutritional, and gastroenterological societies with the aim of reducing complications and enhancing recovery. Surgical procedures are frequently undertaken when other treatment options have been unsuccessful, and, thus, patients may have lost weight and/or required steroids, and are therefore at higher risk of post-operative complications. EEN is used extensively in the paediatric population to induce remission, but is not routinely used in the induction of remission of adult Crohn's disease or in pre-operative optimisation. Large prospective studies regarding the role of pre-operative EEN are lacking. In this review, we evaluate the current literature on the use of EEN in pre-operative settings and its impact on patient outcomes.
Collapse
Affiliation(s)
- Sharafaath Shariff
- Queen Elizabeth Hospital, University Hospital Birmingham NHS Trust, Birmingham B15 2GW, UK; (S.S.); (R.M.C.)
| | - Gordon Moran
- Room D1406 West Block: Queen’s Medical Centre, University of Nottingham, Queen’s Medical Centre, Nottingham NG7 2UH, UK
| | - Caris Grimes
- Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK;
| | - Rachel Margaret Cooney
- Queen Elizabeth Hospital, University Hospital Birmingham NHS Trust, Birmingham B15 2GW, UK; (S.S.); (R.M.C.)
| |
Collapse
|
43
|
Gkikas K, Logan M, Nichols B, Ijaz UZ, Clark CM, Svolos V, Gervais L, Duncan H, Garrick V, Curtis L, Buchanan E, Cardigan T, Armstrong L, Delahunty C, Flynn DM, Barclay AR, Tayler R, Milling S, Hansen R, Russell RK, Gerasimidis K. Dietary triggers of gut inflammation following exclusive enteral nutrition in children with Crohn's disease: a pilot study. BMC Gastroenterol 2021; 21:454. [PMID: 34861829 PMCID: PMC8642954 DOI: 10.1186/s12876-021-02029-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Background The anti-inflammatory effect of exclusive enteral nutrition on the gut of children with Crohn’s disease is rapidly lost after food reintroduction. This study assessed disease dietary triggers following successful treatment with exclusive enteral nutrition. Methods Nutrient intake, dietary patterns and dietary biomarkers in faeces (gluten immunogenic peptides, undigestible starch, short chain fatty acids) were assessed in 14 children with Crohn’s disease during early food reintroduction, following exclusive enteral nutrition. Groups above (Group A) and below (Group B) the median levels of faecal calprotectin after food reintroduction were assigned for comparative analysis. Results Intakes of fibre, gluten-containing cereals and red and processed meat were significantly higher in Group A than Group B; (median [Q1, Q3], g/day; Fibre: 12.1 [11.2, 19.9] vs. 9.9 [7.6, 12.1], p = 0.03; Red and processed meat: 151 [66.7, 190] vs. 63.3 [21.7, 67], p = 0.02; gluten-containing cereals: 289 [207, 402] vs. 203 [61, 232], p = 0.035). A diet consisting of cereals and meat products was predictive (92% accuracy) of higher faecal calprotectin levels after food reintroduction. In faeces, butyrate levels, expressed as absolute concentration and relative abundance, were higher in Group A than Group B by 28.4 µmol/g (p = 0.015) and 6.4% (p = 0.008), respectively. Levels of gluten immunogenic peptide and starch in faeces did not differ between the two groups. Conclusions This pilot study identified potential dietary triggers of gut inflammation in children with Crohn’s disease after food reintroduction following treatment with exclusive enteral nutrition. Trial registration: Clinical trials.gov registration number: NCT02341248; Clinical trials.gov URL: https://clinicaltrials.gov/ct2/show/NCT02341248 (retrospectively registered). Supplementary Information The online version contains supplementary material available at 10.1186/s12876-021-02029-4.
Collapse
Affiliation(s)
- Konstantinos Gkikas
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Michael Logan
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ben Nichols
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Umer Z Ijaz
- Civil Engineering, School of Engineering, University of Glasgow, Glasgow, UK
| | - Clare M Clark
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Vaios Svolos
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Lisa Gervais
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Hazel Duncan
- Department of Paediatrics, Crosshouse Hospital, Kilmarnock, UK
| | - Vikki Garrick
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Lee Curtis
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Elaine Buchanan
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Tracey Cardigan
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | | | | | - Diana M Flynn
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Andrew R Barclay
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Rachel Tayler
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | - Simon Milling
- Institute for Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Richard Hansen
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Royal Hospital for Children, Glasgow, UK
| | | | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
44
|
Wolter M, Grant ET, Boudaud M, Steimle A, Pereira GV, Martens EC, Desai MS. Leveraging diet to engineer the gut microbiome. Nat Rev Gastroenterol Hepatol 2021; 18:885-902. [PMID: 34580480 DOI: 10.1038/s41575-021-00512-7] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/12/2022]
Abstract
Autoimmune diseases, including inflammatory bowel disease, multiple sclerosis and rheumatoid arthritis, have distinct clinical presentations but share underlying patterns of gut microbiome perturbation and intestinal barrier dysfunction. Their potentially common microbial drivers advocate for treatment strategies aimed at restoring appropriate microbiome function, but individual variation in host factors makes a uniform approach unlikely. In this Perspective, we consolidate knowledge on diet-microbiome interactions in local inflammation, gut microbiota imbalance and host immune dysregulation. By understanding and incorporating the effects of individual dietary components on microbial metabolic output and host physiology, we examine the potential for diet-based therapies for autoimmune disease prevention and treatment. We also discuss tools targeting the gut microbiome, such as faecal microbiota transplantation, probiotics and orthogonal niche engineering, which could be optimized using custom dietary interventions. These approaches highlight paths towards leveraging diet for precise engineering of the gut microbiome at a time of increasing autoimmune disease.
Collapse
Affiliation(s)
- Mathis Wolter
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Erica T Grant
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Marie Boudaud
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Alex Steimle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | | | - Eric C Martens
- University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mahesh S Desai
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg. .,Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
45
|
Yanai H, Levine A, Hirsch A, Boneh RS, Kopylov U, Eran HB, Cohen NA, Ron Y, Goren I, Leibovitzh H, Wardi J, Zittan E, Ziv-Baran T, Abramas L, Fliss-Isakov N, Raykhel B, Gik TP, Dotan I, Maharshak N. The Crohn's disease exclusion diet for induction and maintenance of remission in adults with mild-to-moderate Crohn's disease (CDED-AD): an open-label, pilot, randomised trial. Lancet Gastroenterol Hepatol 2021; 7:49-59. [PMID: 34739863 DOI: 10.1016/s2468-1253(21)00299-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The Crohn's disease exclusion diet (CDED) with partial enteral nutrition is effective for induction of remission in children with mild-to-moderate Crohn's disease. We aimed to assess the CDED in adults with Crohn's disease. METHODS We did an open-label, pilot randomised trial at three medical centres in Israel. Eligible patients were biologic naive adults aged 18-55 years with mild-to-moderate Crohn's disease (defined by a Harvey-Bradshaw Index score of 5-14 points), a maximal disease duration of 5 years, with active disease on colonoscopy, or imaging with elevated inflammatory markers (C-reactive protein >5 mg/L or faecal calprotectin concentration >200 μ/g). Patients were randomly assigned (1:1) to CDED plus partial enteral nutrition or CDED alone for 24 weeks. Randomisation was via block randomisation (block sizes of six) using sealed, numbered, and opaque envelopes. Patients and investigators were aware of which group patients were assigned to due to the nature of the different interventions. The primary endpoint was clinical remission, defined as a Harvey-Bradshaw Index score of less than 5 at week 6. The primary endpoint was assessed in the intention-to-treat (ITT) population, which included all patients who used the dietary therapy for at least 48 h. We report results of the final analysis. This trial is registered with ClinicalTrials.gov, NCT02231814. FINDINGS Between Jan 12, 2017, and May 11, 2020, 91 patients were screened, of whom 44 were randomly assigned to the CDED plus partial enteral nutrition group (n=20) or CDED alone group (n=24). 19 patients in the CDED plus partial enteral nutrition group and 21 patients in the CDED alone group received the allocated intervention for at least 48 h and thus were included in the ITT analysis. At week 6, 13 (68%) of 19 patients in the CDED plus partial enteral nutrition group and 12 (57%) of 21 patients in the CDED group had achieved clinical remission (p=0·4618). Among the 25 patients in remission at week 6, 20 (80%) were in sustained remission at week 24 (12 patients in the CDED plus partial enteral nutrition group and eight in the CDED alone group). 14 (35%) of 40 patients were in endoscopic remission at week 24 (eight patients in the CDED plus partial enteral nutrition group and six in the CDED alone group). No serious adverse events or treatment-related adverse events were reported in either group. INTERPRETATION CDED with or without partial enteral nutrition was effective for induction and maintenance of remission in adults with mild-to-moderate biologic naive Crohn's disease and might lead to endoscopic remission. These data suggest that CDED could be used for mild-to-moderate active Crohn's disease and should be assessed in a powered randomised controlled trial. FUNDING Azrieli Foundation and Nestle Health Science.
Collapse
Affiliation(s)
- Henit Yanai
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Arie Levine
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Paediatric Gastroenterology Unit, Wolfson Medical Center, Holon, Israel
| | - Ayal Hirsch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Rotem Sigall Boneh
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Israel PIBD Research Center, Wolfson Medical Center, Holon, Israel
| | - Uri Kopylov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology, Sheba Medical Center, Ramat-Gan, Israel
| | - Hagar Banai Eran
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nathaniel A Cohen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yulia Ron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Idan Goren
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Haim Leibovitzh
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Joram Wardi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Gastroenterology Institute, Wolfson Medical Center, Holon, Israel
| | - Eran Zittan
- The Abraham and Sonia Rochlin IBD Unit, Department of Gastroenterology, Emek Medical Center, Afula, Israel; Rappaport Faculty of Medicine Technion-Israel Institute of Technology, Haifa, Israel
| | - Tomer Ziv-Baran
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lee Abramas
- Israel PIBD Research Center, Wolfson Medical Center, Holon, Israel
| | - Naomi Fliss-Isakov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Barbara Raykhel
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel
| | - Tamar Pfeffer Gik
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nitsan Maharshak
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
46
|
Svolos V, Gkikas K, Gerasimidis K. Diet and gut microbiota manipulation for the management of Crohn's disease and ulcerative colitis. Proc Nutr Soc 2021; 80:1-15. [PMID: 34551834 DOI: 10.1017/s0029665121002846] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aetiology of inflammatory bowel disease (IBD) is multifactorial, with diet and gut microbiota playing an important role. Nonetheless, there are very few studies, particularly clinical research, which have explored the interaction between diet and gut microbiota. In the current review, we summarise the evidence from clinical trials exploring the interactions between the gut microbiota and diet in the management of IBD. Data from the effect of exclusive enteral nutrition (EEN) on the gut microbiota of children with active Crohn's disease (CD), receiving induction treatment, offer opportunities to understand the role of gut microbiota in underlying disease pathogenesis and develop novel dietary and pharmacological microbial therapeutics. In contrast, the evidence which links the effectiveness of food-based dietary therapies for IBD with mechanisms involving the gut microbiota is far less convincing. The microbial signals arising from these dietary therapies are inconsistent and vary compared to the effects of effective treatment with EEN in CD.
Collapse
Affiliation(s)
- Vaios Svolos
- Human Nutrition, School of Medicine, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, G31 2ER, Glasgow, UK
| | - Konstantinos Gkikas
- Human Nutrition, School of Medicine, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, G31 2ER, Glasgow, UK
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, University of Glasgow, New Lister Building, Glasgow Royal Infirmary, G31 2ER, Glasgow, UK
| |
Collapse
|
47
|
Peters V, Dijkstra G, Campmans-Kuijpers MJE. Are all dietary fibers equal for patients with inflammatory bowel disease? A systematic review of randomized controlled trials. Nutr Rev 2021; 80:1179-1193. [PMID: 34486663 PMCID: PMC8990763 DOI: 10.1093/nutrit/nuab062] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
CONTEXT Conflicting practice-based dietary recommendations are sometimes given to patients with inflammatory bowel disease (IBD); whereas intake of fiber should increase during remission, it should be avoided during relapse. Moreover, European countries set daily requirements of total fiber and do not specify any types. OBJECTIVE This systematic review appraised data from randomized clinical trials (RCTs) of the types of fibers beneficial for patients in the treatment of IBD to guide dietary fiber advice. DATA SOURCES The PubMED database was searched following PRISMA guidelines. DATA EXTRACTION RCTs evaluating the effects of any type of fiber on clinical and physiological outcomes in patients with IBD were assessed. Quality assessment of the selected full-text articles was conducted using the Cochrane Risk of Bias Tool. DATA ANALYSIS Eight studies were included reporting on 5 types of fibers. In 2 RCTs, germinated barley foodstuff (GBF) was shown to lower pro-inflammatory cytokines and clinical disease activity scores. Fructo-oligosaccharides (FOS) were demonstrated to lower IBD Questionnaire scores (lower well-being), in contrast to inulin, which decreased disease activity scores. An RCT could not find lower remission rates in the psyllium treatment group, while another RCT reported that administration led to less symptoms in patients. In RCTs, no concrete evidence was found that wheat bran improves disease course. CONCLUSIONS Although the evidence is sparse, GBF and inulin seem propitious and merit further exploration. Evidence on wheat bran and psyllium is still too limited. Adequately powered long-term human RCTs with objective outcomes are needed to improve dietary advice on types of fiber in IBD.
Collapse
Affiliation(s)
- Vera Peters
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Marjo J E Campmans-Kuijpers
- Department of Gastroenterology and Hepatology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
48
|
Mitrev N, Huang H, Hannah B, Kariyawasam VC. Review of exclusive enteral therapy in adult Crohn's disease. BMJ Open Gastroenterol 2021; 8:e000745. [PMID: 34580154 PMCID: PMC8477235 DOI: 10.1136/bmjgast-2021-000745] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Exclusive enteral nutrition (EEN) is a potentially effective but underused therapy for Crohn's disease (CD) in adults. It is first-line induction treatment for paediatric patients but remains a second-line or third-line therapy in adults. OBJECTIVE To analyse the evidence for EEN in adult patients with CD, and summarise this in a narrative review. METHODS In April/May 2020 and July 2021, a literature search was performed using the Medical Subject Headings (MeSH) terms: 'Crohn's disease', 'CD', 'inflammatory bowel disease', 'IBD', 'exclusive enteral nutrition', 'enteral nutrition', 'EEN', in PubMed, Scopus, Cochrane. Additional studies were obtained from references of search result articles as well as general reading. Studies with adult patients with CD treated with EEN were selected. 79 articles of relevance were found. Where data in adults were lacking, data from paediatric studies as extrapolated with care. RESULTS EEN in adult patients been shown to improve clinical, biomarker, endoscopic and radiologic measures of disease activity. EEN avoids the potential adverse effects of recurrent corticosteroids for induction such as metabolic derangements and opportunistic infections. EEN has also demonstrated benefits among adult patients with fistulising and stricturing CD. It may avoid surgery in such patients. Preoperative EEN has also been shown to reduce postoperative complications and recurrence. There appears to be benefits in combing EEN with antitumour necrosis factor agents, however, benefits of combination therapy with other biologics are less clear. A major drawback of EEN therapy in adults has been poor compliance. More palatable polymeric formulations improved patient education and dietitian support may overcome this. Evidence in adults is limited to small studies, often with suboptimal control arms and lack of blinding. Larger scale studies with improved study design are needed to confirm these beneficial effects. CONCLUSION Despite limitations in evidence EEN should be considered in treating adults with CD.
Collapse
Affiliation(s)
- Nikola Mitrev
- Department of Gastroenterology, Blacktown Hospital, Blacktown, New South Wales, Australia
- Western Sydney University Blacktown Mount Druitt Medical School, Blacktown, New South Wales, Australia
| | - Hin Huang
- Western Sydney University Blacktown Mount Druitt Medical School, Blacktown, New South Wales, Australia
| | - Barbara Hannah
- Department of Gastroenterology, Blacktown Hospital, Blacktown, New South Wales, Australia
| | | |
Collapse
|
49
|
Bancil AS, Sandall AM, Rossi M, Chassaing B, Lindsay JO, Whelan K. Food Additive Emulsifiers and Their Impact on Gut Microbiome, Permeability, and Inflammation: Mechanistic Insights in Inflammatory Bowel Disease. J Crohns Colitis 2021; 15:1068-1079. [PMID: 33336247 DOI: 10.1093/ecco-jcc/jjaa254] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The global burden of inflammatory bowel disease [IBD] has increased over the 21st century. Despite multiple studies investigating the pathogenesis of IBD, the causative mechanisms pertaining to its increased prevalence remain unclear. There is growing evidence that aspects of a 'Western diet' increase the risk of developing IBD. More recently, evidence implicating dietary emulsifiers has accumulated, with ecological studies showing a positive correlation between inflammatory bowel disease and emulsifier consumption. Further to these, cell and animal studies have demonstrated plausible mechanisms by which dietary emulsifiers may contribute to IBD pathogenesis through mechanisms including: promotion of pro-inflammatory intestinal microbiota; disruption of mucus architecture; increased intestinal permeability; activation of inflammatory pathways; and disruption of the cell cycle. This review critically analyses the current evidence for these mechanisms that may be of pathological relevance to IBD, evaluates recent dietary trials, acknowledges the challenges of dietary intervention studies, and gives an overview of ongoing and future clinical trials in this important area.
Collapse
Affiliation(s)
- Aaron S Bancil
- King's College London, Department of Nutritional Sciences, London, UK
| | - Alicia M Sandall
- King's College London, Department of Nutritional Sciences, London, UK
| | - Megan Rossi
- King's College London, Department of Nutritional Sciences, London, UK
| | - Benoit Chassaing
- INSERM U1016, CNRS UMR 8104, Université de Paris, Paris, France.,Neuroscience Institute, Georgia State University, Atlanta, GA, USA.,Institute for Biomedical Sciences, Center for Inflammation, Immunity and Infection, Georgia State University, Atlanta, GA, USA
| | - James O Lindsay
- Queen Mary University of London, Blizard Institute, Barts and the London School of Medicine, London, UK
| | - Kevin Whelan
- King's College London, Department of Nutritional Sciences, London, UK
| |
Collapse
|
50
|
Gerasimidis K, Godny L, Sigall-Boneh R, Svolos V, Wall C, Halmos E. Current recommendations on the role of diet in the aetiology and management of IBD. Frontline Gastroenterol 2021; 13:160-167. [PMID: 35300465 PMCID: PMC8862489 DOI: 10.1136/flgastro-2020-101429] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023] Open
Abstract
Diet is a key modifier of risk of inflammatory bowel disease development and potentially a treatment option in patients with established disease. International organisations in gastroenterology and inflammatory bowel disease have published guidelines for the role of diet in disease onset and its management. Here, we discuss the major overarching themes arising from these guidelines and appraise recent literature on the role of diet for inflammatory bowel disease prevention, treatment of active disease and maintenance of remission, considering these themes. Except for exclusive enteral nutrition in active Crohn's disease, we currently possess very little evidence to make any further dietary recommendations for the management of inflammatory bowel disease. There is also currently uncertainty on the extrapolation of epidemiological dietary signals on risk of disease development and preclinical experiments in animal models to management, once disease is established. Until high-quality evidence from clinical research becomes available, the only specific recommendations for inflammatory bowel disease we might safely give are those of healthy eating which apply for the general population for overall health and well-being.
Collapse
Affiliation(s)
| | - Lihi Godny
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva and the Sackler Faculty of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Rotem Sigall-Boneh
- Paediatric Gastroenterology and Nutrition Unit, Wolfson Medical Center, Holon and the Sackler Faculty of Medicine, Tel Aviv University, Israel, Holon, Israel
| | - Vaios Svolos
- Human Nutrition, School of Medicine, University of Glasgow, Glasgow, UK
| | - Catherine Wall
- Department of Medicine and Department of Human Nutrition, University of Otago Dunedin School of Medicine, Christchurch, New Zealand
| | - Emma Halmos
- Department of Gastroenterology, Central Clinical School, Monash University, The Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|