1
|
Frost G, Lutz C, Schwiertz A, Huber H, Missbichler A. Total histamine degrading capacity: Correlation to stool histamine and its usefulness in supporting the diagnosis of histamine intolerance. Anal Biochem 2025; 702:115819. [PMID: 39993492 DOI: 10.1016/j.ab.2025.115819] [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: 08/19/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
The relatively new test (European Patent granted 2022) for determining the total histamine degradation capacity [Totale Histamin Abbau Kapazität/Total Histamine Degrading Capacity] THAK®/THDC of a serum sample has been compared in various settings with the individual clinical history and the levels of histamine in stool. The measurement of THAK can be carried out regardless of the current status of the patient (independent of any acute symptoms), therefore THAK test is a useful and very reliable instrument for supporting the diagnosing histamine intolerance. As soon as the THAK falls below a threshold value the patient is most likely to suffer from HIT, even if the intensity of the symptoms is low. Low THAK values correspond with a significant increase of histamine in stool. Results were evaluated according to both age and gender, no difference between the sexes were found. However, there is a noticeable dependency on age, although this is not diagnostically relevant. THAK® is a registered trademark of Frost Diagnostika.
Collapse
|
2
|
Choi Y, Youn YH, Kang SJ, Shin JE, Cho YS, Jung YS, Shin SY, Huh CW, Lee YJ, Koo HS, Nam K, Lee HS, Kim DH, Park YH, Kim MC, Song HY, Yoon SH, Lee SY, Choi M, Park MI, Sung IK. 2025 Seoul Consensus on Clinical Practice Guidelines for Irritable Bowel Syndrome. J Neurogastroenterol Motil 2025; 31:133-169. [PMID: 40205893 PMCID: PMC11986658 DOI: 10.5056/jnm25007] [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: 01/31/2025] [Accepted: 03/16/2025] [Indexed: 04/11/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic, disabling, and functional bowel disorder that significantly affects social functioning and reduces quality of life and increases social costs. The Korean Society of Neurogastroenterology and Motility published clinical practice guidelines on the management of IBS based on a systematic review of the literature in 2017, and planned to revise these guidelines in light of new evidence on the pathophysiology, diagnosis, and management of IBS. The current revised version of the guidelines is consistent with the previous version and targets adults diagnosed with or suspected of having IBS. These guidelines were developed using a combination of de novo and adaptation methods, with analyses of existing guidelines and discussions within the committee, leading to the identification of key clinical questions. Finally, the guidelines consisted of 22 recommendations, including 3 concerning the definition and risk factors of IBS, 4 regarding diagnostic modalities and strategies, 2 regarding general management, and 13 regarding medical treatment. For each statement, the advantages, disadvantages, and precautions were thoroughly detailed. The modified Delphi method was used to achieve expert consensus to adopt the core recommendations of the guidelines. These guidelines serve as a reference for clinicians (including primary care physicians, general healthcare providers, medical students, residents, and other healthcare professionals) and patients, helping them to make informed decisions regarding IBS management.
Collapse
Affiliation(s)
- Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonngi-do, Korea
| | - Young Hoon Youn
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Joo Kang
- Department of Internal Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Jeong Eun Shin
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Chungcheongnam-do, Korea
| | - Young Sin Cho
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Chungcheongnam-do, Korea
| | - Yoon Suk Jung
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Yong Shin
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Cheal Wung Huh
- Department of Internal Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yoo Jin Lee
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, Korea
| | - Hoon Sup Koo
- Department of Internal Medicine, Konyang University Hospital, Daejeon, Korea
| | - Kwangwoo Nam
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Chungcheongnam-do, Korea
| | - Hong Sub Lee
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Korea
| | - Dong Hyun Kim
- Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| | - Ye Hyun Park
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Min Cheol Kim
- Department of Internal Medicine, Yeungnam University Hospital, Daegu, Korea
| | - Hyo Yeop Song
- Department of Internal Medicine and Digestive Disease Research Institute, Wonkwang University School of Medicine, Iksan, Jeonbuk State, Korea
| | - Sung-Hoon Yoon
- Department of Psychiatry, Wonkwang University Hospital, Iksan, Jeonbuk State, Korea
| | - Sang Yeol Lee
- Department of Psychiatry, Wonkwang University Hospital, Iksan, Jeonbuk State, Korea
| | - Miyoung Choi
- Division of Healthcare Technology Assessment Research, National Evidence-Based Healthcare Collaborating Agency, Seoul, Korea
| | - Moo-In Park
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - In-Kyung Sung
- Department of Internal Medicine, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
3
|
Cuffe MS, Staudacher HM, Aziz I, Adame EC, Krieger-Grubel C, Madrid AM, Ohlsson B, Black CJ, Ford AC. Efficacy of dietary interventions in irritable bowel syndrome: a systematic review and network meta-analysis. Lancet Gastroenterol Hepatol 2025:S2468-1253(25)00054-8. [PMID: 40258374 DOI: 10.1016/s2468-1253(25)00054-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Patients with irritable bowel syndrome (IBS) are often interested in dietary interventions as a means of managing their symptoms. However, the relative efficacy of available diets for the management of IBS is unclear. We aimed to examine the relative efficacy of various dietary interventions in IBS. METHODS For this systematic review and network meta-analysis we searched MEDLINE, EMBASE, EMBASE Classic, and the Cochrane Central Register of Controlled Trials from database inception to Feb 7, 2025, to identify randomised controlled trials comparing an active dietary intervention requiring changes to the intake of more than one food in IBS with either a control intervention, such as a habitual diet, sham diet, a high fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) diet, or alternative miscellaneous dietary advice, or any other active dietary intervention requiring changes to the intake of more than one food. We assessed efficacy using dichotomous assessments of improvement in global IBS symptoms or improvement in individual IBS symptoms, including abdominal pain, abdominal bloating or distension, and bowel habit. We pooled data using a random-effects model, with the efficacy of each intervention reported as pooled relative risks (RRs) with 95% CIs. We ranked interventions according to their P-score, which measures the mean extent of certainty that one intervention is better than another, averaged over all competing interventions. FINDINGS We identified 28 eligible randomised controlled trials (comprising 2338 patients) of 11 different dietary interventions compared with four control interventions, of which six (low FODMAP diet, British Dietetic Association/National Institute for Health and Care Excellence [BDA/NICE] diet, lactose-reduced diet, starch-reduced and sucrose-reduced diet, a personalised diet, and a Mediterranean diet) were studied in more than one trial. For global IBS symptoms, assessed in 28 randomised controlled trials and when considering only the dietary interventions studied in more than one trial, a starch-reduced and sucrose-reduced diet ranked first (RR of global IBS symptoms not improving 0·41 [95% CI 0·26-0·67]; P-score 0·84; two trials), a low FODMAP diet ranked fourth (0·51 [0·37-0·70]; P-score 0·71; 24 trials), and a BDA/NICE diet ranked tenth (0·62 [0·43-0·90]; P-score 0·44; eight trials), versus a habitual diet. For abdominal pain, assessed in 26 trials and when considering only the dietary interventions studied in more than one randomised controlled trial, a starch-reduced and sucrose-reduced diet ranked second (RR of abdominal pain not improving 0·54 [95% CI 0·33-0·90]; P-score 0·73; two trials), and a low FODMAP diet ranked fifth (0·61 [0·42-0·89]; P-score 0·64; 23 trials), versus a habitual diet. For abdominal bloating or distension, assessed in 26 trials and when considering only the dietary interventions studied in more than one randomised trial, only a low FODMAP diet (RR of abdominal bloating or distension not improving 0·55 [95% CI 0·37-0·80]; P-score 0·64; 23 trials) was superior to a habitual diet and ranked fourth. For bowel habit, assessed in 23 randomised trials, none of the dietary interventions was superior to any of the control interventions, but a low FODMAP diet was superior to a BDA/NICE diet (RR of bowel habit not improving 0·79 [95% CI 0·63-0·99]). All comparisons across the network were rated as low or very low confidence, except for direct comparisons between a low FODMAP diet or a starch-reduced and sucrose-reduced diet and habitual diet, both of which were rated as moderate confidence. INTERPRETATION In terms of dietary interventions for IBS, the most evidence exists for a low FODMAP diet, but other promising therapies are emerging and should be the subject of further study. FUNDING None.
Collapse
Affiliation(s)
- Melanie S Cuffe
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Heidi M Staudacher
- Monash University, Department of Medicine, School of Translational Medicine, Melbourne, VIC, Australia
| | - Imran Aziz
- Academic Unit of Gastroenterology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Enrique Coss Adame
- Department of Gastroenterology/GI Motility Lab, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", México City, México
| | - Claudia Krieger-Grubel
- Department of Gastroenterology/Hepatology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Ana Maria Madrid
- Section of Gastroenterology, Department of Medicine, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Malmo, Sweden; Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Christopher J Black
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Alexander C Ford
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK; Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
| |
Collapse
|
4
|
Oh CK, Chung HH, Kim YJ, Kim JB. Comparison of Rifaximin Monotherapy and Rifaximin Combined with Probiotics in Patients with Irritable Bowel Syndrome: A Randomized Controlled Trial. Nutrients 2025; 17:763. [PMID: 40077633 PMCID: PMC11901931 DOI: 10.3390/nu17050763] [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: 01/17/2025] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objective: Rifaximin is a nonabsorbable antibiotic used to treat irritable bowel syndrome (IBS). Recent studies on Helicobacter pylori eradication treatment have reported synergistic effects and low adverse effects when antibiotics are used in combination with probiotics; yet, such studies have not been conducted in IBS. Probiotics can enhance gut microbiota modulation, inhibition of pathogen adhesion to the gut epithelia, improvement in gut barrier function, anti-inflammatory effects, and improvement of gut immunity. Therefore, this study aimed to investigate the efficacy and safety of rifaximin in combination with probiotics compared to rifaximin monotherapy in patients with IBS. Methods: Patients with IBS were randomly allocated to receive rifaximin monotherapy or a combination of rifaximin and probiotics. The primary outcome was the response rate of the total IBS severity scoring system (IBS-SSS) score (>50-point decrease). Secondary outcomes were based on the response rate of the IBS quality of life (IBS-QOL) score and the IBS-SSS1 subscore (>10-point decrease in both scores). Results: Among 70 patients, the responder rates for the total IBS-SSS score were 65.7% in the combination therapy group and 31.4% in the monotherapy group at weeks 4 and 8, respectively (p = 0.004). The responder rates for IBS-QOL were 65.7% versus (vs.) 37.1% and 65.7% vs. 34.2% at weeks 4 and 8, respectively (p = 0.017 and p = 0.009, respectively). The IBS-SSS1 subscore responder rates were 65.7% vs. 40.0% at week 4 and 68.6% vs. 37.1% at 8 weeks (p = 0.031 and p = 0.017, respectively). Conclusions: Rifaximin combined with probiotics was superior to rifaximin monotherapy in patients with IBS. This combination therapy is considered an effective and safe treatment option for patients with IBS. However, further studies are needed to investigate the mechanisms of therapy and long-term outcomes.
Collapse
Affiliation(s)
- Chang Kyo Oh
- Division of Gastroenterology, Department of Internal Medicine, Kangnam Sacred Heart Hospital, College of Medicine, Hallym University of Korea, Seoul 07441, Republic of Korea; (H.H.C.); (Y.J.K.); (J.B.K.)
| | | | | | | |
Collapse
|
5
|
Rexwinkel R, Vermeijden NK, Zeevenhooven J, Kelder J, Groeneweg M, Hummel T, Goede J, van Wering H, Stapelbroek J, Benninga M, Vlieger A. The low FODMAP diet in adolescents functional abdominal in a non-guided setting: a prospective multicenter cohort study. Eur J Pediatr 2025; 184:189. [PMID: 39934502 PMCID: PMC11814023 DOI: 10.1007/s00431-025-05999-9] [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: 10/22/2024] [Revised: 12/23/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
The purpose of this study is to evaluate the efficacy of a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) in adolescents with irritable bowel syndrome (IBS) and functional abdominal pain-not otherwise specified (FAP-NOS) in a non-guided setting, resembling clinical practice. This prospective multicenter cohort study conducted in 13 centers included patients aged 12-18 years diagnosed with IBS or FAP-NOS. Patients received educational material on FODMAPs, including extensive lists of high and low FODMAP foods and additional online information. They were instructed to replace high FODMAP foods with low FODMAP alternatives for the duration of 4 weeks. No dietician was consulted. The primary end point was the proportion of patients with treatment success (≥ 30% reduction of abdominal pain intensity) at 4 weeks. The key secondary outcome was adequate relief of IBS/FAP-NOS symptoms. Of the 325 included patients, 81 patients (24.9%) achieved treatment success (≥ 30% reduction of abdominal pain intensity) after 4 weeks, with higher rates in patients with IBS (29.3%) than FAP-NOS (16.8%, OR 2.16 (1.04-4.48)). Adequate relief was reported in 51 patients (15.7%). There was a significant decrease in abdominal pain intensity (2.2 (1.1) vs. 2.5 (1.0), P < 0.001), daily bloating (2.4 (2.1) vs. 2.8 (2.3), P < 0.001), and flatulence (2.4 (2.1) vs. 2.8 (2.3), P = 0.001). Adverse events were mild and infrequent. CONCLUSION The low FODMAP diet in a non-guided setting, mimicking clinical practice, yielded treatment success in almost 30% adolescents with IBS and 17% in FAP-NOS, suggesting it may not be the first treatment option for these patients. TRIAL REGISTRATION EUCTR2015-003293-32-NL. WHAT IS KNOWN • Irritable bowel syndrome (IBS) and functional abdominal pain-not otherwise specified (FAP-NOS) are common disorders in children which negatively impact quality of life. • While a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) has demonstrated effectiveness in adult IBS, its efficacy in pediatric IBS and FAP-NOS remains uncertain. • Clinical application of the low FODMAP diet often occurs without dietician consultation, contrary to controlled trial settings. WHAT IS NEW • The low FODMAP diet, without dietician guidance, resulted in treatment success in almost 30% of adolescents with IBS and only 17% with FAP-NOS. • With only 15.7% of participants achieving adequate relief of IBS/FAP-NOS symptoms, the non-guided low FODMAP diet may not be the first treatment option for pediatric IBS and FAP-NOS.
Collapse
Affiliation(s)
- Robyn Rexwinkel
- Emma's Children Hospital, Amsterdam UMC, Location AMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Room C2-312, PO Box 22700, 1100 DD, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Centers, Location Academic Medical Center/Emma Children's Hospital, Amsterdam, The Netherlands
- Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute,, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Nicolaas Koen Vermeijden
- Emma's Children Hospital, Amsterdam UMC, Location AMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Room C2-312, PO Box 22700, 1100 DD, Amsterdam, The Netherlands.
- Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Centers, Location Academic Medical Center/Emma Children's Hospital, Amsterdam, The Netherlands.
- Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism Research Institute,, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
- Department of Pediatrics, St Antonius Hospital, Nieuwegein, The Netherlands.
| | - Judith Zeevenhooven
- Emma's Children Hospital, Amsterdam UMC, Location AMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Room C2-312, PO Box 22700, 1100 DD, Amsterdam, The Netherlands
| | - Johannes Kelder
- Department of Cardiology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Michael Groeneweg
- Department of Pediatrics, Maasstad Hospital, Rotterdam, The Netherlands
| | - Thalia Hummel
- Department of Pediatrics, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Joery Goede
- Department of Pediatrics, Spaarne Gasthuis, Haarlem, The Netherlands
| | | | | | - Marc Benninga
- Emma's Children Hospital, Amsterdam UMC, Location AMC, University of Amsterdam, Pediatric Gastroenterology, Hepatology and Nutrition, Room C2-312, PO Box 22700, 1100 DD, Amsterdam, The Netherlands
| | - Arine Vlieger
- Department of Pediatrics, St Antonius Hospital, Nieuwegein, The Netherlands
| |
Collapse
|
6
|
Costa A, Lucarini E. Treating chronic stress and chronic pain by manipulating gut microbiota with diet: can we kill two birds with one stone? Nutr Neurosci 2025; 28:221-244. [PMID: 38889540 DOI: 10.1080/1028415x.2024.2365021] [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: 06/20/2024]
Abstract
Background: Chronic stress and chronic pain are closely linked by the capacity to exacerbate each other, sharing common roots in the brain and in the gut. The strict intersection between these two neurological diseases makes important to have a therapeutic strategy aimed at preventing both to maintain mental health in patients. Diet is an modifiable lifestyle factor associated with gut-brain axis diseases and there is growing interest in its use as adjuvant to main therapies. Several evidence attest the impact of specific diets or nutrients on chronic stress-related disorders and pain with a good degree of certainty. A daily adequate intake of foods containing micronutrients such as amino acids, minerals and vitamins, as well as the reduction in the consumption of processed food products can have a positive impact on microbiota and gut health. Many nutrients are endowed of prebiotic, anti-inflammatory, immunomodulatory and neuroprotective potential which make them useful tools helping the management of chronic stress and pain in patients. Dietary regimes, as intermittent fasting or caloric restriction, are promising, although further studies are needed to optimize protocols according to patient's medical history, age and sex. Moreover, by supporting gut microbiota health with diet is possible to attenuate comorbidities such as obesity, gastrointestinal dysfunction and mood disorders, thus reducing healthcare costs related to chronic stress or pain.Objective: This review summarize the most recent evidence on the microbiota-mediated beneficial effects of macro- and micronutrients, dietary-related factors, specific nutritional regimens and dietary intervention on these pathological conditions.
Collapse
Affiliation(s)
- Alessia Costa
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Area and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| |
Collapse
|
7
|
Zhang H, Su Q. Low-FODMAP Diet for Irritable Bowel Syndrome: Insights from Microbiome. Nutrients 2025; 17:544. [PMID: 39940404 PMCID: PMC11819959 DOI: 10.3390/nu17030544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent gastrointestinal disorder characterized by chronic abdominal pain, bloating, and altered bowel habits. Low-FODMAP diets, which involve restricting fermentable oligosaccharides, disaccharides, monosaccharides, and polyols, have emerged as an effective dietary intervention for alleviating IBS symptoms. This review paper aims to synthesize current insights into the impact of a low-FODMAP diet on the gut microbiome and its mechanisms of action in managing IBS. We explore the alterations in microbial composition and function associated with a low-FODMAP diet and discuss the implications of these changes for gut health and symptom relief. Additionally, we examine the balance between symptom improvement and potential negative effects on microbial diversity and long-term gut health. Emerging evidence suggests that while a low-FODMAP diet can significantly reduce IBS symptoms, it may also lead to reductions in beneficial microbial populations. Strategies to mitigate these effects, such as the reintroduction phase and the use of probiotics, are evaluated. This review highlights the importance of a personalized approach to dietary management in IBS, considering individual variations in microbiome responses. Understanding the intricate relationship between diet, the gut microbiome, and IBS symptomatology will guide the development of more effective, sustainable dietary strategies for IBS patients.
Collapse
Affiliation(s)
- Haoshuai Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qi Su
- Microbiota I-Center (MagIC), Hong Kong SAR, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
8
|
Lei Y, Sun X, Ruan T, Lu W, Deng B, Zhou R, Mu D. Effects of Probiotics and Diet Management in Patients With Irritable Bowel Syndrome: A Systematic Review and Network Meta-analysis. Nutr Rev 2025:nuae217. [PMID: 39862384 DOI: 10.1093/nutrit/nuae217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025] Open
Abstract
CONTEXT The efficacy of probiotics and diet management in irritable bowel syndrome (IBS) is controversial, and their relative effectiveness remains unclear. OBJECTIVE This study aimed to evaluate the effects of probiotics, diet management, and their combination on IBS. DATA SOURCES PubMed, Embase, Cochrane, and Web of Science were searched from inception to July 10, 2023, for relevant studies, including symptom relief, IBS-symptom severity score (-SSS), and IBS-quality of life measure (-QOL). DATA EXTRACTION Two investigators independently performed the data extraction and quality assessment. DATA ANALYSIS A network meta-analysis was performed using a frequentist approach and a random-effects model to estimate the relative risk (RR) and 95% CI. RESULTS Forty-four articles were eligible for this study. In relieving IBS symptoms, compared with a sham diet, a low-fermentable oligosaccharide, disaccharide, monosaccharide, and polyols (low-FODMAP) diet (RR: 3.22; 95% CI: 1.70-6.26) and low-FODMAP diet combined with probiotics (RR: 17.79; 95% CI: 3.27-112.54) significantly relieved IBS symptoms. The control group showed significantly lower effectiveness than the probiotics group (RR: 0.47; 95% CI: 0.32-0.69). According to the surface under the cumulative rank curve (SUCRA), a low-FODMAP diet combined with probiotics (80.4%) had the best effect in relieving IBS symptoms, followed by a low-FODMAP diet (70.8%), probiotics (65.1%), and a gluten-free diet (54.3%). In reducing the total IBS-SSS, the low-FODMAP diet (90.5%) was the most effective, followed by the low-FODMAP diet combined with probiotics (76.6%), probiotics alone (62.3%), and gluten-free diet (28.3%). In reducing total IBS-QOL, probiotics (72.1%) ranked first, followed by gluten-free (57.0%) and low-FODMAP (56.9%) diets. Probiotics (34.9%) were associated with the lowest risk of adverse effects. CONCLUSION A low-FODMAP diet combined with probiotics is most effective in relieving IBS symptoms. A low-FODMAP diet is the most recommended diet for alleviating IBS severity, and probiotics were associated with improving the QOL of patients with IBS, with the fewest adverse events. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42024499113.
Collapse
Affiliation(s)
- Yupeng Lei
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuemei Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tiechao Ruan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenting Lu
- Integrated Care Management Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bixin Deng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ruixi Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Dezhi Mu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan 610041, China
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
9
|
El-Shafie S, Metwaly A. Diet-specific impacts on the gut microbiome and their relation to health and inflammation. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:77-124. [DOI: 10.1016/b978-0-443-18979-1.00005-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
10
|
Khan Z, Muhammad SA, Amin MS, Gul A. The Efficacy of the Low-FODMAP (Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols) Diet in Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis. Cureus 2025; 17:e77053. [PMID: 39917138 PMCID: PMC11799870 DOI: 10.7759/cureus.77053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Irritable bowel syndrome (IBS) is frequently observed in clinical practice and affects people from different parts of the world. The pathogenesis and aetiology are not well-defined or fully understood; however, altered bowel movements, psychological factors, and visceral hypersensitivity may contribute to symptoms via a pathway mediated by serotonin and other enteric neurotransmitters. Altered bowel movements, including diarrhoea and constipation, abdominal pain relieved by passing flatus, and bloating are the main salient features of this condition. This systematic review and meta-analysis aimed to determine the effectiveness and efficacy of a low-fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (low-FODMAP) diet in these patients. Systematic searches were conducted on PubMed, Medline, Google Scholar, and Cochrane Library. Randomised controlled trials (RCTs), systematic trials and cohort studies that included keywords about IBS and a low-FODMAP diet were included. Exclusion criteria included studies that were not in the English language, not relevant to IBS, diet-related to inflammatory bowel disease, or not pertinent to the subject. A total of 41 studies were included in this systematic review and meta-analysis. There was significant heterogeneity among the RCTs; hence, a random-effects model was used. The systematic review included a total of 8460 patients across 36 studies, with follow-up durations ranging from 11 to 16 months. Specifically, the meta-analysis included 15 RCTs with 1118 participants and follow-up durations from two days to nine weeks and six cohort studies including 292 patients with follow-up durations from two weeks to two years. The risk ratio (RR) was 1.21 (95% confidence interval= 0.98-1.51), and the I2 value was 63% for global symptom improvement with a low-FODMAP diet using a random-effects model. There was a low risk of bias in the RCTs. Five studies were included evaluating the effect of a low-FODMAP diet on quality of life, and these studies did not show any statistically significant benefit of a low-FODMAP diet on quality of life, although a mean difference of 4.59 (95% CI 1.50-7.67) was observed. The risk of bias was moderate to severe in the observational studies included in this review. Food intolerance is increasingly recognised as a contributory factor in IBS, and its role in the pathogenesis and precipitation of symptoms is being explored. Specific mechanisms include the fermentation of FODMAPs by the gut microbiota, leading to gas production and subsequent symptoms.
Collapse
Affiliation(s)
- Zahid Khan
- Cardiology, University of South Wales, Pontypridd, GBR
- Cardiology, University of Buckingham, Buckingham, GBR
- Cardiology, Barts Heart Centre, London, GBR
| | - Syed Aun Muhammad
- Cardiology, Mid and South Essex NHS Foundation Trust, Southend-on-Sea, GBR
| | - Mehul S Amin
- Internal Medicine, Southend University Hospital, Southend-on-Sea, GBR
| | - Amresh Gul
- General Practice, GP Clinic, Brisbane, AUS
| |
Collapse
|
11
|
Tursi A, Piovani D, Brandimarte G, Di Mario F, Elisei W, Picchio M, Figlioli G, Bassotti G, Allegretta L, Annunziata ML, Bafutto M, Bianco MA, Colucci R, Conigliaro R, Dumitrascu DL, Escalante R, Ferrini L, Forti G, Franceschi M, Graziani MG, Lammert F, Latella G, Lisi D, Maconi G, Compare D, Nardone G, Camara de Castro Oliveira L, Enio CO, Papagrigoriadis S, Pietrzak A, Pontone S, Stundiene I, Poškus T, Pranzo G, Reichert MC, Rodino S, Regula J, Scaccianoce G, Scaldaferri F, Vassallo R, Zampaletta C, Zullo A, Spaziani E, Bonovas S, Papa A, Danese S. Bowel movement alterations predict the severity of diverticular disease and the risk of acute diverticulitis: a prospective, international study. Intest Res 2025; 23:96-106. [PMID: 39129357 PMCID: PMC11834362 DOI: 10.5217/ir.2024.00046] [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: 03/22/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND/AIMS Patients with diverticular disease (DD) frequently have abnormal bowel movements. However, it is unknown whether the entity of these alterations is associated with the severity of DD. We aimed to assess bowel habits and their relationship with the severity of DD according to Diverticular Inflammation and Complication Assessment (DICA) classification, Combined Overview on Diverticular Assessment (CODA) score, and fecal calprotectin (FC). METHODS An international, multicenter, prospective cohort study was conducted in 43 centers. A 10-point visual analog scale (VAS) was used to assess the severity of constipation and diarrhea. The association of constipation and diarrhea with DICA classification, CODA score, and basal FC was tested using non-parametric tests. Survival methods for censored observations were applied to test the association of constipation and diarrhea with the incidence of acute diverticulitis over a 3-year follow-up. RESULTS Of 871 patients with DD were included in the study. Of these, 208 (23.9%) and 199 (22.9%) reported a VAS score for constipation and diarrhea at least 3 at baseline, respectively. Higher constipation and diarrhea scores were associated with increasing DICA classification, CODA score and basal FC (P< 0.001). Constipation and diarrhea scores were independently associated with an increased hazard of developing acute diverticulitis (hazard ratio [HR]constipation = 1.15 per 1-VAS point increase, 95% confidence interval [CI], 1.04-1.27; P=0.004; and HRdiarrhea =1.14; 95% CI, 1.03-1.26; P=0.014, respectively). CONCLUSIONS In newly diagnosed patients with DD, higher endoscopic and combined scores of DD severity were associated with higher scores of constipation and diarrhea at baseline. Both constipation and diarrhea were independent prognostic factors of acute diverticulitis.
Collapse
Affiliation(s)
- Antonio Tursi
- Territorial Gastroenterology Service, ASL BAT, Andria, Italy
- Department of Medical and Surgical Sciences, School of Medicine, Catholic University, Rome, Italy
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giovanni Brandimarte
- Division of Internal Medicine and Gastroenterology, Cristo Re Hospital, Rome, Italy
| | - Francesco Di Mario
- Department of Medical and Surgical Sciences, Gastroenterology Unit, University of Parma, Parma, Italy
| | - Walter Elisei
- Division of Gastroenterology, San Camillo Hospital, Rome, Italy
| | - Marcello Picchio
- Division of Surgery, P. Colombo Hospital, ASL RM6, Velletri, Italy
| | - Gisella Figlioli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Gabrio Bassotti
- Department of Medicine and Surgery, Gastroenterology and Hepatology Unit, Santa Maria della Misericordia University Hospital, University of Perugia, Perugia, Italy
| | - Leonardo Allegretta
- Division of Gastroenterology, Santa Caterina Novella Hospital, Galatina, Italy
| | | | - Mauro Bafutto
- Institute of Gastroenterology and Digestive Endoscopy, Goiânia, Brazil
| | | | - Raffaele Colucci
- Digestive Endoscopy Unit, San Matteo degli Infermi Hospital, Spoleto, Italy
| | - Rita Conigliaro
- Digestive Endoscopy Unit, Sant’Agostino Estense Hospital, Baggiovara, Italy
| | - Dan L. Dumitrascu
- Second Medical Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ricardo Escalante
- Central University of Venezuela, Loira Medical Center, Caracas, Venezuela
| | - Luciano Ferrini
- Service of Digestive Endoscopy, Villa dei Pini Home Care, Civitanova Marche, Italy
| | - Giacomo Forti
- Digestive Endoscopy Unit, Santa Maria Goretti Hospital, Latina, Italy
| | | | - Maria Giovanna Graziani
- Division of Gastroenterology and Digestive Endoscopy, San Giovanni Addolorata Hospital, Rome, Italy
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
- Health Sciences, Hannover Medical School (MHH), Hannover, Germany
| | - Giovanni Latella
- Division of Gastroenterology, Hepatology and Nutrition, Department of Life, Health & Environmental Sciences, University of L’Aquila, San Salvatore Hospital, L’Aquila, Italy
| | - Daniele Lisi
- Territorial Gastroenterology Service, ASL Rm B, Rome, Italy
| | - Giovanni Maconi
- Division of Gastroenterology, L. Sacco University Hospital, Milan, Italy
| | - Debora Compare
- Department of Clinical Medicine and Surgery, Division of Gastroenterology and Hepatology, Federico II University Hospital, Naples, Italy
| | - Gerardo Nardone
- Department of Clinical Medicine and Surgery, Division of Gastroenterology and Hepatology, Federico II University Hospital, Naples, Italy
| | | | | | | | - Anna Pietrzak
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education and Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Stefano Pontone
- First Division of General Surgery, Umberto I University Hospital, Sapienza University, Rome, Italy
| | - Ieva Stundiene
- Institute of Clinical Medicine, Vilnius University Hospital, Vilnius, Lithuania
| | - Tomas Poškus
- Institute of Clinical Medicine, Vilnius University Hospital, Vilnius, Lithuania
| | - Giuseppe Pranzo
- Digestive Endoscopy Unit, Valle d’Itria Hospital, Martina Franca, Italy
| | | | - Stefano Rodino
- Division of Gastroenterology, Pugliese-Ciaccio Hospital, Catanzaro, Italy
| | - Jaroslaw Regula
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education and Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Franco Scaldaferri
- Division of Internal Medicine and Gastroenterology, IRCCS A. Gemelli Hospital, Fondazione Policlinico Gemelli, Catholic University, Rome, Italy
| | - Roberto Vassallo
- Division of Gastroenterology and Digestive Endoscopy, Buccheri La Ferla Hospital, Palermo, Italy
| | | | - Angelo Zullo
- Division of Gastroenterology, Nuovo Regina Margherita Hospital, Rome, Italy
| | - Erasmo Spaziani
- Department of Surgery, Sapienza University of Rome-Polo Pontino, Terracina, Italy
| | - Stefanos Bonovas
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alfredo Papa
- Division of Internal Medicine and Gastroenterology, IRCCS A. Gemelli Hospital, Fondazione Policlinico Gemelli, Catholic University, Rome, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milan, Italy
| | - DICA International Group
- Territorial Gastroenterology Service, ASL BAT, Andria, Italy
- Department of Medical and Surgical Sciences, School of Medicine, Catholic University, Rome, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
- Division of Internal Medicine and Gastroenterology, Cristo Re Hospital, Rome, Italy
- Department of Medical and Surgical Sciences, Gastroenterology Unit, University of Parma, Parma, Italy
- Division of Gastroenterology, San Camillo Hospital, Rome, Italy
- Division of Surgery, P. Colombo Hospital, ASL RM6, Velletri, Italy
- Department of Medicine and Surgery, Gastroenterology and Hepatology Unit, Santa Maria della Misericordia University Hospital, University of Perugia, Perugia, Italy
- Division of Gastroenterology, Santa Caterina Novella Hospital, Galatina, Italy
- Division of Gastroenterology, San Donato Hospital, San Donato Milanese, Italy
- Institute of Gastroenterology and Digestive Endoscopy, Goiânia, Brazil
- Division of Gastroenterology, T. Maresca Hospital, Torre del Greco, Italy
- Digestive Endoscopy Unit, San Matteo degli Infermi Hospital, Spoleto, Italy
- Digestive Endoscopy Unit, Sant’Agostino Estense Hospital, Baggiovara, Italy
- Second Medical Department, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Central University of Venezuela, Loira Medical Center, Caracas, Venezuela
- Service of Digestive Endoscopy, Villa dei Pini Home Care, Civitanova Marche, Italy
- Digestive Endoscopy Unit, Santa Maria Goretti Hospital, Latina, Italy
- Digestive Endoscopy Unit, ULSS 7 Alto Vicentino, Santorso, Italy
- Division of Gastroenterology and Digestive Endoscopy, San Giovanni Addolorata Hospital, Rome, Italy
- Department of Medicine II, Saarland University Medical Center, Homburg, Germany
- Health Sciences, Hannover Medical School (MHH), Hannover, Germany
- Division of Gastroenterology, Hepatology and Nutrition, Department of Life, Health & Environmental Sciences, University of L’Aquila, San Salvatore Hospital, L’Aquila, Italy
- Territorial Gastroenterology Service, ASL Rm B, Rome, Italy
- Division of Gastroenterology, L. Sacco University Hospital, Milan, Italy
- Department of Clinical Medicine and Surgery, Division of Gastroenterology and Hepatology, Federico II University Hospital, Naples, Italy
- Department of Anorectal Physiology, São José Home Care, Rio de Janeiro, Brazil
- Department of Colorectal Surgery, Federal University of Goiás, Goiânia, Brazil
- Department of Colorectal Surgery, King’s College Hospital, London, UK
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education and Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
- First Division of General Surgery, Umberto I University Hospital, Sapienza University, Rome, Italy
- Institute of Clinical Medicine, Vilnius University Hospital, Vilnius, Lithuania
- Digestive Endoscopy Unit, Valle d’Itria Hospital, Martina Franca, Italy
- Division of Gastroenterology, Pugliese-Ciaccio Hospital, Catanzaro, Italy
- Digestive Endoscopy Unit, F. Perinei Hospital, Altamura, Italy
- Division of Internal Medicine and Gastroenterology, IRCCS A. Gemelli Hospital, Fondazione Policlinico Gemelli, Catholic University, Rome, Italy
- Division of Gastroenterology and Digestive Endoscopy, Buccheri La Ferla Hospital, Palermo, Italy
- Division of Gastroenterology, Belcolle Hospital, Viterbo, Italy
- Division of Gastroenterology, Nuovo Regina Margherita Hospital, Rome, Italy
- Department of Surgery, Sapienza University of Rome-Polo Pontino, Terracina, Italy
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele and University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
12
|
Zhao Y, Zhu S, Dong Y, Xie T, Chai Z, Gao X, Dai Y, Wang X. The Role of Gut Microbiome in Irritable Bowel Syndrome: Implications for Clinical Therapeutics. Biomolecules 2024; 14:1643. [PMID: 39766350 PMCID: PMC11674646 DOI: 10.3390/biom14121643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder (FGID) characterized by chronic or recurrent gastrointestinal symptoms without organic changes, and it is also a common disorder of gut-brain interaction (DGBIs).. The symptoms of IBS not only affect the quality of life for individual patients but also place a significant burden on global healthcare systems. The lack of established and universally applicable biomarkers for IBS, along with the substantial variability in symptoms and progression, presents challenges in developing effective clinical treatments. In recent years, preclinical and clinical studies have linked the pathogenesis of IBS to alterations in the composition and function of the intestinal microbiota. Within the complex microbial community of the gut, intricate metabolic and spatial interactions occur among its members and between microbes and their hosts. Amid the multifaceted pathophysiology of IBS, the role of intestinal microenvironment factors in symptom development has become more apparent. This review aims to delve into the changes in the composition and structure of the gut microbiome in individuals with IBS. It explores how diet-mediated alterations in intestinal microbes and their byproducts play a role in regulating the pathogenesis of IBS by influencing the "brain-gut" axis, intestinal barrier function, immune responses, and more. By doing so, this review seeks to lay a theoretical foundation for advancing the development of clinical therapeutics for IBS.
Collapse
Affiliation(s)
- Yucui Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shixiao Zhu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingling Dong
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tian Xie
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhiqiang Chai
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiumei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
| | - Yongna Dai
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
| | - Xiaoying Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (Y.Z.); (S.Z.); (Y.D.); (T.X.); (Z.C.); (X.G.)
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
13
|
Haghbin H, Hasan F, Gangwani MK, Zakirkhodjaev N, Lee-Smith W, Beran A, Kamal F, Hart B, Aziz M. Efficacy of Dietary Interventions for Irritable Bowel Syndrome: A Systematic Review and Network Meta-Analysis. J Clin Med 2024; 13:7531. [PMID: 39768453 PMCID: PMC11728101 DOI: 10.3390/jcm13247531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/01/2024] [Accepted: 12/07/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction: Irritable bowel syndrome (IBS) is a common condition that alters the quality of life of patients. A variety of dietary interventions have been introduced to address this debilitating condition. The low-FODMAP diet (LFD), gluten-free diet (GFD), and Mediterranean diet are examples showing efficacy. The aim of this network meta-analysis was to compare these interventions to find the best approach. Methods: We performed a systematic review of the available literature through 14 March 2024 in the following databases: Embase, PubMed, MEDLINE OVID, Web of Science, CINAHL Plus, and Cochrane Central. We only included randomized controlled trials (RCTs). We used a random effects model and conducted a direct meta-analysis based on the DerSimonian-Laird approach and a network meta-analysis based on the frequentist approach. Mean differences (MDs) with 95% confidence interval (CI) were calculated. The primary outcomes included IBS quality of life (IBS QOL) and IBS symptom severity scale (IBS-SSS). Results: We finalized 23 studies including 1689 IBS patients. In the direct meta-analysis, there was no statistically significant difference in any IBS score between GFD and either LFD or standard diet. Meanwhile, the LFD was statistically superior to the standard diet in the IBS-SSS (MD: -46.29, CI: -63.72--28.86, p < 0.01) and IBS QOL (MD: 4.06, CI: 0.72-7.41, p = 0.02). On ranking, the Mediterranean diet showed the greatest improvement in IBS-SSS, IBS-QOL, distension, dissatisfaction, and general life interference, followed by the LFD alone or in combination with the GFD. Conclusions: We demonstrated the efficacy of dietary interventions such as the LFD and Mediterranean diet in improving IBS. There is a need for large RCTs with head-to-head comparisons, particularly for the Mediterranean diet.
Collapse
Affiliation(s)
- Hossein Haghbin
- Division of Gastroenterology and Hepatology, Bon Secours Mercy Health, Toledo, OH 43608, USA
| | - Fariha Hasan
- Department of Internal Medicine, Cooper University Hospital, Camden, NJ 08103, USA
| | - Manesh Kumar Gangwani
- Department of Gastroenterology, University of Arkansas Medical Sciences, Little Rock, AR 72205, USA
| | - Nurruddinkhodja Zakirkhodjaev
- Division of Occupational and Environmental Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77021, USA
| | | | - Azizullah Beran
- Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Faisal Kamal
- Division of Gastroenterology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Benjamin Hart
- Division of Gastroenterology, University of Toledo, Toledo, OH 43606, USA
| | - Muhammad Aziz
- Division of Gastroenterology and Hepatology, Bon Secours Mercy Health, Toledo, OH 43608, USA
| |
Collapse
|
14
|
Zhang M, Wu X, Gao H, Zhang L, Li Y, Li M, Zhao C, Wei P, Ou L. Chinese Herbal Medicine for Irritable Bowel Syndrome: A Perspective of Local Immune Actions. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2079-2106. [PMID: 39663262 DOI: 10.1142/s0192415x24500800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Irritable bowel syndrome (IBS) is the functional gastrointestinal disorder, characterized by abdominal pain and altered bowel habits. The interest in intestinal immune activation as a potential disease mechanism for IBS has increased exponentially in recent years. This study was designed to summarize the Chinese herbal medicine (CHM) that potentially exert protective effects against IBS through inhibition of intestinal immune activation. We detailed the current evidence that immune activation contributes to the pathology of IBS and discussed the potential mechanisms involved. Then, therapeutic effects and possible mechanisms related to immune response of herbal medicine prescriptions, extracts, and monomers were analyzed. The reasons leading to the aberrant and persistent immune activation noted in IBS are mainly associated with the increased number of mast cells, CD3[Formula: see text] T cells, and CD4[Formula: see text] T cells. The mechanisms mainly focused on the gut microbiota disorder induced alteration of the PGE2/COX2/SERT/5-HT, TLR4/MyD88/NF-κB, and BDNF/TrkB pathways. Most of the CHM alleviated IBS through interventions of intestinal immune activation via gut microbiota related to the TLR4/MyD88/NF-κB and SCF/c-kit pathways. We hope this review will provide some clues for the further development of novel candidate agents for IBS and other intestinal immune disorders.
Collapse
Affiliation(s)
- Mengmeng Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Xu Wu
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
- Engineering Technology Research Center of Shaanxi, Administration of Chinese Herbal Pieces, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Huanqing Gao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Lin Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Yao Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Min Li
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Chongbo Zhao
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
- Engineering Technology Research Center of Shaanxi, Administration of Chinese Herbal Pieces, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Peifeng Wei
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| | - Li Ou
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang 712046, P. R. China
| |
Collapse
|
15
|
Abeltino A, Hatem D, Serantoni C, Riente A, De Giulio MM, De Spirito M, De Maio F, Maulucci G. Unraveling the Gut Microbiota: Implications for Precision Nutrition and Personalized Medicine. Nutrients 2024; 16:3806. [PMID: 39599593 PMCID: PMC11597134 DOI: 10.3390/nu16223806] [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: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
Recent studies have shown a growing interest in the complex relationship between the human gut microbiota, metabolism, and overall health. This review aims to explore the gut microbiota-host association, focusing on its implications for precision nutrition and personalized medicine. The objective is to highlight how gut microbiota modulate metabolic and immune functions, contributing to disease susceptibility and wellbeing. The review synthesizes recent research findings, analyzing key studies on the influence of gut microbiota on lipid and carbohydrate metabolism, intestinal health, neurobehavioral regulation, and endocrine signaling. Data were drawn from both experimental and clinical trials examining microbiota-host interactions relevant to precision nutrition. Our findings highlight the essential role of gut microbiota-derived metabolites in regulating host metabolism, including lipid and glucose pathways. These metabolites have been found to influence immune responses and gut barrier integrity. Additionally, the microbiota impacts broader physiological processes, including neuroendocrine regulation, which could be crucial for dietary interventions. Therefore, understanding the molecular mechanisms of dietary-microbiota-host interactions is pivotal for advancing personalized nutrition strategies. Tailored dietary recommendations based on individual gut microbiota compositions hold promise for improving health outcomes, potentially revolutionizing future healthcare approaches across diverse populations.
Collapse
Affiliation(s)
- Alessio Abeltino
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Duaa Hatem
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Cassandra Serantoni
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Alessia Riente
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Michele Maria De Giulio
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Marco De Spirito
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Flavio De Maio
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Maulucci
- Metabolic Intelligence Lab, Department of Neuroscience, Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy; (A.A.); (D.H.); (C.S.); (A.R.); (M.M.D.G.); (M.D.S.)
- UOC Physics for Life Sciences, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| |
Collapse
|
16
|
Harris CI, Nasar B, Finnerty CC. Nutritional Implications of Mast Cell Diseases. J Acad Nutr Diet 2024; 124:1387-1396. [PMID: 38754765 DOI: 10.1016/j.jand.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Affiliation(s)
| | | | - Celeste C Finnerty
- Division of Surgical Sciences, Department of Surgery, University of Texas Medical Branch, Galveston, Texas; The Mast Cell Disease Society, Inc., Sterling, Massachusetts
| |
Collapse
|
17
|
Lambiase C, Rossi A, Morganti R, Cancelli L, Grosso A, Tedeschi R, Rettura F, Mosca M, de Bortoli N, Bellini M. Adapted Low-FODMAP Diet in IBS Patients with and without Fibromyalgia: Long-Term Adherence and Outcomes. Nutrients 2024; 16:3419. [PMID: 39408383 PMCID: PMC11478509 DOI: 10.3390/nu16193419] [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: 09/05/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES A low-FODMAPs Diet (LFD) is considered a "second line" dietary strategy for irritable bowel syndrome (IBS) but, after a period of strict restriction of all FODMAP foods, it has to be adapted and tailored to each patient (AdLFD). Fibromyalgia often coexists with IBS in up to 65% of cases. Our aims were to evaluate if comorbid fibromyalgia influenced the long-term clinical outcomes and adherence to an AdLFD in IBS patients. METHODS IBS patients with or without fibromyalgia who had started an AdLFD were enrolled. Patients had been evaluated before starting the LFD (T0). After a mean follow-up of 62.5 ± 22.7 months (T1), they were re-evaluated using questionnaires on disease severity, bowel habits, psychological status, and adherence to AdLFD. RESULTS In total, 51 IBS patients entered the study. Nineteen of them had comorbid fibromyalgia. Thirty patients reported a reduction in symptom severity at T1 in comparison with T0. Despite some slight differences in single IBS Symptom Severity Score items, comorbid fibromyalgia did not influence the IBS-SSS total score at T1. Patients with comorbid fibromyalgia showed a higher Hospital Anxiety and Depression Scale (HADS) score at baseline. A total of 44 patients showed good long-term adherence to the AdLFD. All patients improved their HADS score and had long-term adherence to the AdLFD. CONCLUSIONS Comorbid fibromyalgia showed only a slight influence on long-term outcomes of an AdLFD on IBS symptoms, without affecting the relief of global symptoms. No influence on long-term adherence to AdLFD was detected. Hence, this approach can be taken into account in fibromyalgia patients for a nonpharmacological management of IBS symptoms. However, multicentric studies on larger samples would be welcome in the future.
Collapse
Affiliation(s)
- Christian Lambiase
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
- National Institute for Health and Care Research Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| | - Alessandra Rossi
- Rheumatology Division, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Riccardo Morganti
- Clinical Trial Statistical Support Unit, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy
| | - Lorenzo Cancelli
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Antonio Grosso
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Riccardo Tedeschi
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Francesco Rettura
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
- Gastroenterology Unit, Annunziata Hospital, 87100 Cosenza, Italy
| | - Marta Mosca
- Rheumatology Division, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Nicola de Bortoli
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| | - Massimo Bellini
- Gastrointestinal Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
18
|
Green N, Chan C, Ooi CY. The gastrointestinal microbiome, small bowel bacterial overgrowth, and microbiome modulators in cystic fibrosis. Pediatr Pulmonol 2024; 59 Suppl 1:S70-S80. [PMID: 39105345 DOI: 10.1002/ppul.26913] [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: 08/27/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 08/07/2024]
Abstract
People with cystic fibrosis (pwCF) have an altered gastrointestinal microbiome. These individuals also demonstrate propensity toward developing small intestinal bacterial overgrowth (SIBO). The dysbiosis present has intestinal and extraintestinal implications, including potential links with the higher rates of gastrointestinal malignancies described in CF. Given these implications, there is growing interest in therapeutic options for microbiome modulation. Alternative therapies, including probiotics and prebiotics, and current CF transmembrane conductance regulator gene modulators are promising interventions for ameliorating gut microbiome dysfunction in pwCF. This article will characterize and discuss the current state of knowledge and expert opinions on gut dysbiosis and SIBO in the context of CF, before reviewing the current evidence supporting gut microbial modulating therapies in CF.
Collapse
Affiliation(s)
- Nicole Green
- Department of Pediatrics, Division of Gastroenterology and Hepatology, Seattle Children's Hospital, University of Washington, Seattle, Washington, USA
| | - Christopher Chan
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Chee Y Ooi
- Discipline of Paediatrics & Child Health, Randwick Clinical Campus, School of Clinical Medicine, UNSW Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Gastroenterology, Sydney Children's Hospital, Randwick, New South Wales, Australia
| |
Collapse
|
19
|
Conley TE, Slater R, Moss S, Bulmer DC, Negro JDLR, Ijaz UZ, Pritchard DM, Parkes M, Probert C. Microbiome-driven IBS metabotypes influence response to the low FODMAP diet: insights from the faecal volatome. EBioMedicine 2024; 107:105282. [PMID: 39173527 PMCID: PMC11388012 DOI: 10.1016/j.ebiom.2024.105282] [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: 02/13/2024] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a common and debilitating disorder manifesting with abdominal pain and bowel dysfunction. A mainstay of treatment is dietary modification, including restriction of FODMAPs (fermentable oligosaccharides, disaccharides, monosaccharides and polyols). A greater response to a low FODMAP diet has been reported in those with a distinct IBS microbiome termed IBS-P. We investigated whether this is linked to specific changes in the metabolome in IBS-P. METHODS Solid phase microextraction gas chromatography-mass spectrometry was used to examine the faecal headspace of 56 IBS cases (each paired with a non-IBS household control) at baseline, and after four-weeks of a low FODMAP diet (39 pairs). 50% cases had the IBS-P microbial subtype, while the others had a microbiome that more resembled healthy controls (termed IBS-H). Clinical response to restriction of FODMAPs was measured with the IBS-symptom severity scale, from which a pain sub score was calculated. FINDINGS Two distinct metabotypes were identified and mapped onto the microbial subtypes. IBS-P was characterised by a fermentative metabolic profile rich in short chain fatty acids (SCFAs). After FODMAP restriction significant reductions in SCFAs were observed in IBS-P. SCFA levels did not change significantly in the IBS-H group. The magnitude of pain and overall symptom improvement were significantly greater in IBS-P compared to IBS-H (p = 0.016 and p = 0.026, respectively). Using just five metabolites, a biomarker model could predict microbial subtype with accuracy (AUROC 0.797, sensitivity 78.6% (95% CI: 0.78-0.94), specificity 71.4% (95% CI: 0.55-0.88). INTERPRETATION A metabotype high in SCFAs can be manipulated by restricting fermentable carbohydrate, and is associated with an enhanced clinical response to this dietary restriction. This implies that SCFAs harbour pro-nociceptive potential when produced in a specific IBS niche. By ascertaining metabotype, microbial subtype can be predicted with accuracy. This could allow targeted FODMAP restriction in those seemingly primed to respond best. FUNDING This research was co-funded by Addenbrooke's Charitable Trust, Cambridge University Hospitals and the Wellcome Sanger Institute, and supported by the NIHR Cambridge Biomedical Research Centre (BRC-1215-20014).
Collapse
Affiliation(s)
- Thomas Edward Conley
- University of Liverpool Institute of Systems, Molecular and Integrative Biology, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Department of Gastroenterology, Liverpool, UK.
| | - Rachael Slater
- University of Liverpool Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Stephen Moss
- Cambridge University Hospitals NHS Foundation Trust, Department of Gastroenterology, Cambridge, UK; University of Cambridge Department of Medicine, Gastroenterology and Hepatology, Cambridge, Cambridgeshire, UK
| | - David Colin Bulmer
- University of Cambridge Department of Pharmacology, Cambridge, Cambridgeshire, UK
| | - Juan de la Revilla Negro
- Cambridge University Hospitals NHS Foundation Trust, Department of Gastroenterology, Cambridge, UK
| | - Umer Zeeshan Ijaz
- University of Glasgow, Mazumdar-Shaw Advanced Research Centre, Glasgow, UK
| | - David Mark Pritchard
- University of Liverpool Institute of Systems, Molecular and Integrative Biology, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Department of Gastroenterology, Liverpool, UK
| | - Miles Parkes
- Cambridge University Hospitals NHS Foundation Trust, Department of Gastroenterology, Cambridge, UK; University of Cambridge Department of Medicine, Gastroenterology and Hepatology, Cambridge, Cambridgeshire, UK
| | - Chris Probert
- University of Liverpool Institute of Systems, Molecular and Integrative Biology, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Department of Gastroenterology, Liverpool, UK
| |
Collapse
|
20
|
Crucillà S, Caldart F, Michelon M, Marasco G, Costantino A. Functional Abdominal Bloating and Gut Microbiota: An Update. Microorganisms 2024; 12:1669. [PMID: 39203511 PMCID: PMC11357468 DOI: 10.3390/microorganisms12081669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
(1) Background: Functional abdominal bloating and distension (FAB/FAD) are common disorders of the gut-brain interaction. Their physiopathology is complex and not completely clarified, although gut microbiota imbalances play a central role. The treatment of FAB/FAD still represents a clinical challenge for both patients and healthcare providers. Gut microbiota modulation strategies might play a crucial role in their management. The aim of this narrative review was to update the current evidence on FAB/FAD, with a focus on gut microbiota. (2) Methods: In October 2023, a review was conducted through the Medline, PubMed, and Embase databases. Selected literature included all available English-edited studies (randomized controlled trials and cross-sectional, cohort, and case-control studies). (3) Results: Twelve studies were selected, most of which investigated the relationship between IBS and microbiota, with bloating being one of its symptoms. The studies suggest that restoring a balanced microbiome appears to be the most promising solution for better management of FAB/FAD. Targeted approaches, such as the use of probiotics, prebiotics, antibiotics such as rifaximin or dietary modifications, may hold the key to alleviating symptoms. Other therapeutic options, such as diet, neuromodulators, and brain-gut behavioral therapies (i.e., cognitive-behavioral therapy) have shown promising outcomes, but strong data are still lacking. (4) Conclusions: Targeted approaches that focus on the gut microbiota, such as the use of probiotics, prebiotics, and antibiotics, are essential in managing FAB/FAD. Understanding the complex relationship between gut microbiota and FAB/FAD is crucial for developing effective treatments. Further studies are needed to explore the specific roles of different microbial populations in patients with FAB/FAD to enhance therapeutic strategies.
Collapse
Affiliation(s)
- Salvatore Crucillà
- Gastroenterology B Unit, Pancreas Center, University of Verona, 37134 Verona, Italy;
| | - Federico Caldart
- Gastroenterology B Unit, Pancreas Center, University of Verona, 37134 Verona, Italy;
| | - Marco Michelon
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
| | - Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy;
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Andrea Costantino
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
- Unit of Gastroenterology and Endoscopy, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
21
|
Dean G, Chey SW, Singh P, Chey WD. A diet low in fermentable oligo-, di-, monosaccharides and polyols improves abdominal and overall symptoms in persons with all subtypes of irritable bowel syndrome. Neurogastroenterol Motil 2024; 36:e14845. [PMID: 38887150 DOI: 10.1111/nmo.14845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/02/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND A diet low in fermentable oligo-, di-, monosaccharides and polyols (LFD) improves symptoms in patients with irritable bowel syndrome (IBS). Previous studies have focused on patients with IBS and diarrhea (IBS-D). It is unclear whether LFD is effective for IBS with constipation (IBS-C) or IBS with mixed bowel habits (IBS-M). This open-label, real-world study evaluates the relative effectiveness of the LFD among IBS subtypes. METHODS This study analyzes data from a service that provides low-FODMAP meals to individuals with IBS. Participants met with a registered dietitian and completed the IBS symptom severity survey (IBS-SSS) before and after undergoing a 2-4-week period of FODMAP restriction. The primary endpoint was the proportion of participants with ≥50-point decrease in IBS-SSS between the three IBS subtypes. KEY RESULTS After FODMAP restriction, 90% of participants with IBS-D, 75% with IBS-C, and 84% with IBS-M met the primary endpoint (p = 0.045). Similar improvement was seen for a 100-point decrease, but the difference between IBS subtypes was not significant (p = 0.46). After FODMAP restriction, all groups had statistically significant improvement in total IBS-SSS as well as individual symptom categories. Improvement in IBS-SSS subcategories was similar among the groups except for the categories of bloating severity (IBS-M had greatest improvement) and bowel movement satisfaction (IBS-C had less improvement). CONCLUSION & INFERENCES Though the proportion of responders was highest for IBS-D and lowest for IBS-C, the LFD led to robust improvement in overall symptoms in all IBS subtypes. Key individual symptoms also showed significant improvements in all IBS subtypes.
Collapse
Affiliation(s)
- Gregory Dean
- Department of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Samuel W Chey
- Department of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Prashant Singh
- Department of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - William D Chey
- Department of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
22
|
Lange S, Tsohataridis S, Boland N, Ngo L, Hahad O, Münzel T, Wild P, Daiber A, Schuppan D, Lurz P, Keppeler K, Steven S. Effects of Short-Term Gluten-Free Diet on Cardiovascular Biomarkers and Quality of Life in Healthy Individuals: A Prospective Interventional Study. Nutrients 2024; 16:2265. [PMID: 39064708 PMCID: PMC11279490 DOI: 10.3390/nu16142265] [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: 06/23/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
INTRODUCTION The exposome concept includes nutrition as it significantly influences human health, impacting the onset and progression of diseases. Gluten-containing wheat products are an essential source of energy for the world's population. However, a rising number of non-celiac healthy individuals tend to reduce or completely avoid gluten-containing cereals for health reasons. AIM AND METHODS This prospective interventional human study aimed to investigate whether short-term gluten avoidance improves cardiovascular endpoints and quality of life (QoL) in healthy volunteers. A cohort of 27 participants followed a strict gluten-free diet (GFD) for four weeks. Endothelial function measured by flow-mediated vasodilation (FMD), blood testing, plasma proteomics (Olink®) and QoL as measured by the World Health Organisation Quality-of-Life (WHOQOL) survey were investigated. RESULTS GFD resulted in decreased leucocyte count and C-reactive protein levels along with a trend of reduced inflammation biomarkers determined by plasma proteomics. A positive trend indicated improvement in FMD, whereas other cardiovascular endpoints remained unchanged. In addition, no improvement in QoL was observed. CONCLUSION In healthy individuals, a short-term GFD demonstrated anti-inflammatory effects but did not result in overall cardiovascular improvement or enhanced quality of life.
Collapse
Affiliation(s)
- Simon Lange
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Simeon Tsohataridis
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Niklas Boland
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Lisa Ngo
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Omar Hahad
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Philipp Wild
- Department of Preventive Cardiology and Medical Prevention, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Andreas Daiber
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Philipp Lurz
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Karin Keppeler
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
| | - Sebastian Steven
- Department of Cardiology, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany (S.T.); (O.H.); (T.M.); (A.D.); (P.L.)
- Department of Cardiology, Goethe University Frankfurt, 60596 Main, Germany
| |
Collapse
|
23
|
Qadri H, Shah AH, Almilaibary A, Mir MA. Microbiota, natural products, and human health: exploring interactions for therapeutic insights. Front Cell Infect Microbiol 2024; 14:1371312. [PMID: 39035357 PMCID: PMC11257994 DOI: 10.3389/fcimb.2024.1371312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/03/2024] [Indexed: 07/23/2024] Open
Abstract
The symbiotic relationship between the human digestive system and its intricate microbiota is a captivating field of study that continues to unfold. Comprising predominantly anaerobic bacteria, this complex microbial ecosystem, teeming with trillions of organisms, plays a crucial role in various physiological processes. Beyond its primary function in breaking down indigestible dietary components, this microbial community significantly influences immune system modulation, central nervous system function, and disease prevention. Despite the strides made in microbiome research, the precise mechanisms underlying how bacterial effector functions impact mammalian and microbiome physiology remain elusive. Unlike the traditional DNA-RNA-protein paradigm, bacteria often communicate through small molecules, underscoring the imperative to identify compounds produced by human-associated bacteria. The gut microbiome emerges as a linchpin in the transformation of natural products, generating metabolites with distinct physiological functions. Unraveling these microbial transformations holds the key to understanding the pharmacological activities and metabolic mechanisms of natural products. Notably, the potential to leverage gut microorganisms for large-scale synthesis of bioactive compounds remains an underexplored frontier with promising implications. This review serves as a synthesis of current knowledge, shedding light on the dynamic interplay between natural products, bacteria, and human health. In doing so, it contributes to our evolving comprehension of microbiome dynamics, opening avenues for innovative applications in medicine and therapeutics. As we delve deeper into this intricate web of interactions, the prospect of harnessing the power of the gut microbiome for transformative medical interventions becomes increasingly tantalizing.
Collapse
Affiliation(s)
- Hafsa Qadri
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Abdul Haseeb Shah
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Abdullah Almilaibary
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
- Department of Family and Community Medicine, Faculty of Medicine, Al Baha University, Al Bahah, Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| |
Collapse
|
24
|
Chu NHS, Chow E, Chan JCN. The Therapeutic Potential of the Specific Intestinal Microbiome (SIM) Diet on Metabolic Diseases. BIOLOGY 2024; 13:498. [PMID: 39056692 PMCID: PMC11273990 DOI: 10.3390/biology13070498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024]
Abstract
Exploring the intricate crosstalk between dietary prebiotics and the specific intestinal microbiome (SIM) is intriguing in explaining the mechanisms of current successful dietary interventions, including the Mediterranean diet and high-fiber diet. This knowledge forms a robust basis for developing a new natural food therapy. The SIM diet can be measured and evaluated to establish a reliable basis for the management of metabolic diseases, such as diabetes, metabolic (dysfunction)-associated fatty liver disease (MAFLD), obesity, and metabolic cardiovascular disease. This review aims to delve into the existing body of research to shed light on the promising developments of possible dietary prebiotics in this field and explore the implications for clinical practice. The exciting part is the crosstalk of diet, microbiota, and gut-organ interactions facilitated by producing short-chain fatty acids, bile acids, and subsequent metabolite production. These metabolic-related microorganisms include Butyricicoccus, Akkermansia, and Phascolarctobacterium. The SIM diet, rather than supplementation, holds the promise of significant health consequences via the prolonged reaction with the gut microbiome. Most importantly, the literature consistently reports no adverse effects, providing a strong foundation for the safety of this dietary therapy.
Collapse
Affiliation(s)
- Natural H. S. Chu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
| | - Juliana C. N. Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China; (E.C.); (J.C.N.C.)
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
25
|
Van den Houte K, Colomier E, Routhiaux K, Mariën Z, Schol J, Van den Bergh J, Vanderstappen J, Pauwels N, Joos A, Arts J, Caenepeel P, De Clerck F, Matthys C, Meulemans A, Jones M, Vanuytsel T, Carbone F, Tack J. Efficacy and Findings of a Blinded Randomized Reintroduction Phase for the Low FODMAP Diet in Irritable Bowel Syndrome. Gastroenterology 2024; 167:333-342. [PMID: 38401741 DOI: 10.1053/j.gastro.2024.02.008] [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: 03/01/2023] [Revised: 01/30/2024] [Accepted: 02/05/2024] [Indexed: 02/26/2024]
Abstract
BACKGROUND & AIMS The efficacy of a low fermentable oligo-, di-, monosaccharides and polyols (FODMAP) diet in irritable bowel syndrome (IBS) is well established. After the elimination period, a reintroduction phase aims to identify triggers. We studied the impact of a blinded reintroduction using FODMAP powders to objectively identify triggers and evaluated the effect on symptoms, quality of life, and psychosocial comorbidities. METHODS Responders to a 6-week low FODMAP diet, defined by a drop in IBS symptom severity score (IBS-SSS) compared with baseline, entered a 9-week blinded randomized reintroduction phase with 6 FODMAP powders (fructans, fructose, galacto-oligosaccharides, lactose, mannitol, sorbitol) or control (glucose). A rise in IBS-SSS (≥50 points) defined a FODMAP trigger. Patients completed daily symptom diaries and questionnaires for quality of life and psychosocial comorbidities. RESULTS In 117 recruited patients with IBS, IBS-SSS improved significantly after the elimination period compared with baseline (150 ± 116 vs 301 ± 97, P < .0001, 80% responders). Symptom recurrence was triggered in 85% of the FODMAP powders, by an average of 2.5 ± 2 FODMAPs/patient. The most prevalent triggers were fructans (56%) and mannitol (54%), followed by galacto-oligosaccharides, lactose, fructose, sorbitol, and glucose (respectively 35%, 28%, 27%, 23%, and 26%) with a significant increase in abdominal pain at day 1 for sorbitol/mannitol, day 2 for fructans/galacto-oligosaccharides, and day 3 for lactose. CONCLUSION We confirmed the significant benefit of the low FODMAP diet in tertiary-care IBS. A blinded reintroduction revealed a personalized pattern of symptom recurrence, with fructans and mannitol as the most prevalent, and allows the most objective identification of individual FODMAP triggers. Ethical commission University hospital of Leuven reference number: s63629; Clinicaltrials.gov number: NCT04373304.
Collapse
Affiliation(s)
- Karen Van den Houte
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Esther Colomier
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Karen Routhiaux
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Zoë Mariën
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Jolien Schol
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | | | | | - Nelle Pauwels
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Annick Joos
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Joris Arts
- Department of Gastroenterology, Algemeen Ziekenhuis Sint-Lucas, Brugge, Belgium
| | - Philip Caenepeel
- Department of Gastroenterology, Hospital Oost-Limburg, Genk, Belgium
| | | | - Christophe Matthys
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism KU Leuven, Leuven, Belgium
| | - Ann Meulemans
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism KU Leuven, Leuven, Belgium
| | - Mike Jones
- School of Psychological Sciences, Faculty of Medicine, Health & Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium; Department of Gastroenterology, University Hospitals Leuven (UZ Leuven), Belgium
| | - Florencia Carbone
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium; Department of Gastroenterology, University Hospitals Leuven (UZ Leuven), Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Diseases (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium; Department of Gastroenterology, University Hospitals Leuven (UZ Leuven), Belgium.
| |
Collapse
|
26
|
Vakili O, Adibi Sedeh P, Pourfarzam M. Metabolic biomarkers in irritable bowel syndrome diagnosis. Clin Chim Acta 2024; 560:119753. [PMID: 38821336 DOI: 10.1016/j.cca.2024.119753] [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: 04/14/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Irritable bowel syndrome (IBS) is a chronic gastrointestinal (GI) disorder characterized by altered bowel habits and abdominal discomfort during defecation. It significantly impacts life quality and work productivity for those affected. Global data suggests a slightly higher prevalence in females than in males. Today, unambiguous diagnosis of IBS remains challenging due to the absence of a specific biochemical, histopathological, or radiological test. Current diagnosis relies heavily on thorough symptom evaluation. Efforts by the Rome committees have established standardized diagnostic criteria (Rome I-IV), improving consistency and clinical applicability. Recent studies in this framework, seem to have successfully employed metabolomics techniques to identify distinct metabolite profiles in breath and stool samples of IBS patients, differentiating them from healthy controls and those with other functional GI disorders, such as inflammatory bowel disease (IBD). Building on this success, researchers are investigating the presence of similar metabolites in easily accessible biofluids such as urine, potentially offering a less invasive diagnostic approach. Accordingly, this review focuses on key metabolites specifically detected in IBS patients' biological specimens, with a focus on urinary metabolites, using various methods, particularly mass spectrometry (MS)-based techniques, including gas chromatography-MS (GC-MS), liquid chromatography-tandem MS (LC-MS/MS), and capillary electrophoresis-MS (CE-MS) metabolomics assays. These findings may make provision for a new set of non-invasive biomarkers for IBS diagnosis and management.
Collapse
Affiliation(s)
- Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Peyman Adibi Sedeh
- Gastroenterology and Hepatology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Morteza Pourfarzam
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
27
|
Ford AC, Staudacher HM, Talley NJ. Postprandial symptoms in disorders of gut-brain interaction and their potential as a treatment target. Gut 2024; 73:1199-1211. [PMID: 38697774 DOI: 10.1136/gutjnl-2023-331833] [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: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/05/2024]
Abstract
Postprandial, or meal-related, symptoms, such as abdominal pain, early satiation, fullness or bloating, are often reported by patients with disorders of gut-brain interaction, including functional dyspepsia (FD) or irritable bowel syndrome (IBS). We propose that postprandial symptoms arise via a distinct pathophysiological process. A physiological or psychological insult, for example, acute enteric infection, leads to loss of tolerance to a previously tolerated oral food antigen. This enables interaction of both the microbiota and the food antigen itself with the immune system, causing a localised immunological response, with activation of eosinophils and mast cells, and release of inflammatory mediators, including histamine and cytokines. These have more widespread systemic effects, including triggering nociceptive nerves and altering mood. Dietary interventions, including a diet low in fermentable oligosaccharides, disaccharides, monosaccharides and polyols, elimination of potential food antigens or gluten, IgG food sensitivity diets or salicylate restriction may benefit some patients with IBS or FD. This could be because the restriction of these foods or dietary components modulates this pathophysiological process. Similarly, drugs including proton pump inhibitors, histamine-receptor antagonists, mast cell stabilisers or even tricyclic or tetracyclic antidepressants, which have anti-histaminergic actions, all of which are potential treatments for FD and IBS, act on one or more of these mechanisms. It seems unlikely that food antigens driving intestinal immune activation are the entire explanation for postprandial symptoms in FD and IBS. In others, fermentation of intestinal carbohydrates, with gas release altering reflex responses, adverse reactions to food chemicals, central mechanisms or nocebo effects may dominate. However, if the concept that postprandial symptoms arise from food antigens driving an immune response in the gastrointestinal tract in a subset of patients is correct, it is paradigm-shifting, because if the choice of treatment were based on one or more of these therapeutic targets, patient outcomes may be improved.
Collapse
Affiliation(s)
- Alexander C Ford
- Leeds Gastroenterology Institute, St James's University Hospital, Leeds, UK
| | - Heidi M Staudacher
- Deakin University-Geelong Waterfront Campus, Geelong, Victoria, Australia
| | - Nicholas J Talley
- Health, University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
28
|
Biniszewska O, Jacenik D, Tarasiuk A, Fichna J. Current and future pharmacotherapies for the management of constipation-predominant irritable bowel syndrome. Expert Opin Pharmacother 2024; 25:1039-1049. [PMID: 38856704 DOI: 10.1080/14656566.2024.2366993] [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: 02/16/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) is a common gastrointestinal disorder affecting 9-23% of the world's population, with a higher prevalence among women. IBS is a complex disorder influenced by psychosocial, physiological, and genetic factors, exacerbated by stress. AREAS COVERED Research confirms that the most common subtype of IBS is IBS-C. Therefore, new therapies are being developed to speed up bowel movement and reduce constipation, with drugs such as linaclotide, plecanatide, lubiprostone, or tegaserod available to reduce IBS-C symptoms. In addition, patients' condition is improved by foods rich in fiber and low in FODMAP and the use of biotics. EXPERT OPINION The topic is of great importance due to the growing number of patients suffering from IBS-C and its significant impact on quality of life. Current clinical trials of new therapeutic options are not too successful, and it seems that one of the plausible treatment options could be the multi-drug cocktail with some, or perhaps even all its ingredients emerging from drug re-purposing. Another important path that needs to be explored further in IBS-C patients is the adjustment of dietary habits and/or introduction of dietary or nutritional intervention.
Collapse
Affiliation(s)
- Olga Biniszewska
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Aleksandra Tarasiuk
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
29
|
McKay DM, Defaye M, Rajeev S, MacNaughton WK, Nasser Y, Sharkey KA. Neuroimmunophysiology of the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2024; 326:G712-G725. [PMID: 38626403 PMCID: PMC11376980 DOI: 10.1152/ajpgi.00075.2024] [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: 03/07/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/18/2024]
Abstract
Gut physiology is the epicenter of a web of internal communication systems (i.e., neural, immune, hormonal) mediated by cell-cell contacts, soluble factors, and external influences, such as the microbiome, diet, and the physical environment. Together these provide the signals that shape enteric homeostasis and, when they go awry, lead to disease. Faced with the seemingly paradoxical tasks of nutrient uptake (digestion) and retarding pathogen invasion (host defense), the gut integrates interactions between a variety of cells and signaling molecules to keep the host nourished and protected from pathogens. When the system fails, the outcome can be acute or chronic disease, often labeled as "idiopathic" in nature (e.g., irritable bowel syndrome, inflammatory bowel disease). Here we underscore the importance of a holistic approach to gut physiology, placing an emphasis on intercellular connectedness, using enteric neuroimmunophysiology as the paradigm. The goal of this opinion piece is to acknowledge the pace of change brought to our field via single-cell and -omic methodologies and other techniques such as cell lineage tracing, transgenic animal models, methods for culturing patient tissue, and advanced imaging. We identify gaps in the field and hope to inspire and challenge colleagues to take up the mantle and advance awareness of the subtleties, intricacies, and nuances of intestinal physiology in health and disease by defining communication pathways between gut resident cells, those recruited from the circulation, and "external" influences such as the central nervous system and the gut microbiota.
Collapse
Affiliation(s)
- Derek M McKay
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Manon Defaye
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sruthi Rajeev
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Wallace K MacNaughton
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Inflammation Research Network, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Keith A Sharkey
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Gastrointestinal Research Group, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [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: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
31
|
Khalighi Sikaroudi M, Soltani S, Ghoreishy SM, Ebrahimi Z, Shidfar F, Dehnad A. Effects of a low FODMAP diet on the symptom management of patients with irritable bowel syndrome: a systematic umbrella review with the meta-analysis of clinical trials. Food Funct 2024; 15:5195-5208. [PMID: 38711328 DOI: 10.1039/d3fo03717g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
A low FODMAP diet (LFD) is a common restrictive diet to manage the symptoms of irritable bowel syndrome (IBS). However, there is no consensus on the alleviating effects of this diet. Herein, a systematic umbrella review with meta-analysis was conducted to investigate the effect of an LFD on IBS symptoms and its secondary outcomes in patients, which were not reported in previous meta-analyses. We performed a systematic literature search in PubMed, Scopus, and ISI Web of Science up to December 2023. The methodological quality of systematic reviews and their included trials was evaluated using AMSTAR 2 and the Cochrane risk of bias, respectively. The certainty of the evidence tool was evaluated using the GRADE approach. The data related to IBS symptoms, quality of life (QoL), microbiome diversity, and stool short-chain fatty acids were extracted. A random-effect (if RCTs ≥ 6) or fixed-effect model (if RCTs < 5) was used to recalculate effect sizes and 95% CIs and report them in both qualitative and quantitative terms (pooled risk ratio, Hedges' g, and weighted mean difference). A total of 658 articles were initially identified, with 11 meta-analyses and 24 RCTs reporting 28 outcomes with 1646 participants included. An LFD significantly affected the clinical improvement of total symptoms according to the IBS-SSS questionnaire (RR: 1.42; 95% CI: 1.02, 1.97; P = 0.04) in all the subtypes of IBS and also had favorable effects on stool consistency (WMD: -0.48; 95% CI: -0.902, -0.07) and frequency (WMD: -0.36; 95% CI: -0.61, -0.10) and some other GI symptoms in both less and more than 4 weeks of diet intervention except for stool consistency, which needed more than 4 weeks of LFD implementation. A significant QoL improvement was observed but not in the anxiety and depression state. Furthermore, some studies showed that an LFD may increase fecal pH and dysbiosis and reduce SCFA and the abundance of Bifidobacterium. In conclusion, an LFD can alleviate symptoms and QoL in IBS patients, although dysbiosis may occur. Considering the low certainty of evidence, strong RCTs with more appropriate designs are needed.
Collapse
Affiliation(s)
- Masoumeh Khalighi Sikaroudi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Soltani
- Yazd Cardiovascular Research Center, Non-communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mojtaba Ghoreishy
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Zohreh Ebrahimi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Farzad Shidfar
- Nutritional Sciences Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran.
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Afsaneh Dehnad
- Department of English Language, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Mousavi Khaneghah A, Mostashari P. Decoding food reactions: a detailed exploration of food allergies vs. intolerances and sensitivities. Crit Rev Food Sci Nutr 2024:1-45. [DOI: 10.1080/10408398.2024.2349740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
| | - Parisa Mostashari
- Department of Food Science and Technology, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
33
|
Xu C, Song Z, Hu JY, Li CC, Shen H. Global research trend and hotspot in the low FODMAP diet: a bibliometric analysis. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2024; 43:63. [PMID: 38741213 PMCID: PMC11092139 DOI: 10.1186/s41043-024-00567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND According to national guidelines, a diet low in fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAP) is a second-line therapy option for irritable bowel syndrome (IBS) and improves functional intestinal symptoms. Numerous noteworthy results have been published in this field over the past fifteen years. This study aims to analyze the global research trend and hotspot of the low FODMAP diet research, and provide a comprehensive perspective and direction for researchers. METHODS The Science Citation Index-Expanded of the Web of Science Core Collection (WoSCC) was used to identify low FODMAP diet-related articles and reviews. Three bibliometric programs (CiteSpace, VOSviewer, Scimago Graphic) were utilized to analyze and visualize the annual publications, authors, countries, institutions, journals, citations, and keywords. RESULTS In total, 843 documents related to the low FODMAP diet research were published in 227 journals by 3,343 authors in 1,233 institutions from 59 countries. The United States, which was the most engaged nation in international collaboration, had the largest annual production and the fastest growth. The most productive organization was Monash University, and the most fruitful researcher was Gibson PR. Nutrients ranked first in terms of the number of published documents. The article "A diet low in FODMAPs reduces symptoms of irritable bowel syndrome" (Halmos EP, 2014) received the most co-citations. Keywords that appear frequently in the literature mainly involve two main aspects: the clinical efficacy evaluation and mechanism exploration of the low FODMAP diet. The term "gut microbiota" stands out as the most prominent keyword among the burst keywords that have remained prevalent till date. CONCLUSION The restriction stage of the low FODMAP diet is superior to other dietary therapies for IBS in terms of symptom response, but it has a negative impact on the abundance of gut Bifidobacteria and diet quality. Identification of biomarkers to predict response to the low FODMAP diet is of great interest and has become the current research hotspot.
Collapse
Affiliation(s)
- Cheng Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen Song
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Yi Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chong-Chao Li
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
34
|
Conley TE, Parkes M, Moss S, Probert C. Assessing 'response' to the low-FODMAP diet in irritable bowel syndrome: Should we be reporting harder primary endpoints? Clin Nutr 2024; 43:1079-1086. [PMID: 38579370 DOI: 10.1016/j.clnu.2024.03.017] [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: 01/08/2024] [Revised: 03/04/2024] [Accepted: 03/22/2024] [Indexed: 04/07/2024]
Abstract
BACKGROUND & AIMS The low-FODMAP diet (LFD) has become almost synonymous with IBS care, yet the challenges associated with this rigorous therapeutic approach are often underacknowledged. Despite positive outcomes in RCTs, comparator groups frequently exhibit substantial response rates, raising questions about the definition of 'response'. Whilst the assessment of response in drug trials has evolved to utilize the more stringent FDA/EMA primary clinical endpoints, trials of the LFD have not yet followed. The aim of this article is to opine whether the current approach to the measurement of clinical response to the LFD in clinical trials should be reconsidered. METHODS A comprehensive literature review of LFD clinical trials from the past decade was conducted, focusing on recorded response metrics for primary clinical endpoints. RESULTS While response definitions vary, the 50-point IBS-SSS delta emerged as the predominant metric. Notably, no trials to date have adopted the more stringent primary clinical endpoints used in drug trials. Other response measures included binary response metrics (such as 'adequate clinical response'), changes in visual analogue scales or stool form/output, reductions in abdominal pain, as well as changes the magnitude of the IBS-SSS delta. Whether these metrics correspond to a clinically meaningful improvement for the patient is less clear, and as such aligning patient-clinician expectations can be challenging. CONCLUSIONS A paradigm shift in the conceptualization of 'response' coupled with an emphasis on harder clinical endpoints in the context of clinical trials may serve to better justify the trade-off between symptom-improvement and the inherent challenges associated with this burdensome therapeutic approach.
Collapse
Affiliation(s)
- Thomas Edward Conley
- University of Liverpool Institute of Integrative Biology, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Department of Gastroenterology, Liverpool, UK.
| | - Miles Parkes
- University of Cambridge Department of Medicine, Gastroenterology and Hepatology, Cambridge, Cambridgeshire, UK; Cambridge University Hospitals NHS Foundation Trust, Department of Gastroenterology, Cambridge, UK
| | - Stephen Moss
- University of Cambridge Department of Medicine, Gastroenterology and Hepatology, Cambridge, Cambridgeshire, UK; Cambridge University Hospitals NHS Foundation Trust, Department of Gastroenterology, Cambridge, UK
| | - Chris Probert
- University of Liverpool Institute of Integrative Biology, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Department of Gastroenterology, Liverpool, UK
| |
Collapse
|
35
|
Wu H, Li J, Li F, Lun W. Causal association of gastroesophageal reflux disease on irritable bowel syndrome: a two-sample Mendelian randomization study. Front Genet 2024; 15:1328327. [PMID: 38601073 PMCID: PMC11004226 DOI: 10.3389/fgene.2024.1328327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Background Recently, observational studies have reported that gastroesophageal reflux disease (GERD) is commonly associated with irritable bowel syndrome (IBS), but the causal relationship is unclear. Methods We conducted a two-sample Mendelian randomization study using summary data from genome-wide association studies (GWASs) to explore a causal relationship between GERD (N cases = 129,080) and IBS (N cases = 4,605) of European ancestry. Furthermore, the inverse-variance weighted (IVW) method and a series of sensitivity analyses were used to assess the accuracy and confidence of our results. Results We found a significant association of GERD with IBS (NSNP = 74; OR: 1.375; 95% CI: 1.164-1.624; p < 0.001). Reverse MR analysis showed no evidence of a causal association for IBS with GERD (NSNP = 6; OR: 0.996; 95% CI: 0.960-1.034; p = 0.845). Conclusion This study provides evidence that the presence of GERD increases the risk of developing IBS, and it is observed from the reverse MR results that IBS did not increase the risk of GERD.
Collapse
Affiliation(s)
- Huihuan Wu
- Department of Gastroenterology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - FeiFei Li
- Department of Cardiology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Weijian Lun
- Department of Gastroenterology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, Guangdong, China
| |
Collapse
|
36
|
Ashonibare VJ, Akorede BA, Ashonibare PJ, Akhigbe TM, Akhigbe RE. Gut microbiota-gonadal axis: the impact of gut microbiota on reproductive functions. Front Immunol 2024; 15:1346035. [PMID: 38482009 PMCID: PMC10933031 DOI: 10.3389/fimmu.2024.1346035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/30/2024] [Indexed: 04/12/2024] Open
Abstract
The influence of gut microbiota on physiological processes is rapidly gaining attention globally. Despite being under-studied, there are available data demonstrating a gut microbiota-gonadal cross-talk, and the importance of this axis in reproduction. This study reviews the impacts of gut microbiota on reproduction. In addition, the possible mechanisms by which gut microbiota modulates male and female reproduction are presented. Databases, including Embase, Google scholar, Pubmed/Medline, Scopus, and Web of Science, were explored using relevant key words. Findings showed that gut microbiota promotes gonadal functions by modulating the circulating levels of steroid sex hormones, insulin sensitivity, immune system, and gonadal microbiota. Gut microbiota also alters ROS generation and the activation of cytokine accumulation. In conclusion, available data demonstrate the existence of a gut microbiota-gonadal axis, and role of this axis on gonadal functions. However, majority of the data were compelling evidences from animal studies with a great dearth of human data. Therefore, human studies validating the reports of experimental studies using animal models are important.
Collapse
Affiliation(s)
- Victory J. Ashonibare
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Bolaji A. Akorede
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Biomedical Sciences, University of Wyoming, Laramie, WY, United States
| | - Precious J. Ashonibare
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Tunmise M. Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Breeding and Genetic Unit, Department of Agronomy, Osun State University, Ejigbo, Osun State, Nigeria
| | - Roland Eghoghosoa Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
37
|
Bertin L, Zanconato M, Crepaldi M, Marasco G, Cremon C, Barbara G, Barberio B, Zingone F, Savarino EV. The Role of the FODMAP Diet in IBS. Nutrients 2024; 16:370. [PMID: 38337655 PMCID: PMC10857121 DOI: 10.3390/nu16030370] [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: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
The low FODMAP (fermentable oligosaccharide, disaccharide, monosaccharide, and polyol) diet is a beneficial therapeutic approach for patients with irritable bowel syndrome (IBS). However, how the low FODMAP diet works is still not completely understood. These mechanisms encompass not only traditionally known factors such as luminal distension induced by gas and water but also recent evidence on the role of FOMAPs in the modulation of visceral hypersensitivity, increases in intestinal permeability, the induction of microbiota changes, and the production of short-chain fatty acids (SCFAs), as well as metabolomics and alterations in motility. Although most of the supporting evidence is of low quality, recent trials have confirmed its effectiveness, even though the majority of the evidence pertains only to the restriction phase and its effectiveness in relieving abdominal bloating and pain. This review examines potential pathophysiological mechanisms and provides an overview of the existing evidence on the effectiveness of the low FODMAP diet across various IBS subtypes. Key considerations for its use include the challenges and disadvantages associated with its practical implementation, including the need for professional guidance, variations in individual responses, concerns related to microbiota, nutritional deficiencies, the development of constipation, the necessity of excluding an eating disorder before commencing the diet, and the scarcity of long-term data. Despite its recognized efficacy in symptom management, acknowledging these limitations becomes imperative for a nuanced comprehension of the role of a low FODMAP diet in managing IBS. By investigating its potential mechanisms and evidence across IBS subtypes and addressing emerging modulations alongside limitations, this review aims to serve as a valuable resource for healthcare practitioners, researchers, and patients navigating the intricate landscape of IBS.
Collapse
Affiliation(s)
- Luisa Bertin
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| | - Miriana Zanconato
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| | - Martina Crepaldi
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (C.C.); (G.B.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Cesare Cremon
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (C.C.); (G.B.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (G.M.); (C.C.); (G.B.)
- Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy
| | - Brigida Barberio
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| | - Fabiana Zingone
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| | - Edoardo Vincenzo Savarino
- Department of Surgery, Oncology, Gastroenterology, University of Padua, 35121 Padua, Italy; (L.B.); (M.Z.); (M.C.); (B.B.); (F.Z.)
- Gastroenterology Unit, Azienda Ospedale-Università Padova, 35128 Padua, Italy
| |
Collapse
|
38
|
Zhou J, Wei H, Zhou A, Xiao X, Xie X, Tang B, Lin H, Tang L, Meng R, Yuan X, Zhang J, Huang C, Huang B, Liao X, Zhong T, He S, Gu S, Yang S. The gut microbiota participates in the effect of linaclotide in patients with irritable bowel syndrome with constipation (IBS-C): a multicenter, prospective, pre-post study. J Transl Med 2024; 22:98. [PMID: 38263117 PMCID: PMC10807057 DOI: 10.1186/s12967-024-04898-1] [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: 10/27/2023] [Accepted: 01/14/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND Interindividual variation characterizes the relief experienced by constipation-predominant irritable bowel syndrome (IBS-C) patients following linaclotide treatment. Complex bidirectional interactions occur between the gut microbiota and various clinical drugs. To date, no established evidence has elucidated the interactions between the gut microbiota and linaclotide. We aimed to explore the impact of linaclotide on the gut microbiota and identify critical bacterial genera that might participate in linaclotide efficacy. METHODS IBS-C patients were administered a daily linaclotide dose of 290 µg over six weeks, and their symptoms were then recorded during a four-week posttreatment observational period. Pre- and posttreatment fecal samples were collected for 16S rRNA sequencing to assess alterations in the gut microbiota composition. Additionally, targeted metabolomics analysis was performed for the measurement of short-chain fatty acid (SCFA) concentrations. RESULTS Approximately 43.3% of patients met the FDA responder endpoint after taking linaclotide for 6 weeks, and 85% of patients reported some relief from abdominal pain and constipation. Linaclotide considerably modified the gut microbiome and SCFA metabolism. Notably, the higher efficacy of linaclotide was associated with enrichment of the Blautia genus, and the abundance of Blautia after linaclotide treatment was higher than that in healthy volunteers. Intriguingly, a positive correlation was found for the Blautia abundance and SCFA concentrations with improvements in clinical symptoms among IBS-C patients. CONCLUSION The gut microbiota, especially the genus Blautia, may serve as a significant predictive microbe for symptom relief in IBS-C patients receiving linaclotide treatment. TRIAL REGISTRATION This trial was registered with the Chinese Clinical Trial Registry (Chictr.org.cn, ChiCTR1900027934).
Collapse
Affiliation(s)
- Jianyun Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Haoqi Wei
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - An Zhou
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Xu Xiao
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Xia Xie
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Bo Tang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Hui Lin
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Li Tang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Ruiping Meng
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Xiaoying Yuan
- Department of Gastroenterology, The Ninth People's Hospital of Chongqing, No. 69 Jialing Village, Beibei District, Chongqing, China, 400700
| | - Jing Zhang
- Department of Gastroenterology, Chongqing University Jiangjin Hospital, No.725, Jiangzhou Avenue, Jiangjin District, Chongqing, China, 402260
| | - Cheng Huang
- Department of Gastroenterology, Chonggang General Hospital, No. 1 Dayan Sancun, Dadukou District, Chongqing, China, 400000
| | - Baobao Huang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Xiping Liao
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Tingting Zhong
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037
| | - Suyu He
- Department of Gastroenterology, Suining Central Hospital, 22 Youfang Street, Chuanshan District, Suining, China, 629000.
| | - Sai Gu
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University Jinshan Campus, 50 Jinyu Dadao, Liangjiang New District, Chongqing, China, 401112.
| | - Shiming Yang
- Department of Gastroenterology, The Second Affiliated Hospital, The Third Military Medical University, Xinqiaozheng Street, Chongqing, China, 400037.
| |
Collapse
|
39
|
O’Neill S, Minehan M, Knight-Agarwal CR, Pyne DB. Alterations in gut microbiota caused by major depressive disorder or a low FODMAP diet and where they overlap. Front Nutr 2024; 10:1303405. [PMID: 38260072 PMCID: PMC10800578 DOI: 10.3389/fnut.2023.1303405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Beneficial changes in microbiota observed in individuals with a major depressive disorder (MDD) may be initiated with a low fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) elimination diet. Academic Search Ultimate, APA PsychINFO, Cochrane Library, MEDLINE, Scopus and Web of Science were searched for original research documenting differences in microbiota in MDD or changes with a low FODMAP diet in adults (age 18 years +). Studies with fecal microbiota, 16 s RNA sequencing and QIIME pipelines were included. Studies using antibiotics, probiotics, and medications such as antidepressants were excluded. Additionally, studies based on a single gender were excluded as gender impacts microbiota changes in MDD. Four studies addressed differences in microbiota with MDD and another four assessed shifts occurring with a low FODMAP diet. The abundance of Bacteroidetes, Bacteroidaceae and Bacteroides were lower in individuals with MDD but increased with a low FODMAP diet. Abundance of Ruminoccaceae was lower and Bilophila was higher with both a low FODMAP diet and MDD. These results provide preliminary evidence that a low FODMAP diet might drive changes in microbiota that also benefit people with MDD. Further research to assess whether a low FODMAP diet can treat MDD through modification of targeted microbiota is warranted.
Collapse
Affiliation(s)
- Simone O’Neill
- University of Canberra Research Institute for Sport and Exercise, Faculty of Health, University of Canberra, Canberra, ACT, Australia
| | - Michelle Minehan
- University of Canberra Research Institute for Sport and Exercise, Faculty of Health, University of Canberra, Canberra, ACT, Australia
- Faculty of Health, University of Canberra, Canberra, ACT, Australia
| | | | - David B. Pyne
- University of Canberra Research Institute for Sport and Exercise, Faculty of Health, University of Canberra, Canberra, ACT, Australia
| |
Collapse
|
40
|
Scarpellini E, Balsiger LM, Broeders B, Houte KVD, Routhiaux K, Raymenants K, Carbone F, Tack J. Nutrition and Disorders of Gut-Brain Interaction. Nutrients 2024; 16:176. [PMID: 38202005 PMCID: PMC10780945 DOI: 10.3390/nu16010176] [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: 11/13/2023] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Disorders of gut-brain interaction (DGBIs) have a complex pathophysiology that is often characterized by a relationship between food ingestion and triggering of symptoms. Understanding of the underlying mechanisms and the role of nutrients as a therapeutic target are rapidly evolving. AIMS AND METHODS We performed a narrative review of the literature using the following keywords, their acronyms, and their associations: nutrients, disorders of gut-brain interaction; functional dyspepsia; malabsorption; irritable bowel syndrome; diarrhea; constipation. RESULTS Functional dyspepsia displayed a significant correlation between volume, fat and/or wheat abundance, chemical composition of ingested food and symptoms of early satiety, fullness and weight loss. Carbohydrate malabsorption is related to enzyme deficiency throughout the GI tract. Food composition and richness in soluble vs. non-soluble fibers is related to constipation and diarrhea. The elimination of fermentable oligo-, di-, monosaccharides and polyols (FODMAPs) has a significant and non-unidirectional impact on irritable bowel syndrome (IBS) symptoms. CONCLUSIONS Food volume, nutritive and chemical composition, and its malabsorption are associated with symptom generation in DGBIs. Further multicenter, randomized-controlled clinical trials are needed to clarify the underlying pathophysiology.
Collapse
Affiliation(s)
- Emidio Scarpellini
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
- Internal Medicine Unit, “Madonna del Soccorso” General Hospital, Via Luciano Manara 7, 63074 San Benedetto del Tronto, Italy
| | - Lukas Michaja Balsiger
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Bert Broeders
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Karen Van Den Houte
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Karen Routhiaux
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Karlien Raymenants
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Florencia Carbone
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| | - Jan Tack
- Translational Research in Gastrointestinal Disoerders (T.A.R.G.I.D.), Gasthuisberg University Hospital, KU Leuven, Herestraat 49, 3000 Lueven, Belgium; (E.S.); (L.M.B.); (B.B.); (K.V.D.H.); (K.R.); (K.R.); (F.C.)
| |
Collapse
|
41
|
Tayama J, Hamaguchi T, Koizumi K, Yamamura R, Okubo R, Kawahara JI, Inoue K, Takeoka A, Fukudo S. Efficacy of an eHealth self-management program in reducing irritable bowel syndrome symptom severity: a randomized controlled trial. Sci Rep 2024; 14:4. [PMID: 38172498 PMCID: PMC10764726 DOI: 10.1038/s41598-023-50293-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
This study aimed to verify whether an eHealth-based self-management program can reduce irritable bowel syndrome (IBS) symptom severity. An open-label simple randomized controlled trial was conducted that compared an intervention group (n = 21) participating in an eHealth self-management program, which involved studying IBS-related information from an established self-help guide followed by in-built quizzes, with a treatment-as-usual group (n = 19) that, except for pharmacotherapy, had no treatment restrictions. Participants were female Japanese university students. The eHealth group received unlimited access to the self-management program for 8 weeks on computers and mobile devices. The primary outcome, participants' severity of IBS symptoms assessed using the IBS-severity index (IBS-SI), and the secondary outcomes of participants' quality of life, gut bacteria, and electroencephalography alpha and beta power percentages were measured at baseline and 8 weeks. A significant difference was found in the net change in IBS-SI scores between the eHealth and treatment-as-usual groups, and the former had significantly lower IBS-SI scores following the 8-week intervention than at baseline. Moreover, there was a significant difference in the net change in phylum Cyanobacteria between the eHealth and treatment-as-usual groups. Thus, the eHealth-based self-management program successfully reduced the severity of IBS symptoms.
Collapse
Affiliation(s)
- Jun Tayama
- Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama, 359-1192, Japan.
| | - Toyohiro Hamaguchi
- Department of Occupational Therapy, School of Health and Social Services, Saitama Prefectural University, 820, Sannomiya, Koshigaya, Saitama, 343-8540, Japan
| | - Kohei Koizumi
- Department of Occupational Therapy, School of Health and Social Services, Saitama Prefectural University, 820, Sannomiya, Koshigaya, Saitama, 343-8540, Japan
| | - Ryodai Yamamura
- Division of Biomedical Oncology, Institute for Genetic Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-0815, Japan
| | - Ryo Okubo
- Department of Psychiatry and Neurology, National Hospital Organization Obihiro Hospital, 16, Kita 2, Nishi 18, Obihiro, Hokkaido, 080-0048, Japan
| | - Jun-Ichiro Kawahara
- Department of Psychology, Hokkaido University, Kita 10, Nishi 7, Kita-Ku, Sapporo, Hokkaido, 060-0815, Japan
| | - Kenji Inoue
- Center for Student Success Research and Practice, Osaka University, 1-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsushi Takeoka
- Health Center, Nagasaki University, 1-14, Bunkyo, Nagasaki, Nagasaki, 852-8521, Japan
| | - Shin Fukudo
- Department of Behavioral Medicine, Tohoku University Graduate School of Medicine, 2-1, Seiryo-Machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
42
|
Kraimi N, Ross T, Pujo J, De Palma G. The gut microbiome in disorders of gut-brain interaction. Gut Microbes 2024; 16:2360233. [PMID: 38949979 PMCID: PMC11218806 DOI: 10.1080/19490976.2024.2360233] [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: 01/30/2024] [Accepted: 05/21/2024] [Indexed: 07/03/2024] Open
Abstract
Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.
Collapse
Affiliation(s)
- Narjis Kraimi
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Taylor Ross
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Julien Pujo
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| | - Giada De Palma
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Canada
| |
Collapse
|
43
|
Kazyulin A, Goncharenko A, Pavleeva E, Lyubozhnova I, Kalyagin I. Understanding similarities and differences of irritable bowel syndrome and conditions with IBS-like symptoms. RUSSIAN JOURNAL OF EVIDENCE-BASED GASTROENTEROLOGY 2024; 13:66. [DOI: 10.17116/dokgastro20241301166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
44
|
Herfindal AM, van Megen F, Gilde MKO, Valeur J, Rudi K, Skodje GI, Lundin KEA, Henriksen C, Bøhn SK. Effects of a low FODMAP diet on gut microbiota in individuals with treated coeliac disease having persistent gastrointestinal symptoms - a randomised controlled trial. Br J Nutr 2023; 130:2061-2075. [PMID: 37272479 PMCID: PMC10657752 DOI: 10.1017/s0007114523001253] [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: 11/22/2022] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
Individuals with coeliac disease (CeD) often experience gastrointestinal symptoms despite adherence to a gluten-free diet (GFD). While we recently showed that a diet low in fermentable oligo-, di-, monosaccharides and polyols (FODMAP) successfully provided symptom relief in GFD-treated CeD patients, there have been concerns that the low FODMAP diet (LFD) could adversely affect the gut microbiota. Our main objective was therefore to investigate whether the LFD affects the faecal microbiota and related variables of gut health. In a randomised controlled trial GFD-treated CeD adults, having persistent gastrointestinal symptoms, were randomised to either consume a combined LFD and GFD (n 39) for 4 weeks or continue with GFD (controls, n 36). Compared with the control group, the LFD group displayed greater changes in the overall faecal microbiota profile (16S rRNA gene sequencing) from baseline to follow-up (within-subject β-diversity, P < 0·001), characterised by lower and higher follow-up abundances (%) of genus Anaerostipes (Pgroup < 0·001) and class Erysipelotrichia (Pgroup = 0·02), respectively. Compared with the control group, the LFD led to lower follow-up concentrations of faecal propionic and valeric acid (GC-FID) in participants with high concentrations at baseline (Pinteraction ≤ 0·009). No differences were found in faecal bacterial α-diversity (Pgroup ≥ 0·20) or in faecal neutrophil gelatinase-associated lipocalin (ELISA), a biomarker of gut integrity and inflammation (Pgroup = 0·74), between the groups at follow-up. The modest effects of the LFD on the gut microbiota and related variables in the CeD patients of the present study are encouraging given the beneficial effects of the LFD strategy to treat functional GI symptoms (Registered at clinicaltrials.gov as NCT03678935).
Collapse
Affiliation(s)
- Anne Mari Herfindal
- Department of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Frida van Megen
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Unit for Clinical Nutrition, Division of Cancer Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Mari K. O. Gilde
- Department of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
| | - Knut Rudi
- Department of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Gry I. Skodje
- Healthy Life Centre, Municipality of Nes, Nes, Norway
| | - Knut E. A. Lundin
- KG Jebsen Coeliac Disease Research Centre, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Christine Henriksen
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Siv Kjølsrud Bøhn
- Department of Chemistry, Biotechnology and Food Sciences, Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
45
|
Marginean CM, Popescu M, Drocas AI, Cazacu SM, Mitrut R, Marginean IC, Iacob GA, Popescu MS, Docea AO, Mitrut P. Gut–Brain Axis, Microbiota and Probiotics—Current Knowledge on Their Role in Irritable Bowel Syndrome: A Review. GASTROINTESTINAL DISORDERS 2023; 5:517-535. [DOI: 10.3390/gidisord5040043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a common digestive disorder with a significant impact on both individuals and society in terms of quality of life and healthcare costs. A growing body of research has identified various communication pathways between the microbiota and the brain in relation to motility disorders, with the gut–brain axis being key to the pathogenesis of IBS. Multiple factors contribute to the pathogenetic pathways in IBS, including immune mechanisms, psychosocial factors, increased oxidative stress and pro-inflammatory cytokine release, as well as genetic and hormonal factors. Increased permeability of the normal intestinal barrier allows bacterial products to access the lamina propria, providing a mechanism for perpetuating chronic inflammation and characteristic symptoms. The microbiota influences inflammatory processes in IBS by altering the balance between pro-inflammatory factors and host defence. Probiotics modulate the pathophysiological mechanisms involved in IBS by influencing the composition of the microbiota and improving intestinal motility disorders, visceral hypersensitivity, immune function of the intestinal epithelium, metabolic processes in the intestinal lumen, dysfunction of the microbiota-GBA, and are recognised as effective and safe in IBS therapy. Our study aimed to provide a comprehensive overview of the relationship between the gut–brain axis, microbiota, and IBS, based on current information.
Collapse
Affiliation(s)
- Cristina Maria Marginean
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Popescu
- Department of Endocrinology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Andrei Ioan Drocas
- Department of Urology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Sergiu Marian Cazacu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Radu Mitrut
- Department of Cardiology, University and Emergency Hospital, 050098 Bucharest, Romania
| | | | - George Alexandru Iacob
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marian Sorin Popescu
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Paul Mitrut
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
46
|
Arora R, Chandel AK. Unlocking the potential of low FODMAPs sourdough technology for management of irritable bowel syndrome. Food Res Int 2023; 173:113425. [PMID: 37803764 DOI: 10.1016/j.foodres.2023.113425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 10/08/2023]
Abstract
Consumption of high FODMAP (Fermentable Oligo-, Di-, and Monosaccharides and Polyols) diet is the leading cause of alteration in the human gut microbiome, thereby, causing irritable bowel syndrome (IBS). Therefore, sourdough technology can be exploited for reduction of FODMAPs in various foods to alleviate the symptoms of IBS. Several microorganisms viz. Pichia fermentans, Lactobacillus fetmentum, Saccharomyces cerevisiae, Torulaspora delbrueckii, Kluyveromyces marxianus etc. have been identified for the production of low FODMAP type II sourdough fermented products. However, more research on regulation of end-product and volatilome profile is required for maximal exploitation of FODMAP-reducing microorganisms. Therefore, the present review is focused on utilisation of lactic acid bacteria and yeasts, alone and in synergy, for the production of low FODMAP sourdough foods. Moreover, the microbial bioprocessing of cereal and non-cereal based low FODMAP fermented sourdough products along with their nutritional and therapeutic benefits have been elaborated. The challenges and future prospects for the production of sourdough fermented low FODMAP foods, thereby, bringing out positive alterations in gut microbiome, have also been discussed.
Collapse
Affiliation(s)
- Richa Arora
- Department of Microbiology, Punjab Agricultural University, Ludhiana, Punjab 141004, India
| | - Anuj K Chandel
- Department of Biotechnology, Engineering School of Lorena (EEL), University of São Paulo, Lorena SP 12.602-810, Brazil.
| |
Collapse
|
47
|
Zheng H, Zhang C, Zhang J, Duan L. "Sentinel or accomplice": gut microbiota and microglia crosstalk in disorders of gut-brain interaction. Protein Cell 2023; 14:726-742. [PMID: 37074139 PMCID: PMC10599645 DOI: 10.1093/procel/pwad020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023] Open
Abstract
Abnormal brain-gut interaction is considered the core pathological mechanism behind the disorders of gut-brain interaction (DGBI), in which the intestinal microbiota plays an important role. Microglia are the "sentinels" of the central nervous system (CNS), which participate in tissue damage caused by traumatic brain injury, resist central infection and participate in neurogenesis, and are involved in the occurrence of various neurological diseases. With in-depth research on DGBI, we could find an interaction between the intestinal microbiota and microglia and that they are jointly involved in the occurrence of DGBI, especially in individuals with comorbidities of mental disorders, such as irritable bowel syndrome (IBS). This bidirectional regulation of microbiota and microglia provides a new direction for the treatment of DGBI. In this review, we focus on the role and underlying mechanism of the interaction between gut microbiota and microglia in DGBI, especially IBS, and the corresponding clinical application prospects and highlight its potential to treat DGBI in individuals with psychiatric comorbidities.
Collapse
Affiliation(s)
- Haonan Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Cunzheng Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Jindong Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| | - Liping Duan
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing 100191, China
| |
Collapse
|
48
|
Napolitano M, Fasulo E, Ungaro F, Massimino L, Sinagra E, Danese S, Mandarino FV. Gut Dysbiosis in Irritable Bowel Syndrome: A Narrative Review on Correlation with Disease Subtypes and Novel Therapeutic Implications. Microorganisms 2023; 11:2369. [PMID: 37894027 PMCID: PMC10609453 DOI: 10.3390/microorganisms11102369] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder characterized by chronic abdominal pain and altered bowel habits. It can be subclassified in different subtypes according to the main clinical manifestation: constipation, diarrhea, mixed, and unclassified. Over the past decade, the role of gut microbiota in IBS has garnered significant attention in the scientific community. Emerging research spotlights the intricate involvement of microbiota dysbiosis in IBS pathogenesis. Studies have demonstrated reduced microbial diversity and stability and specific microbial alterations for each disease subgroup. Microbiota-targeted treatments, such as antibiotics, probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and even diet, offer exciting prospects for managing IBS. However, definitive conclusions are hindered by the heterogeneity of these studies. Further research should focus on elucidating the mechanisms, developing microbiome-based diagnostics, and enabling personalized therapies tailored to an individual's microbiome profile. This review takes a deep dive into the microscopic world inhabiting our guts, and its implications for IBS. Our aim is to elucidate the complex interplay between gut microbiota and each IBS subtype, exploring novel microbiota-targeted treatments and providing a comprehensive overview of the current state of knowledge.
Collapse
Affiliation(s)
- Maria Napolitano
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Ernesto Fasulo
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| | - Federica Ungaro
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Luca Massimino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Division of Immunology, Transplantation and Infectious Disease, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Emanuele Sinagra
- Gastroenterology & Endoscopy Unit, Fondazione Istituto G. Giglio, Contrada Pietra Pollastra Pisciotto, 90015 Cefalù, Italy;
| | - Silvio Danese
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
- Gastroenterology and Endoscopy, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Francesco Vito Mandarino
- Department of Gastroenterology and Gastrointestinal Endoscopy, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.F.); (F.U.); (L.M.); (S.D.); (F.V.M.)
| |
Collapse
|
49
|
Nordin E, Hellström PM, Vuong E, Ribbenstedt A, Brunius C, Landberg R. IBS randomized study: FODMAPs alter bile acids, phenolic- and tryptophan metabolites, while gluten modifies lipids. Am J Physiol Regul Integr Comp Physiol 2023; 325:R248-R259. [PMID: 37399002 DOI: 10.1152/ajpregu.00016.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/10/2023] [Accepted: 06/17/2023] [Indexed: 07/04/2023]
Abstract
Diet is considered a culprit for symptoms in irritable bowel syndrome (IBS), although the mechanistic understanding of underlying causes is lacking. Metabolomics, i.e., the analysis of metabolites in biological samples may offer a diet-responsive fingerprint for IBS. Our aim was to explore alterations in the plasma metabolome after interventions with fermentable oligosaccharides, disaccharides, monosaccharides, and polyols (FODMAPs) or gluten versus control in IBS, and to relate such alterations to symptoms. People with IBS (n = 110) were included in a double-blind, randomized, crossover study with 1-wk provocations of FODMAPs, gluten, or placebo. Symptoms were evaluated with the IBS severity scoring system (IBS-SSS). Untargeted metabolomics was performed on plasma samples using LC-qTOF-MS. Discovery of metabolite alterations by treatment was performed using random forest followed by linear mixed modeling. Associations were studied using Spearman correlation. The metabolome was affected by FODMAP [classification rate (CR) 0.88, P < 0.0001], but less by gluten intake CR 0.72, P = 0.01). FODMAP lowered bile acids, whereas phenolic-derived metabolites and 3-indolepropionic acid (IPA) were higher compared with placebo. IPA and some unidentified metabolites correlated weakly to abdominal pain and quality of life. Gluten affected lipid metabolism weakly, but with no interpretable relationship to IBS. FODMAP affected gut microbial-derived metabolites relating to positive health outcomes. IPA and unknown metabolites correlated weakly to IBS severity. Minor symptom worsening by FODMAP intake must be weighed against general positive health aspects of FODMAP. The gluten intervention affected lipid metabolism weakly with no interpretable association to IBS severity. Registration: www.clinicaltrials.gov as NCT03653689.NEW & NOTEWORTHY In irritable bowel syndrome (IBS), fermentable oligo-, di-, monosaccharides, and polyols (FODMAPs) affected microbial-derived metabolites relating to positive health outcomes such as reduced risk of colon cancer, inflammation, and type 2 diabetes, as shown in previous studies. The minor IBS symptom induction by FODMAP intake must be weighed against the positive health aspects of FODMAP consumption. Gluten affected lipids weakly with no association to IBS severity.
Collapse
Affiliation(s)
- Elise Nordin
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Per M Hellström
- Department of Medical Sciences, Gastroenterology/Hepatology, Uppsala University, Uppsala, Sweden
| | - Eddie Vuong
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Anton Ribbenstedt
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Carl Brunius
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Rikard Landberg
- Department of Biology and Biological Engineering, Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| |
Collapse
|
50
|
Sarvepalli SS, Vemula SL, Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR. Digesting the Impact of Diet on Irritable Bowel Syndrome (IBS): Exploring Solutions for Controlling IBS. Cureus 2023; 15:e45279. [PMID: 37846263 PMCID: PMC10576851 DOI: 10.7759/cureus.45279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/18/2023] Open
Abstract
Irritable bowel syndrome (IBS) plagues nearly a fifth of the general population. It is a chronic illness that can significantly lower quality of life (QoL) and work productivity. The relationship between diet and the functional gastrointestinal (GI) symptoms present in IBS is gaining more and more attention. In addition to being a factor in the pathophysiology of IBS, diet also has a significant impact on symptoms and overall well-being. Recent research has also shown that short-chain fermentable carbohydrates increase colonic gas production and small intestine water volume, which in turn causes functional GI symptoms in those with visceral hypersensitivity. This review article has consolidated various studies highlighting the association between certain foods and the pathophysiology of IBS. It has also talked about how restricting certain food items from the diet of affected individuals can relieve symptoms and in some cases can be more effective than pharmacotherapy. Although the low reduced fermentable oligosaccharide, disaccharide, monosaccharide, and polyol (FODMAP) diet (LFD) is a well-known method of treating IBS symptoms, over a third of individuals do not benefit from it. This article has also discussed the effectiveness and applicability of the LFD compared to other dietary therapies for the long-term management of IBS.
Collapse
Affiliation(s)
| | - Shree Laya Vemula
- Department of Research, Anam Chenchu Subba Reddy (ACSR) Government Medical College, Nellore, IND
| | | | - Raam Mannam
- Department of General Surgery, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Department of Research, Narayana Medical College, Nellore, IND
| | | |
Collapse
|