51
|
Naik TD, Tubaki BR, Patankar DS. Efficacy of whole system ayurveda protocol in irritable bowel syndrome – A Randomized controlled clinical trial. J Ayurveda Integr Med 2022; 14:100592. [PMID: 36371363 PMCID: PMC10105243 DOI: 10.1016/j.jaim.2022.100592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the clinically challenging disorders. It has a significant effect on health, cost and quality of life. Ayurveda management through whole system approach in IBS is explored. OBJECTIVE To evaluate the efficacy of whole system Ayurveda approach in IBS. METHODS The present trial is a randomized controlled parallel group study. 48 patients diagnosed as IBS (Rome IV Criteria) between the age group of 20-60 yrs were recruited in the study. Patients were randomly divided into 2 groups. KC group intervened with Kalingadi Churna 3 gm twice a day, before food with buttermilk. WS group intervened with whole system ayurveda protocol (WSAP). Duration of intervention was 60 days with follow up on every 15th day. Assessments were through various clinical measures like IBS Symptom Severity Score (IBS-SSS), IBS Adequate Relief (IBS-AR), Gastrointestinal symptom rating scale (GSRS), IBS-VAS, Complete Spontaneous Bowel Movements (CSBMs), Bristol Stool Form (BSF), Hamilton Anxiety Rating Scale (HARS), Hamilton Depression Rating Scale (HDRS), IBS quality of life (IBS-QoL) at every follow up. Hemoglobin, Erythrocyte sedimentation rate and stool examination was conducted at pre and post study. RESULTS Study showed that WS group had significant improvement compared to KC group in IBS-SSS, IBS-AR, IBS-VAS, CSBM, BSF-Diarrhea and BSF-Constipation. Both groups were comparable in GSRS, HARS, HDRS and IBS-QOL. Blood and stool parameters assessments showed comparable improvements in both the groups. Within group significant improvements in all the clinical assessment scales were observed in both the groups. CONCLUSION WSAP was effective in management of IBS (IBS constipation and IBS diarrhea). Improvements were observed in abdominal pain, stool frequency, consistency and adequate relief.
Collapse
Affiliation(s)
- Teja D Naik
- Department of Kayachikitsa, Shri BMK Ayurveda Mahavidyalaya, A Constituent Unit of KLE Academy of Higher Education & Research, Belagavi, Karnataka, 590003, India
| | - Basavaraj R Tubaki
- Department of Kayachikitsa, Shri BMK Ayurveda Mahavidyalaya, A Constituent Unit of KLE Academy of Higher Education & Research, Belagavi, Karnataka, 590003, India.
| | - Devayani S Patankar
- Department of Kayachikitsa, Shri BMK Ayurveda Mahavidyalaya, A Constituent Unit of KLE Academy of Higher Education & Research, Belagavi, Karnataka, 590003, India
| |
Collapse
|
52
|
Wang Y, Chen N, Niu F, Li Y, Guo K, Shang X, E F, Yang C, Yang K, Li X. Probiotics therapy for adults with diarrhea-predominant irritable bowel syndrome: a systematic review and meta-analysis of 10 RCTs. Int J Colorectal Dis 2022; 37:2263-2276. [PMID: 36251040 DOI: 10.1007/s00384-022-04261-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Accumulating evidence showed that probiotics therapy might be effective in treating diarrhea-predominant irritable bowel syndrome (IBS-D). This study aimed to evaluate the effectiveness and safety of probiotics therapy for the treatment of IBS-D. METHODS We performed a comprehensive literature search in eight electronic databases, and gray literature from inception to August 4, 2021. Randomized controlled trials (RCTs) of probiotics therapy for the treatment of IBS-D were included and the quality was assessed using the risk of bias tool recommended by the Cochrane Handbook version 5.1.0. RevMan 5.4 software was used to perform the meta-analysis on the outcomes of IBS-D symptoms, abdominal pain, quality of life, and abdominal distension. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach was used to assess the certainty of evidence. RESULTS Ten RCTs evaluating 943 patients were identified. Only one study had unclear risk of bias, while nine studies had a high risk of bias. The meta-analysis results showed that, compared to the placebo, probiotics therapy significantly decreased the score of IBS-D symptoms (SMD = - 0.55, 95% CI: [- 0.83, - 0.27], P < 0.05), abdominal pain (SMD = - 0.43, 95% CI: [- 0.57, - 0.29], P < 0.05), and abdominal distension (SMD = - 0.45, 95%CI: [- 0.81, - 0.09], P < 0.05). There was no statistical difference in the quality of life. However, all the certainty of evidence was very low. CONCLUSION Very low certainty evidence showed that probiotics might be an effective treatment for improving the IBS-D symptoms, abdominal pain, and abdominal distension, in adult IBS-D patients. However, these conclusions should be supported by high-quality evidence.
Collapse
Affiliation(s)
- Yan Wang
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Nan Chen
- Research and Education Department, Shaanxi Provincial Rehabilitation Hospital, Xi'an, China
| | - Fangfen Niu
- First Hospital of Lanzhou University, Lanzhou, China
| | - Yanfei Li
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Kangle Guo
- Gansu Provincial Hospital, Lanzhou, China
| | - Xue Shang
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Fenfen E
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Chaoqun Yang
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China.,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Kehu Yang
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China. .,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China. .,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.
| | - Xiuxia Li
- Evidence-Based Social Science Research Center & Health Technology Assessment Center, School of Public Health, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China. .,Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China. .,Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China.
| |
Collapse
|
53
|
Novel Drug Therapeutics in Celiac Disease: A Pipeline Review. Drugs 2022; 82:1515-1526. [PMID: 36251239 DOI: 10.1007/s40265-022-01784-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/03/2022]
Abstract
Celiac disease (CeD) is a chronic, autoimmune systemic disorder triggered by the ingestion of gluten, a protein found in foods such as wheat, rye, and barley. The only effective treatment for CeD is complete removal of gluten from the diet. A strict gluten-free diet (GFD) results in symptomatic, serologic, and histologic remission in most patients. However, GFD may fail to induce clinical or histologic improvement and some patients may alternatively have difficulty strictly adhering to the GFD for other reasons. Despite this, there are currently no FDA-approved drugs for the treatment of CeD. The complex pathogenic process of CeD is becoming increasingly studied and better understood, enabling the identification of various targets for future therapies. Mechanisms under evaluation include probiotics, digestion of peptides, gluten sensitization, tight junction modulation, deamidation, and immune targets. Multiple investigational drugs are in the pipeline, and several drug candidates have entered late-phase clinical trials. Indeed, current and future studies are needed to target specific etiological mechanisms and provide an alternative to GFD alone. This review provides a broad overview of the various investigative treatment approaches for CeD, summarizing the latest progress in the pipeline.
Collapse
|
54
|
Lv L, Ruan G, Ping Y, Cheng Y, Tian Y, Xiao Z, Zhao X, Chen D, Wei Y. Clinical study on sequential treatment of severe diarrhea irritable bowel syndrome with precision probiotic strains transplantation capsules, fecal microbiota transplantation capsules and live combined bacillus subtilis and enterococcus faecium capsules. Front Cell Infect Microbiol 2022; 12:1025889. [PMID: 36250045 PMCID: PMC9555570 DOI: 10.3389/fcimb.2022.1025889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To study the effect of precision probiotic strains transplantation capsules on diarrhea irritable bowel syndrome compared with fecal microbiota transplantation capsules and live combined bacillus subtilis and enterococcus faecium capsules. Methods Two patients with severe irritable bowel syndrome were treated with precision probiotic strains transplantation capsules, fecal microbiota transplantation capsules and live combined bacillus subtilis and enterococcus faecium capsules in sequence. IBS-SSS, IBS-QoL, GSRS, stool frequency, stool character, degree of abdominal pain, GAD-7, and PHQ9 scores of patients at 0, 2, 4, 6, 8, 10, and 12 weeks of treatment were monitored and recorded, and stool samples were collected for metagenomics and metabolomics. Results It was found that the IBS-SSS score of patient case 1 decreased by 175 points and that of patient case 2 decreased by 100 points after treatment of precision probiotic strains transplantation capsules. There was no significant decrease after fecal microbiota transplantation capsules and live combined bacillus subtilis and enterococcus faecium capsules were used. At the same time, compared with fecal microbiota transplantation and live combined bacillus subtilis and enterococcus faecium capsules, the IBS QoL, stool frequency, stool character, degree of abdominal pain and GAD-7 score of patient case 1 improved more significantly by the precision probiotic strains transplantation capsules. And the stool frequency and stool character score of patient case 2 decreased more significantly. Intestinal microbiota also improved more significantly after the precise capsule transplantation treatment. And we found Eubacterium_ Eligens showed the same change trend in the treatment of two patients, which may play a role in the treatment. Conclusion precision probiotic strains transplantation capsules is more beneficial to improve the intestinal microbiota of patients than microbiota transplantation capsule and live combined bacillus subtilis and enterococcus faecium capsules, so as to better alleviate clinical symptoms. This study provides a more perfect and convenient therapeutic drugs for the treatment of IBS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanling Wei
- *Correspondence: Dongfeng Chen, ; Yanling Wei,
| |
Collapse
|
55
|
Zoghi S, Abbasi A, Heravi FS, Somi MH, Nikniaz Z, Moaddab SY, Ebrahimzadeh Leylabadlo H. The gut microbiota and celiac disease: Pathophysiology, current perspective and new therapeutic approaches. Crit Rev Food Sci Nutr 2022; 64:2176-2196. [PMID: 36154539 DOI: 10.1080/10408398.2022.2121262] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Celiac disease (CD) as a chronic gluten-sensitive intestinal condition, mainly affects genetically susceptible hosts. The primary determinants of CD have been identified as environmental and genetic variables. The development of CD is significantly influenced by environmental factors, including the gut microbiome. Therefore, gut microbiome re-programming-based therapies using probiotics, prebiotics, postbiotics, gluten-free diet, and fecal microbiota transplantation have shown promising results in the modification of the gut microbiome. Due to the importance and paucity of information regarding the CD pathophysiology, in this review, we have covered the association between CD development and gut microbiota, the effects of infectious agents, particularly the recent Covid-19 infection in CD patients, and the efficacy of potential therapeutic approaches in the CD have been discussed. Hence, scientific literature indicates that the diverse biological functions of the gut microbiota against immunomodulatory responses have made microbiome-based therapy an alternative therapeutic paradigm to ameliorate the symptoms of CD and quality of life. However, the exact potential of microbiota-based techniques that aims to quantitatively and qualitatively alter the gut microbiota to be used in the treatment and ameliorate the symptoms of CD will be determined with further research in the future.
Collapse
Affiliation(s)
- Sevda Zoghi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Yaghoub Moaddab
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
56
|
Merra G, Capacci A, De Lorenzo A, Di Renzo L, Gualtieri P, Frank G, Marchetti M. Does our microbiota eat with or without gluten? EXPLORATION OF MEDICINE 2022:275-279. [DOI: 10.37349/emed.2022.00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/25/2022] [Indexed: 01/05/2025] Open
Affiliation(s)
- Giuseppe Merra
- 1Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Annunziata Capacci
- 2Department of Medical and Surgical Sciences, Agostino Gemelli General Hospital Foundation-IRCCS, 00168 Rome, Italy
| | - Antonino De Lorenzo
- 1Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Laura Di Renzo
- 1Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Paola Gualtieri
- 1Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giulia Frank
- 3School of Specialisation in Food Science, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Marco Marchetti
- 1Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
57
|
Vacca M, Porrelli A, Calabrese FM, Lippolis T, Iacobellis I, Celano G, Pinto D, Russo F, Giannelli G, De Angelis M. How Metabolomics Provides Novel Insights on Celiac Disease and Gluten-Free Diet: A Narrative Review. Front Microbiol 2022; 13:859467. [PMID: 35814671 PMCID: PMC9260055 DOI: 10.3389/fmicb.2022.859467] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/27/2022] [Indexed: 12/12/2022] Open
Abstract
Celiac disease (CD) is an inflammatory autoimmune disorder triggered by the ingestion of gluten from wheat and other cereals. Nowadays, its positive diagnosis is based on invasive approaches such as the histological examination of intestinal biopsies and positive serology screening of antibodies. After proven diagnosis, the only admissible treatment for CD individuals is strict life-long adherence to gluten-free diet (GFD), although it is not a conclusive therapy. Acting by different mechanisms and with different etiologies, both CD and GFD have a great impact on gut microbiota that result in a different taxa composition. Altered production of specific metabolites reflects these microbiota changes. In this light, the currently available literature reports some suggestions about the possible use of specific metabolites, detected by meta-omics analyses, as potential biomarkers for a CD non-invasive diagnosis. To highlight insights about metabolomics application in CD study, we conducted a narrative dissertation of selected original articles published in the last decade. By applying a systematic search, it clearly emerged how the metabolomic signature appears to be contradictory, as well as poorly investigated.
Collapse
Affiliation(s)
- Mirco Vacca
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Porrelli
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
- *Correspondence: Francesco Maria Calabrese,
| | - Tamara Lippolis
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Ilaria Iacobellis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppe Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| | - Daniela Pinto
- Human Microbiome Advanced Project-HMPA, Giuliani SpA, Milan, Italy
| | - Francesco Russo
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Gianluigi Giannelli
- National Institute of Gastroenterology “S. de Bellis,” Institute of Research, Castellana Grotte, Italy
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
58
|
Parker S, Palsson O, Sanders DS, Simren M, Sperber AD, Törnblom H, Urwin H, Whitehead W, Aziz I. Functional Gastrointestinal Disorders and Associated Health Impairment in Individuals with Celiac Disease. Clin Gastroenterol Hepatol 2022; 20:1315-1325.e4. [PMID: 34298190 DOI: 10.1016/j.cgh.2021.07.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/28/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Individuals with celiac disease (CD) can experience persisting gastrointestinal symptoms despite adhering to a gluten-free diet (GFD). This may be due to functional gastrointestinal disorders (FGIDs), although there is little data on its prevalence and associated factors. METHODS An online health questionnaire was completed by adult members of Celiac UK in October 2018. The survey included validated questions on Rome IV FGIDs, nongastrointestinal somatic symptoms, anxiety, depression, quality of life, health care use, GFD duration, and its adherence using the celiac dietary adherence test score (with a value ≤ 13 indicating optimal adherence). The prevalence of FGIDs and associated health impairment in the celiac cohort was compared against an age- and sex-matched population-based control group. RESULTS Of the 863 individuals with CD (73% female; mean age, 61 years), all were taking a GFD for at least 1 year, with 96% declaring that they have been on the diet for 2 or more years (2-4 years, 20%; ≥5 years, 76%). The adherence to a GFD was deemed optimal in 61% (n = 523), with the remaining 39% (n = 340) nonadherent. Those adhering to a GFD fulfilled criteria for a FGID in approximately one-half of cases, although this was significantly lower than nonadherent subjects (51% vs 75%; odds ratio [OR], 2.0; P < .001). However, the prevalence of FGIDs in GFD-adherent subjects was significantly higher than in matched population-based controls (35%; OR, 2.0; P < .001). This was accounted for by functional bowel (46% vs 31%; OR, 1.9; P < .0001) and anorectal disorders (14.5% vs 9.3%; OR, 1.7; P = .02) but not functional esophageal (7.6% vs 6.1%; P = .36) or gastroduodenal disorders (8.7% vs 7.4%; P = .47). Finally, GFD-adherent subjects with FGIDs were significantly more likely than their counterparts without FGIDs to have abnormal levels of anxiety (5% vs 2%; OR, 2.8; P = .04), depression (7% vs 2%; OR, 3.6; P = .01), somatization (31% vs 8%; OR, 5.1; P < .0001), and reduced quality of life (P < .0001). CONCLUSION One in 2 people with CD, despite having been on a GFD for a number of years and demonstrating optimal adherence, have ongoing symptoms compatible with a Rome IV FGID. This is 2-fold the odds of FGIDs seen in age- and sex-matched controls. The presence of FGIDs is associated with significant health impairment, including psychological comorbidity. Addressing disorders of gut-brain interaction might improve outcomes in this specific group of patients.
Collapse
Affiliation(s)
- Sophie Parker
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, United Kingdom; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Olafur Palsson
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina, Chapel Hill, North Carolina
| | - David S Sanders
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, United Kingdom; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Magnus Simren
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina, Chapel Hill, North Carolina; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ami D Sperber
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hans Törnblom
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Heidi Urwin
- Coeliac UK, High Wycombe, Buckinghamshire, United Kingdom
| | - William Whitehead
- Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina, Chapel Hill, North Carolina
| | - Imran Aziz
- Academic Department of Gastroenterology, Sheffield Teaching Hospitals, Sheffield, United Kingdom; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
59
|
Bu Z, Ye X, Huang B, Liu R, Peng L. Bifidobacteria Was Decreased in Adult Patients With Irritable Bowel Syndrome Based on PCR and Bacterial Culture: A Systematic Review and Meta-Analysis. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2022; 33:368-376. [PMID: 35678794 PMCID: PMC11163281 DOI: 10.5152/tjg.2022.21543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/08/2021] [Indexed: 06/15/2023]
Abstract
The causes of irritable bowel syndrome remain unknown. Studies and meta-analyses revealed that intestinal microbiota disturbance was one of the causes of irritable bowel syndrome, but the results remained controversial. Therefore, we performed a systematic review and meta-analysis to identify the association between them. We performed a systematic meta-analysis of case-control studies from January 2000 to December 2020 to compare fecal microbes based on polymerase chain reaction and bacterial cul- ture between adult irritable bowel syndrome patients and healthy controls. The standardized mean difference value and a 95% CI were calculated. Two professional researchers used Newcastle-Ottawa Scale to reassess selected literature and extract high- quality studies. Six studies were included in our analysis. When all eligible studies were pooled into the meta-analysis, compared with healthy controls, the standardized mean differences of Bifidobacteria (standardized mean difference = -1.01, 95% CI =: -2.01 to -0.01) in irritable bowel syndrome patients decreased significantly, whereas the standardized mean differences of Enterococcus, Enterobacter, Lactobacillus, Bacteroides, and Escherichia coli did not change significantly in irritable bowel syndrome patients. However, heterogeneity was significant to perform sensitivity analysis and stratified analysis in all these special intestinal microbes. In summary, this study indicated that only Bifidobacteria was decreased in irritable bowel syndrome patients compared with healthy controls using Newcastle-Ottawa Scale standards to extract high-quality literature. Future studies are warranted to further dem- onstrate the relationship between them.
Collapse
Affiliation(s)
- Zhibin Bu
- Department of Ultrasound, Zhejiang University Faculty of Medicine Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Xianghua Ye
- Department of Radiotherapy, Zhejiang University Faculty of Medicine The First Affiliated Hospital, Hangzhou, Zhejiang Province, China
| | - Bin Huang
- Department of Ultrasound, Zhejiang University Faculty of Medicine Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Rui Liu
- Department of Radiotherapy, Xi’an Jiaotong University The First Affiliated Hospital, Xi’an, Shaanxi Province, China
| | - Ling Peng
- Department of Pulmonary and Critical Care Medicine, Hangzhou Medical College Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
60
|
Hata S, Nakajima H, Hashimoto Y, Miyoshi T, Hosomi Y, Okamura T, Majima S, Nakanishi N, Senmaru T, Osaka T, Okada H, Ushigome E, Hamaguchi M, Asano M, Yamazaki M, Fukui M. Effects of probiotic Bifidobacterium bifidum G9-1 on the gastrointestinal symptoms of patients with type 2 diabetes mellitus treated with metformin: An open-label, single-arm, exploratory research trial. J Diabetes Investig 2022; 13:489-500. [PMID: 34665938 PMCID: PMC8902400 DOI: 10.1111/jdi.13698] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 12/20/2022] Open
Abstract
AIMS/INTRODUCTION Metformin is associated with the risk of gastrointestinal complications, and probiotic Bifidobacterium bifidum G9-1 (BBG9-1) can improve the symptoms of diarrhea. This study aimed to clarify the effects of probiotic BBG9-1 on the gastrointestinal symptoms of type 2 diabetes mellitus patients using metformin. MATERIALS AND METHODS In this open-label single-arm exploratory study, 40 patients (mean age 64.0 ± 9.4 years) were given probiotic BBG9-1 for 10 weeks. Changes in the gastrointestinal symptom rating scale total score, which was the primary end-point, gastrointestinal symptom rating scale subscale scores, glycated hemoglobin levels and gut microbiota after the administration of probiotic BBG9-1 were evaluated by the Student's t-test. RESULTS The gastrointestinal symptom rating scale total score significantly improved (from 2.02 ± 0.51 to 1.59 ± 0.43, change, -0.43 ± 0.49, P < 0.001). Furthermore, all gastrointestinal symptom rating scale subscale scores, including diarrhea (from 2.32 ± 1.14 to 1.89 ± 0.99, change, -0.42 ± 0.95, P = 0.007) and constipation (from 3.00 ± 1.16 to 2.20 ± 1.07, change, -0.80 ± 1.19, P < 0.001), scores also significantly improved. However, the glycated hemoglobin levels did not change (from 7.0 ± 0.7 to 7.0 ± 0.6%, change, 0.0 ± 0.4, P = 0.91). The relative abundance of the genus Sutterella decreased by the use of probiotic BBG9-1 (from 0.011 ± 0.009 to 0.008 ± 0.006, change, -0.003 ± 0.006, P = 0.002). CONCLUSIONS Type 2 diabetes mellitus patients treated with metformin showed significant improvement in all gastrointestinal symptom rating scores after using probiotic BBG9-1 without changing the glucose control. This study showed the potential usefulness of probiotic BBG9-1 for improving gastrointestinal symptoms, including constipation and diarrhea, in type 2 diabetes mellitus patients treated with metformin.
Collapse
Affiliation(s)
- Shinnosuke Hata
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hanako Nakajima
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yoshitaka Hashimoto
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Tomoki Miyoshi
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yukako Hosomi
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Takuro Okamura
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Saori Majima
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Naoko Nakanishi
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Takafumi Senmaru
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Takafumi Osaka
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Hiroshi Okada
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Emi Ushigome
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahide Hamaguchi
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Mai Asano
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Yamazaki
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Michiaki Fukui
- Department of Endocrinology and MetabolismGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
61
|
Treppiccione L, Luongo D, Maurano F, Rossi M. Next generation strategies to recover immunological tolerance in celiac disease. Int Rev Immunol 2022; 42:237-245. [PMID: 35225129 DOI: 10.1080/08830185.2022.2044807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Celiac disease (CD) is an autoimmune disease that occurs in genetically predisposed individuals following the ingestion of gluten. Its prevalence is rising worldwide. A gluten-free (GF) diet is mandatory for the management of CD. However, several issues persist regarding the nutritional quality of GF products. Importantly, deep knowledge about the pathogenic mechanisms in CD highlights the central role of CD4+ T cell-mediated immunity in CD. Furthermore, intestinal T regulatory cells are functional in CD, but cytokines such as IL-15, produced under inflammatory conditions, hamper their activity. This paves the way for the development of immunomodulatory strategies to the GF diet. From this perspective, microbiological approaches were considered able to modulate the gluten-specific immune response. Interestingly, gliadin peptide-based immunotherapy to abolish the inflammatory CD4+T cell-mediated response has been explored in CD patients. Furthermore, different biotechnological approaches based on the use of chemically/enzymatically modified gluten molecules have been proved effective in different models of CD. However, the choice of the right age in infants to introduce the antigen and thus induce tolerance still remains an important issue to solve. Addressing all these points should help to design an effective intervention strategy for preventing CD.
Collapse
Affiliation(s)
| | | | | | - Mauro Rossi
- Institute of Food Sciences, CNR, Avellino, Italy
| |
Collapse
|
62
|
Feng HY, Chan CH, Chu YC, Qu XM, Wang YH, Wei JCC. Patients with ankylosing spondylitis have high risk of irritable bowel syndrome. A long-term nationwide population-based cohort study. Postgrad Med 2022; 134:290-296. [PMID: 35139724 DOI: 10.1080/00325481.2022.2041338] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Ankylosing spondylitis (AS) is a chronic inflammatory disease, might carry a high risk of irritable bowel syndrome (IBS) due to abnormal gut microbiota or inflammatory reaction. METHODS We conducted a 14-year retrospective cohort study based on Taiwan's National Health Insurance Research Database (NHIRD). A total of 4007 patients with newly diagnosed AS (outpatient visits≧3 times, or hospitalization≧1 time) and 988,084 non-AS comparisons were enrolled during 2000-2012. To ensure baseline comparability, the propensity score was matched by age, gender, comorbidities, and other possible confounders. The outcome was the incidence of IBS, followed up to the end of 2013. Cox proportional hazard model calculated adjusted hazard ratio (aHR) and the cumulative incidence of both groups was analyzed by the Kaplan-Meier method. RESULT After propensity score matching, baseline demographic characteristics were comparable between AS patients and the comparison group. The crude HR for IBS in the AS group was significantly higher 2.41 (95%C.I. =1.84-3.16) than comparison group. After adjusting for possible confounders, adjusted HR was 2.50 (95%C.I.=1.91-3.29). The cumulative incidence of IBS in AS was significantly higher than non-AS comparisons during the 14-year follow-up (P<0.001). CONCLUSION This nationwide population-based cohort study showed that patients with AS have higher risks of IBS than those of the non-AS comparison group.
Collapse
Affiliation(s)
- Hao-Yuan Feng
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Chi-Ho Chan
- Department of Microbiology and Immunology, Chung Shan Medical University, Taichung 402, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Yu-Cheng Chu
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Xin-Man Qu
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
| | - Yu-Hsun Wang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - James Cheng-Chung Wei
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan.,Department of Allergy, Immunology & Rheumatology, Chung Shan Medical University Hospital, Taichung 402, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan.,Graduate Institute of Integrated Medicine, China Medical University, Taichung 402, Taiwan
| |
Collapse
|
63
|
Patra F, Duary RK. Determination and Safety Aspects of Probiotic Cultures. PROBIOTICS, PREBIOTICS AND SYNBIOTICS 2022:122-160. [DOI: 10.1002/9781119702160.ch6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
64
|
Singh A, Kaur H, Midha V, Sood A. Microorganisms in the Pathogenesis and Management of Celiac Disease (CeD). ROLE OF MICROORGANISMS IN PATHOGENESIS AND MANAGEMENT OF AUTOIMMUNE DISEASES 2022:287-307. [DOI: 10.1007/978-981-19-4800-8_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
65
|
Mirzaie Z, Bastani A, Haji-Aghamohammadi AA, Rashidi Nooshabadi M, Ahadinezhad B, Khadem Haghighian H. Effects of Ellagic Acid on Oxidative Stress Index, Inflammatory Markers and Quality of Life in Patients With Irritable Bowel Syndrome: Randomized Double-blind Clinical Trial. Clin Nutr Res 2022; 11:98-109. [PMID: 35558999 PMCID: PMC9065395 DOI: 10.7762/cnr.2022.11.2.98] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a common disorder that affects the large intestine. Oxidative stress and inflammation play a major role in IBS. Considering the antioxidant properties of ellagic acid (EA), this study was designed to evaluate the effect of EA on oxidative stress index, inflammatory markers, and quality of life in patients with IBS. This research was conducted as a randomized, double-blind, placebo-controlled clinical trial; 44 patients with IBS were recruited. Patients who met the inclusion criteria were randomly allocated to consume a capsule containing 180 mg of EA per day (n = 22) or a placebo (n = 22) for 8 weeks. Serum levels of total antioxidant capacity (TAC), malondialdehyde (MDA), C-reactive protein (CRP), and interleukin-6 (IL-6) were measured at the beginning and the end of the study. Also, quality of life was assessed using a self-report questionnaire for IBS patients (IBS-QOL). At the end of the study, we saw a significant decrease and increase in the MDA and TAC in the intervention group, respectively (p < 0.05). Also, EA consumption reduced CRP and IL-6 levels, and these changes were significant in comparison with placebo group changes (p < 0.05). The overall score of IBS-QOL significantly decreased, and quality of life was increased (p < 0.05), but there were no significant changes in the placebo group. According to these findings, receiving polyphenols, such as EA, may help maintain intestinal health by modulating inflammation and oxidative stress and ultimately improving the quality of life in IBS patients.
Collapse
Affiliation(s)
- Zahra Mirzaie
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Ali Bastani
- Department of Internal Medicine, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Ali Akbar Haji-Aghamohammadi
- Department of Internal Medicine, Velayat Clinical Research Development Unit, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | | | - Bahman Ahadinezhad
- Social Determinants of Health Research Center, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| | - Hossein Khadem Haghighian
- Department of Nutrition, School of Health, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin 34197-59811, Iran
| |
Collapse
|
66
|
Yoosuf S, Therrien A, Leffler DA. Non-dietary therapies for celiac disease. COELIAC DISEASE AND GLUTEN-RELATED DISORDERS 2022:111-160. [DOI: 10.1016/b978-0-12-821571-5.00011-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
67
|
Meurman J, Stamatova I. Probiotics for oral health and disease treatment. PROBIOTICS FOR HUMAN NUTRITION IN HEALTH AND DISEASE 2022:413-430. [DOI: 10.1016/b978-0-323-89908-6.00008-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
68
|
|
69
|
Wu X, Qian L, Liu K, Wu J, Shan Z. Gastrointestinal microbiome and gluten in celiac disease. Ann Med 2021; 53:1797-1805. [PMID: 34647492 PMCID: PMC8519548 DOI: 10.1080/07853890.2021.1990392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/30/2021] [Indexed: 01/11/2023] Open
Abstract
Coeliac disease (CD), also known as gluten sensitive enteropathy, is an autoimmune intestinal disease induced by gluten in genetically susceptible individuals. Gluten is a common ingredient in daily diet and is one of the main environmental factors to induce coeliac disease. Adhering to gluten free diet (GFD) is an effective method for treating CD. Microbiota plays an extremely important role in maintaining human health, and diet is the main factor to regulate the composition and function of gut microbiota. Recent studies have shown that gluten metabolism is closely related to gastrointestinal tract (GIT) microbiota. With the increasing prevalence of coeliac disease, there is a need for alternative treatments to GFD. In this review, biological medication of gluten, relationship between gluten and gut microflora, effect of GFD on GIT microflora, and effect of probiotics on CD were reviewed. By analysing the research progress on relationship between gluten and gastrointestinal microbiome in coeliac disease, this review tried to explore the prospective and potential mechanism of microecological agents in treating coeliac disease.
Collapse
Affiliation(s)
- Xingxing Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lin Qian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kexin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wu
- Institute of Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing University, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaowei Shan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
70
|
Fabiano V, Indrio F, Verduci E, Calcaterra V, Pop TL, Mari A, Zuccotti GV, Cullu Cokugras F, Pettoello-Mantovani M, Goulet O. Term Infant Formulas Influencing Gut Microbiota: An Overview. Nutrients 2021; 13:4200. [PMID: 34959752 PMCID: PMC8708119 DOI: 10.3390/nu13124200] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/19/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023] Open
Abstract
Intestinal colonization of the neonate is highly dependent on the term of pregnancy, the mode of delivery, the type of feeding [breast feeding or formula feeding]. Postnatal immune maturation is dependent on the intestinal microbiome implementation and composition and type of feeding is a key issue in the human gut development, the diversity of microbiome, and the intestinal function. It is well established that exclusive breastfeeding for 6 months or more has several benefits with respect to formula feeding. The composition of the new generation of infant formulas aims in mimicking HM by reproducing its beneficial effects on intestinal microbiome and on the gut associated immune system (GAIS). Several approaches have been developed currently for designing new infant formulas by the addition of bioactive ingredients such as human milk oligosaccharides (HMOs), probiotics, prebiotics [fructo-oligosaccharides (FOSs) and galacto-oligosaccharides (GOSs)], or by obtaining the so-called post-biotics also known as milk fermentation products. The aim of this article is to guide the practitioner in the understanding of these different types of Microbiota Influencing Formulas by listing and summarizing the main concepts and characteristics of these different models of enriched IFs with bioactive ingredients.
Collapse
Affiliation(s)
- Valentina Fabiano
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Flavia Indrio
- Department of Medical and Surgical Sciences, University of Foggia, 71100 Foggia, Italy
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
| | - Elvira Verduci
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Valeria Calcaterra
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
- Pediatric and Adolescent Unit, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Tudor Lucian Pop
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Second Paediatric Clinic, Department of Mother and Child, University of Medicine and Pharmacy Iuliu Hatieganu, 400177 Cluj-Napoca, Romania
| | - Alessandra Mari
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Department of Pediatrics, Vittore Buzzi Children’s Hospital, Università degli Studi di Milano, 20154 Milan, Italy; (V.F.); (E.V.); (V.C.); (A.M.); (G.V.Z.)
| | - Fugen Cullu Cokugras
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Paediatric Gastroenterology, Hepatology and Nutrition, Cerrahpasa Medical Faculty, Istanbul University, Istanbul 34000, Turkey
| | - Massimo Pettoello-Mantovani
- European Paediatric Association/Union of National European Paediatric Societies and Associations, 10115 Berlin, Germany; (T.L.P.); (F.C.C.); (M.P.-M.)
- Department of Pediatrics, Scientific Institute ‘Casa Sollievo della Sofferenza’, University of Foggia, 71122 Foggia, Italy
- Association pour l’Activité et la Recherche Scìentifiques, EPA-UNEPSA/ARS, 2000 Neuchâtel, Switzerland
| | - Olivier Goulet
- Department of Paediatric Gastroenterology, and Nutrition, Intestinal Failure Rehabilitation Centre, National Reference Centre for Rare Digestive Diseases, Necker-Enfants Malades Hospital, Paris Centre University and Paris-Descartes School of Medicine, 75000 Paris, France;
| |
Collapse
|
71
|
Yu D, Meng X, de Vos WM, Wu H, Fang X, Maiti AK. Implications of Gut Microbiota in Complex Human Diseases. Int J Mol Sci 2021; 22:12661. [PMID: 34884466 PMCID: PMC8657718 DOI: 10.3390/ijms222312661] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Humans, throughout the life cycle, from birth to death, are accompanied by the presence of gut microbes. Environmental factors, lifestyle, age and other factors can affect the balance of intestinal microbiota and their impact on human health. A large amount of data show that dietary, prebiotics, antibiotics can regulate various diseases through gut microbes. In this review, we focus on the role of gut microbes in the development of metabolic, gastrointestinal, neurological, immune diseases and, cancer. We also discuss the interaction between gut microbes and the host with respect to their beneficial and harmful effects, including their metabolites, microbial enzymes, small molecules and inflammatory molecules. More specifically, we evaluate the potential ability of gut microbes to cure diseases through Fecal Microbial Transplantation (FMT), which is expected to become a new type of clinical strategy for the treatment of various diseases.
Collapse
Affiliation(s)
- Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Xin Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Amit K. Maiti
- Department of Genetics and Genomics, Mydnavar, 2645 Somerset Boulevard, Troy, MI 48084, USA
| |
Collapse
|
72
|
Alterations in Gut Microbiome Composition and Function in Irritable Bowel Syndrome and Increased Probiotic Abundance with Daily Supplementation. mSystems 2021; 6:e0121521. [PMID: 34726487 PMCID: PMC8562475 DOI: 10.1128/msystems.01215-21] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Irritable bowel syndrome (IBS) is characterized by abdominal discomfort and irregular bowel movements and stool consistency. As such, the gut microbiome has been posited as being influential for the syndrome. However, identifying microbial features associated with IBS symptom heterogeneity is difficult without large cohorts. Our aim was to identify microbial features associated with IBS and IBS subtypes compared to healthy controls and to determine if a synbiotic supplementation intervention could decrease the proportion of those microbial features. Stool samples from 490 individuals with IBS (including all dominant subtypes) and 122 individuals without IBS were analyzed with metagenomic sequencing. One hundred thirty-four IBS subjects were followed over time while receiving daily synbiotic supplementation, the composition of which varied between participants. IBS participants had significantly lower alpha diversity, an enrichment in Gram-negative bacteria, and a reduction in pathways associated with short-chain fatty acid and vitamin synthesis. Shigella species were significantly associated with IBS, while Eubacterium rectale and Faecalibacterium prausnitzii were associated with healthy controls. Random forest identified unique and overlapping microbial features associated with each IBS subtype. Longitudinal assessment of 134 IBS subjects receiving synbiotic supplements demonstrated a significant difference in microbial features and an increase in probiotic abundance across time. We identified microbial features that differentiate healthy and IBS subtypes. Synbiotic supplementation in IBS subjects did not result in alpha diversity change in the microbiome but did demonstrate changes in microbial features. Future work is needed to determine if the observed microbiome changes are associated with IBS symptom improvement. IMPORTANCE An estimated 35 million people in the United States and 11.5% of the population globally are affected by IBS. Immunity, genetics, environment, diet, small intestinal bacterial overgrowth (SIBO), and the gut microbiome are all factors that contribute to the onset or triggers of IBS. With strong supporting evidence that the gut microbiome may influence symptoms associated with IBS, elucidating the important microbes that contribute to the symptoms and severity is important to make decisions for targeted treatment. As probiotics have become more common in treating IBS symptoms, identifying effective probiotics may help inform future studies and treatment.
Collapse
|
73
|
Varesi A, Deumer US, Ananth S, Ricevuti G. The Emerging Role of Gut Microbiota in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Current Evidence and Potential Therapeutic Applications. J Clin Med 2021; 10:jcm10215077. [PMID: 34768601 PMCID: PMC8584653 DOI: 10.3390/jcm10215077] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
The well-known symptoms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) are chronic pain, cognitive dysfunction, post-exertional malaise and severe fatigue. Another class of symptoms commonly reported in the context of ME/CFS are gastrointestinal (GI) problems. These may occur due to comorbidities such as Crohn's disease or irritable bowel syndrome (IBS), or as a symptom of ME/CFS itself due to an interruption of the complex interplay between the gut microbiota (GM) and the host GI tract. An altered composition and overall decrease in diversity of GM has been observed in ME/CFS cases compared to controls. In this review, we reflect on genetics, infections, and other influences that may factor into the alterations seen in the GM of ME/CFS individuals, we discuss consequences arising from these changes, and we contemplate the therapeutic potential of treating the gut to alleviate ME/CFS symptoms holistically.
Collapse
Affiliation(s)
- Angelica Varesi
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy
- Almo Collegio Borromeo, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| | - Undine-Sophie Deumer
- Department of Biological Sciences, Faculty of Natural Sciences and Mathematics, University of Cologne, 50674 Cologne, Germany;
| | - Sanjana Ananth
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London SW7 2AZ, UK;
| | - Giovanni Ricevuti
- Department of Drug Sciences, School of Pharmacy, University of Pavia, 27100 Pavia, Italy
- Correspondence: (A.V.); (G.R.)
| |
Collapse
|
74
|
Conte M, Porpora M, Nigro F, Nigro R, Budelli AL, Barone MV, Nanayakkara M. Pro-Pre and Postbiotic in Celiac Disease. APPLIED SCIENCES 2021; 11:8185. [DOI: 10.3390/app11178185] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Celiac Disease (CD) is an autoimmune disease characterized by inflammation of the intestinal mucosa due to an immune response to wheat gliadins. It presents in subjects with genetic susceptibility (HLA-DQ2/DQ8 positivity and non-HLA genes) and under the influence of environmental triggers, such as viral infections and intestinal microbiota dysbiosis. The only treatment currently available in CD is a gluten-free diet for life. Despite this, the intestinal dysbiosis that is recorded in celiac subjects persists, even with adherence to dietary therapy. In this review, we have analyzed the literature over the past several decades, which have focused on the use of pro-, pre- and post-biotics in vitro and in vivo in CD. The study of probiotics and their products in CD could be interesting for observing their various effects on several different pathways, including anti-inflammatory properties.
Collapse
Affiliation(s)
- Mariangela Conte
- Department of Translational Medical Science (Section of Pediatrics), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Monia Porpora
- Department of Translational Medical Science (Section of Pediatrics), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Federica Nigro
- School of Engineering, Niccoló Cusano University, 00166 Rome, Italy
- I.T.P. Innovation and Technology Provider S.r.l., Via Bisignano a Chiaia 68, 80121 Naples, Italy
| | - Roberto Nigro
- DICMAPI, University of Naples Federico II, 80125 Naples, Italy
| | - Andrea Luigi Budelli
- School of Engineering, Niccoló Cusano University, 00166 Rome, Italy
- DICMAPI, University of Naples Federico II, 80125 Naples, Italy
| | - M. Vittoria Barone
- Department of Translational Medical Science (Section of Pediatrics), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| | - Merlin Nanayakkara
- Department of Translational Medical Science (Section of Pediatrics), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples Federico II, Via S. Pansini 5, 80131 Naples, Italy
| |
Collapse
|
75
|
Le Morvan de Sequeira C, Kaeber M, Cekin SE, Enck P, Mack I. The Effect of Probiotics on Quality of Life, Depression and Anxiety in Patients with Irritable Bowel Syndrome: A Systematic Review and Meta-Analysis. J Clin Med 2021; 10:jcm10163497. [PMID: 34441793 PMCID: PMC8397103 DOI: 10.3390/jcm10163497] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Functional gastrointestinal disorders such as irritable bowel syndrome (IBS) report clinical improvement following probiotic therapy, but whether psychiatric comorbidity and quality-of-life in IBS improves directly or in directly is unknown. This meta-analysis synthesized the evidence regarding the effects of probiotics on quality of life (QoL), anxiety and depression in IBS. Methods: The review was executed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines using the databases PubMed, Web of Science and Cochrane Library. For QoL, the data were meta-analyzed, and for anxiety and depression a qualitative analysis was performed. Results: Thirty-five placebo-controlled studies were included of which 11 were eligible for meta-analysis. QoL improved with probiotic and placebo similarly, with the probiotic interventions slightly superior (mean QoL difference—0.36 (95% CI: 0.07, 0.64); p = 0.01). Anxiety and depression were similar between placebo and probiotic groups following therapy. Conclusion: For IBS, probiotic therapy improved QoL, but had no effects on anxiety and depression. However, the applied probiotics were not developed for selective effects on psyche and the brain. Therefore, it remains to be shown whether or not patients with IBS would benefit from second generation probiotics developed for these central effects (psychobiotics).
Collapse
|
76
|
Elsouri K, Arboleda V, Heiser S, Kesselman MM, Demory Beckler M. Microbiome in Rheumatoid Arthritis and Celiac Disease: A Friend or Foe. Cureus 2021; 13:e15543. [PMID: 34277168 PMCID: PMC8269990 DOI: 10.7759/cureus.15543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/08/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis (RA) and celiac disease (CD) are both autoimmune diseases with increasing global prevalence. These two diseases have been connected based on similar HLA mutations, serological markers, rheumatological, and gastrointestinal manifestations. In this review, we discuss the role of the oral and gut microbiome in the development and progression of RA and CD. Here, we highlight similar microbial dysbiosis and how these alterations in composition can lead to worsening disease severity in both CD and RA. Additionally, we analyze the role of probiotics in regulating the microbiome and improving symptoms associated with RA and CD.
Collapse
Affiliation(s)
- Kawther Elsouri
- Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Vania Arboleda
- Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, USA
| | - Samantha Heiser
- Medicine, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| | - Marc M Kesselman
- Rheumatology, Nova Southeastern University Dr. Kiran C. Patel College of Osteopathic Medicine, Davie, USA
| | - Michelle Demory Beckler
- Microbiology and Immunology, Nova Southeastern University Dr. Kiran C. Patel College of Allopathic Medicine, Fort Lauderdale, USA
| |
Collapse
|
77
|
Segura V, Ruiz-Carnicer Á, Sousa C, Moreno MDL. New Insights into Non-Dietary Treatment in Celiac Disease: Emerging Therapeutic Options. Nutrients 2021; 13:2146. [PMID: 34201435 PMCID: PMC8308370 DOI: 10.3390/nu13072146] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
To date, the only treatment for celiac disease (CD) consists of a strict lifelong gluten-free diet (GFD), which has numerous limitations in patients with CD. For this reason, dietary transgressions are frequent, implying intestinal damage and possible long-term complications. There is an unquestionable need for non-dietary alternatives to avoid damage by involuntary contamination or voluntary dietary transgressions. In recent years, different therapies and treatments for CD have been developed and studied based on the degradation of gluten in the intestinal lumen, regulation of the immune response, modulation of intestinal permeability, and induction of immunological tolerance. In this review, therapeutic lines for CD are evaluated with special emphasis on phase III and II clinical trials, some of which have promising results.
Collapse
Affiliation(s)
| | | | | | - María de Lourdes Moreno
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (V.S.); (Á.R.-C.); (C.S.)
| |
Collapse
|
78
|
The Modification of the Gut Microbiota via Selected Specific Diets in Patients with Crohn's Disease. Nutrients 2021; 13:nu13072125. [PMID: 34206152 PMCID: PMC8308385 DOI: 10.3390/nu13072125] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal symptoms in Crohn’s disease (CD) are common and affect the quality of life of patients; consequently, a growing number of studies have been published on diet interventions in this group. The role of the gut microbiota in the pathogenesis and the progression of inflammatory bowel diseases (IBD), including CD, has been widely discussed. Mainly, a decreased abundance of Firmicutes, species of the Bifidobacterium genus, and the Faecalibacterium prausnitzii species as well as a reduced general diversity have been described. In this review article, we summarize available data on the influence of reduction diets on the microbiome of patients with CD. One of the most frequently used elimination diets in CD patients is the low-FODMAP (Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols) diet. Although many papers show it may reduce abdominal pain, diarrhea, or bloating, it also reduces the intake of prebiotic substances, which can negatively affect the gut microbiota composition, decreasing the abundance of Bifidobacterium species and Faecalibacterium prausnitzii. Other elimination diets used by IBD patients, such as lactose-free or gluten-free diets, have also been shown to disturb the microbial diversity. On the other hand, CDED (Crohn’s disease exclusion diet) with partial enteral nutrition not only induces the remission of CD but also has a positive influence on the microbiota. The impact of diet interventions on the microbiota and, potentially, on the future course of the disease should be considered when nutritional guidelines for IBD patients are designed. Dietetic recommendations should be based not only on the regulation of the symptoms but also on the long-term development of the disease.
Collapse
|
79
|
Li X, Hu D, Tian Y, Song Y, Hou Y, Sun L, Zhang Y, Man C, Zhang W, Jiang Y. Protective effects of a novel Lactobacillus rhamnosus strain with probiotic characteristics against lipopolysaccharide-induced intestinal inflammation in vitro and in vivo. Food Funct 2021; 11:5799-5814. [PMID: 32568317 DOI: 10.1039/d0fo00308e] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Lipopolysaccharides (LPS), a main component of the Gram-negative bacterial cell wall, can damage the epithelial wall barrier and induce chronic intestinal inflammation. The purpose of this study is to evaluate whether the novel L. rhamnosus could alleviate intestinal inflammation and damage induced by LPS and explore the possible underlying molecular mechanism. L. rhamnosus JL-1 was selected from five L. rhamnosus strains due to its strong adherence capacity to Caco-2 cells (92.89%) and it could survive in simulated gastrointestinal juices. Whole genome sequencing analysis showed that there were no translocation and inversion regions in the genome of L. rhamnosus JL-1 compared with L. rhamnosus GG. Comparative genomic analysis showed that there were encoding genes related to adhesion, acid resistance and bile salt resistance in the genome of L. rhamnosus JL-1. Both in vitro and in vivo experiments indicated that LPS challenge inhibited the mRNA and protein expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6). However, the mRNA and protein expressions of pro-inflammatory cytokines were inhibited by pre-treatment with L. rhamnosus JL-1 in a dose-dependent manner. The result of histopathology analysis of ileum showed that oral administration of L. rhamnosus JL-1 reduced pathological damage induced by LPS. Furthermore, it was revealed that L. rhamnosus JL-1 could inhibit the mRNA and protein expressions of TLR4 and NF-κB. These results strongly suggested that L. rhamnosus JL-1 relieved LPS-induced intestinal inflammation by inhibiting the TLR4/NF-κB signaling pathway. To sum up, L. rhamnosus JL-1 has a potential probiotic function and plays an important role in preventing LPS-induced intestinal inflammation and damage.
Collapse
Affiliation(s)
- Xuesong Li
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Dong Hu
- Institute of Genetics and Physiology, Hebei Academy of Agriculture and Forestry Sciences, Shijiazhuang, 050051, China
| | - Yazhen Tian
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yang Song
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yichao Hou
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Linlin Sun
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yu Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
80
|
De Angelis M, Siragusa S, Vacca M, Di Cagno R, Cristofori F, Schwarm M, Pelzer S, Flügel M, Speckmann B, Francavilla R, Gobbetti M. Selection of Gut-Resistant Bacteria and Construction of Microbial Consortia for Improving Gluten Digestion under Simulated Gastrointestinal Conditions. Nutrients 2021; 13:992. [PMID: 33808622 PMCID: PMC8003469 DOI: 10.3390/nu13030992] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED This work aimed to define the microbial consortia that are able to digest gluten into non-toxic and non-immunogenic peptides in the human gastrointestinal tract. METHODS 131 out of 504 tested Bacillus and lactic acid bacteria, specifically Bacillus (64), lactobacilli (63), Pediococcus (1), and Weissella (3), showed strong gastrointestinal resistance and were selected for their PepN, PepI, PepX, PepO, and PepP activities toward synthetic substrates. Based on multivariate analysis, 24 strains were clearly distinct from the other tested strains based on having the highest enzymatic activities. As estimated by RP-HPLC and nano-ESI-MS/MS, 6 cytoplasmic extracts out of 24 selected strains showed the ability to hydrolyze immunogenic epitopes, specifically 57-68 of α9-gliadin, 62-75 of A-gliadin, 134-153 of γ-gliadin, and 57-89 (33-mer) of α2-gliadin. Live and lysed cells of selected strains were combined into different microbial consortia for hydrolyzing gluten under gastrointestinal conditions. Commercial proteolytic enzymes (Aspergillusoryzae E1, Aspergillusniger E2, Bacillussubtilis Veron HPP, and Veron PS proteases) were also added to each microbial consortium. Consortium activity was evaluated by ELISA tests, RP-HPLC-nano-ESI-MS/MS, and duodenal explants from celiac disease patients. RESULTS two microbial consortia (Consortium 4: Lactiplantibacillus (Lp.) plantarum DSM33363 and DSM33364, Lacticaseibacillus (Lc.) paracasei DSM33373, Bacillussubtilis DSM33298, and Bacilluspumilus DSM33301; and Consortium 16: Lp. plantarum DSM33363 and DSM33364, Lc. paracasei DSM33373, Limosilactobacillusreuteri DSM33374, Bacillusmegaterium DSM33300, B.pumilus DSM33297 and DSM33355), containing commercial enzymes, were able to hydrolyze gluten to non-toxic and non-immunogenic peptides under gastrointestinal conditions. CONCLUSIONS the results of this study provide evidence that selected microbial consortia could potentially improve the digestion of gluten in gluten-sensitive patients by hydrolyzing the immunogenic peptides during gastrointestinal digestion.
Collapse
Affiliation(s)
- Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Sonya Siragusa
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.D.A.); (S.S.); (M.V.)
| | - Raffaella Di Cagno
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine-Pediatric Section, University of Bari Aldo Moro, Ospedale Pediatrico Giovanni XXIII, 70125 Bari, Italy; (F.C.); (R.F.)
| | - Michael Schwarm
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Stefan Pelzer
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Monika Flügel
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Bodo Speckmann
- Evonik Operations GmbH, 63457 Hanau-Wolfgang, Germany; (M.S.); (S.P.); (M.F.); (B.S.)
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine-Pediatric Section, University of Bari Aldo Moro, Ospedale Pediatrico Giovanni XXIII, 70125 Bari, Italy; (F.C.); (R.F.)
| | - Marco Gobbetti
- Faculty of Science and Technology, Free University of Bozen, 39100 Bolzano, Italy;
| |
Collapse
|
81
|
Kivelä L, Caminero A, Leffler DA, Pinto-Sanchez MI, Tye-Din JA, Lindfors K. Current and emerging therapies for coeliac disease. Nat Rev Gastroenterol Hepatol 2021; 18:181-195. [PMID: 33219355 DOI: 10.1038/s41575-020-00378-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Coeliac disease is a common enteropathy that occurs in genetically susceptible individuals in response to the ingestion of gluten proteins present in wheat, rye and barley. Currently, the only available treatment for the condition is a strict, life-long gluten-free diet that, despite being safe and often effective, is associated with several challenges. Due to the high cost, particularly restrictive nature and perception of decreased quality of life associated with the diet, some patients are continuously exposed to gluten, which prevents an adequate disease control. Moreover, a subgroup of patients does not respond to the diet adequately, and healing of the small-bowel mucosa can be incomplete. Thus, there is a need for alternative treatment forms. The increasingly understood pathogenetic process of coeliac disease has enabled the identification of various targets for future therapies. Multiple investigational therapies ranging from tolerogenic to immunological approaches are in the pipeline, and several drug candidates have entered phase II/III clinical trials. This Review gives a broad overview of the different investigative treatment modalities for coeliac disease and summarizes the latest advances in this field.
Collapse
Affiliation(s)
- Laura Kivelä
- Tampere Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland.,Children's Hospital and Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Daniel A Leffler
- Harvard Celiac Disease Research Program, Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA, USA.,Takeda Pharmaceuticals, Cambridge, MA, USA
| | - Maria Ines Pinto-Sanchez
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jason A Tye-Din
- Immunology Division, The Walter and Eliza Hall Institute, Parkville, and Gastroenterology Department, The Royal Melbourne Hospital, Parkville, Australia
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
82
|
Cristofori F, Dargenio VN, Dargenio C, Miniello VL, Barone M, Francavilla R. Anti-Inflammatory and Immunomodulatory Effects of Probiotics in Gut Inflammation: A Door to the Body. Front Immunol 2021; 12:578386. [PMID: 33717063 PMCID: PMC7953067 DOI: 10.3389/fimmu.2021.578386] [Citation(s) in RCA: 438] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/14/2021] [Indexed: 02/05/2023] Open
Abstract
Hosting millions of microorganisms, the digestive tract is the primary and most important part of bacterial colonization. On one side, in cases of opportunistic invasion, the abundant bacterial population inside intestinal tissues may face potential health problems such as inflammation and infections. Therefore, the immune system has evolved to sustain the host-microbiota symbiotic relationship. On the other hand, to maintain host immune homeostasis, the intestinal microflora often exerts an immunoregulatory function that cannot be ignored. A field of great interest is the association of either microbiota or probiotics with the immune system concerning clinical uses. This microbial community regulates some of the host's metabolic and physiological functions and drives early-life immune system maturation, contributing to their homeostasis throughout life. Changes in gut microbiota can occur through modification in function, composition (dysbiosis), or microbiota-host interplays. Studies on animals and humans show that probiotics can have a pivotal effect on the modulation of immune and inflammatory mechanisms; however, the precise mechanisms have not yet been well defined. Diet, age, BMI (body mass index), medications, and stress may confound the benefits of probiotic intake. In addition to host gut functions (permeability and physiology), all these agents have profound implications for the gut microbiome composition. The use of probiotics could improve the gut microbial population, increase mucus-secretion, and prevent the destruction of tight junction proteins by decreasing the number of lipopolysaccharides (LPSs). When LPS binds endothelial cells to toll-like receptors (TLR 2, 4), dendritic cells and macrophage cells are activated, and inflammatory markers are increased. Furthermore, a decrease in gut dysbiosis and intestinal leakage after probiotic therapy may minimize the development of inflammatory biomarkers and blunt unnecessary activation of the immune system. In turn, probiotics improve the differentiation of T-cells against Th2 and development of Th2 cytokines such as IL-4 and IL-10. The present narrative review explores the interactions between gut microflora/probiotics and the immune system starting from the general perspective of a biological plausibility to get to the in vitro and in vivo demonstrations of a probiotic-based approach up to the possible uses for novel therapeutic strategies.
Collapse
Affiliation(s)
- Fernanda Cristofori
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vanessa Nadia Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Costantino Dargenio
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Vito Leonardo Miniello
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Michele Barone
- Gastroenterology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Ruggiero Francavilla
- Department of Biomedical Science and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
83
|
Gupta AK, Maity C. Efficacy and safety of Bacillus coagulans LBSC in irritable bowel syndrome: A prospective, interventional, randomized, double-blind, placebo-controlled clinical study [CONSORT Compliant]. Medicine (Baltimore) 2021; 100:e23641. [PMID: 33545934 PMCID: PMC7837859 DOI: 10.1097/md.0000000000023641] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
GOALS To evaluate safety and efficacy of Bacillus coagulans LBSC [DSM17654] in irritable bowel syndrome (IBS) through a prospective, interventional, randomized, double-blind, and placebo-controlled, CONSORT compliant clinical trial. BACKGROUND Bacteriotherapy shows promising impact on alleviating clinical conditions of IBS and associated functional gastrointestinal disorders. B coagulans LBSC is a genetically and phenotypically safe probiotic strain used in this study to study its impact on ameliorating IBS symptoms and improving quality of life. METHODS In this interventional, randomized, double-blind, placebo-controlled clinical study, total 40 subjects (18-65 years) were screened through Rome IV criteria and randomized into 2 groups, that is, interventional and placebo arm (n = 20/arm). Similar dosages were received by both the arm, that is, placebo (vehicle) and interventional arm (B coagulans LBSC, 6 billion/d) for a period of 80 days. Study completed with per protocol subjects (n = 38) and results were considered to evaluate the primary and secondary endpoints. RESULTS Assessment through Digestive Symptom Frequency Questionnaire 5 point Likert scale showed significant improvement in interventional arm compared to placebo on symptoms such as bloating/cramping, abdominal pain, diarrhea, constipation, stomach rumbling, nausea, vomiting, headache, and anxiety. Maximum of "no symptoms" cases and mild to moderate gastrointestinal symptoms along with improved stool consistency were from interventional arm tested following IBS severity scoring system and Bristol stool form scale. Upper gastrointestinal endoscopy revealed no clinical difference of gastrointestinal mucosa between both the arms. B coagulans LBSC was well tolerated with no serious adverse events. CONCLUSIONS B coagulans LBSC was safe for human consumption and efficacious in alleviating overall pathophysiological symptoms of IBS and thereby improving inclusive quality of life evaluated.
Collapse
|
84
|
Kaur J, Singh BP, Chaudhary V, Elshaghabee FMF, Singh J, Singh A, Rokana N, Panwar H. Probiotics as Live Bio-therapeutics: Prospects and Perspectives. MICROORGANISMS FOR SUSTAINABILITY 2021:83-120. [DOI: 10.1007/978-981-15-6795-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
85
|
Caminero A, Verdu E. Therapeutic targets based on the modulation of immune function by gut bacteria. BIOTECHNOLOGICAL STRATEGIES FOR THE TREATMENT OF GLUTEN INTOLERANCE 2021:221-237. [DOI: 10.1016/b978-0-12-821594-4.00004-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
86
|
Bio-synthesis of silver nanoparticles with the brackish water blue-green alga Oscillatoria princeps and antibacterial assessment. APPLIED NANOSCIENCE 2020. [DOI: 10.1007/s13204-020-01593-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
87
|
Hashimoto Y, Nakajima H, Hata S, Miyoshi T, Hosomi Y, Majima S, Nakanishi N, Senmaru T, Osaka T, Okada H, Ushigome E, Hamaguchi M, Asano M, Yamazaki M, Fukui M. Effect of probiotics, Bifidobacterium bifidum G9-1, on gastrointestinal symptoms in patients with type 2 diabetes mellitus: study protocol for open-label, single-arm, exploratory research trial (Big STAR study). J Clin Biochem Nutr 2020; 67:223-227. [PMID: 33293761 PMCID: PMC7705076 DOI: 10.3164/jcbn.20-100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 12/12/2022] Open
Abstract
Metformin is associated with risks of gastrointestinal complications in patients with type 2 diabetes. In contrast, probiotic Bifidobacterium bifidum G9-1 (BBG9-1) could improve the symptoms of diarrhea caused by metformin in animal models. Thus, the primary outcome of this study will be the effect of the probiotic BBG9-1 on gastrointestinal symptoms, including diarrhea, in patients with type 2 diabetes who use metformin. This open-label, single-arm, and exploratory study will examine 40 patients with type 2 diabetes who use metformin and have symptoms of constipation or diarrhea. After the baseline examination (objective 1), patients will be administered probiotic BBG9-1 for 10 ± 2 weeks. Then, examinations will be performed (objective 2). The primary outcome will be changes in the symptoms of constipation or diarrhea from objective 1 to objective 2. Secondary outcomes will include changes in gut microbiota, and correlations between changes in fecal properties and biomarkers, including HbA1c level and body mass index. This is the first study to investigate the effect of probiotic BBG9-1 on the change in the symptom of constipation or diarrhea in patients with type 2 diabetes who use metformin.
Collapse
Affiliation(s)
- Yoshitaka Hashimoto
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hanako Nakajima
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shinnosuke Hata
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tomoki Miyoshi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yukako Hosomi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Saori Majima
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Naoko Nakanishi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takafumi Senmaru
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takafumi Osaka
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hiroshi Okada
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Emi Ushigome
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Mai Asano
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahiro Yamazaki
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
88
|
Akobeng AK, Singh P, Kumar M, Al Khodor S. Role of the gut microbiota in the pathogenesis of coeliac disease and potential therapeutic implications. Eur J Nutr 2020; 59:3369-3390. [PMID: 32651763 PMCID: PMC7669811 DOI: 10.1007/s00394-020-02324-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 07/01/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Although genetic predisposition and exposure to dietary gluten are considered necessary triggers for the development of coeliac disease, alterations in the gut microbial composition may also contribute towards the pathogenesis of coeliac disease. This review aims to provide an overview of the available data on the potential mechanisms through which the gut microbiota plays a role in the causation of coeliac disease and to discuss the potential therapeutic strategies that could diminish the consequences of microbial dysbiosis. METHOD A search of the literature was performed using the PubMed, Embase, and JSTOR databases; relevant articles were included. RESULTS Recent studies in patients with coeliac disease have reported an increase in the relative amounts of gram negative bacterial genera such as Bacteroides, Prevotella, and Escherichia, and reduced amounts of protective anti-inflammatory bacteria such as Bifidobacteria and Lactobacilli. Dysbiotic microbiota may lead to a dysregulated immune response that may contribute to the pathogenesis of coeliac disease. In infancy, antibiotic use and certain infant feeding practices may lead to alterations in the developing gut microbiota to influence the immune maturation process and predispose to coeliac disease. CONCLUSION The induction of the intestinal immune system and gluten intolerance may be influenced by the relative abundance of certain microbiota. Factors such as infant feeding practices, diet, antibiotics, and infections, may be involved in the development of coeliac disease due to their influence on gut microbial composition. The efficacy of potential modulators of the gut microbiota such as probiotics, prebiotics, and fecal microbial transplant as adjunctive treatments to gluten-free diet in coeliac disease is unproven and requires further investigation.
Collapse
Affiliation(s)
- Anthony K Akobeng
- Division of Gastroenterology, Hepatology, and Nutrition, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Cornell University, Doha, Qatar
| | - Parul Singh
- Research Department, Sidra Medicine, Doha, Qatar
| | - Manoj Kumar
- Research Department, Sidra Medicine, Doha, Qatar
| | | |
Collapse
|
89
|
Cristofori F, Francavilla R, Capobianco D, Dargenio VN, Filardo S, Mastromarino P. Bacterial-Based Strategies to Hydrolyze Gluten Peptides and Protect Intestinal Mucosa. Front Immunol 2020; 11:567801. [PMID: 33224137 PMCID: PMC7669986 DOI: 10.3389/fimmu.2020.567801] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/29/2020] [Indexed: 12/23/2022] Open
Abstract
Gluten is a mixture of proteins highly resistant to hydrolysis, resulting in the emergence of toxic peptides responsible for gluten-related disorders. Currently, a gluten-free diet (GFD) is the unique proven therapy for celiac disease (CD). Several research groups and pharmaceutical companies are developing new nondietetic therapeutic strategies for CD. Probiotics are viable microorganisms thought to have a healthy effect on the host. The proteolytic mechanism of lactic acid bacteria comprises an extracellular serine protease, di- and oligopeptide-specific transport systems, and several intracellular peptidases that might affect gluten degradation. Therefore, probiotic supplementation is an attractive therapy because of its possible anti-inflammatory and immunomodulatory properties. Several studies have been performed to assess the effectiveness of various specific probiotic strains, showing positive effects on immune-modulation (inhibition of pro-inflammatory cytokine TNF-α) restoring gut microbiota and decrease of immunogenic peptides. The present review aims to summarize the current knowledge on the ability of probiotic strain (single or mixtures) to digest gliadin peptides in vitro and to modulate the inflammatory response in the gut.
Collapse
Affiliation(s)
- Fernanda Cristofori
- Interdisciplinary Department of Medicine—Pediatric Section, Università di Bari Aldo Moro, Bari, Italy
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine—Pediatric Section, Università di Bari Aldo Moro, Bari, Italy
| | - Daniela Capobianco
- Department of Public Health and Infectious Disease, Università La Sapienza di Roma, Rome, Italy
| | - Vanessa Nadia Dargenio
- Interdisciplinary Department of Medicine—Pediatric Section, Università di Bari Aldo Moro, Bari, Italy
| | - Simone Filardo
- Department of Public Health and Infectious Disease, Università La Sapienza di Roma, Rome, Italy
| | - Paola Mastromarino
- Department of Public Health and Infectious Disease, Università La Sapienza di Roma, Rome, Italy
| |
Collapse
|
90
|
Rej A, Aziz I, Sanders DS. Coeliac disease and noncoeliac wheat or gluten sensitivity. J Intern Med 2020; 288:537-549. [PMID: 32573000 DOI: 10.1111/joim.13120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 12/15/2022]
Abstract
Coeliac disease (CD) and noncoeliac wheat or gluten sensitivity (NCWS/NCGS) are common gluten-related disorders. Both conditions can present with gastrointestinal and extraintestinal manifestations, which can be a challenge for physicians to discern between. Whilst coeliac serology and histological assessment are required for the diagnosis of CD, there are no clear biomarkers for the diagnosis of NCGS. The management of both conditions is with a gluten-free diet (GFD), although the duration, as well as strictness of adherence to a GFD in NCGS, is unclear. Adherence to a GFD in CD can also be challenging, with recent developments of noninvasive assessments, although histological assessment via duodenal biopsies remains the gold standard. The management of refractory coeliac disease remains particularly challenging, often requiring specialist input. Whilst wheat is noted to be a trigger for symptom generation in NCGS, it is unclear which components of wheat are responsible for symptom generation in this group, with further research required to elucidate the pathophysiology.
Collapse
Affiliation(s)
- A Rej
- From the, Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - I Aziz
- From the, Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK.,Department of Infection, Immunity and Cardiovascular Disease, Academic Unit of Gastroenterology, University of Sheffield, Sheffield, UK
| | - D S Sanders
- From the, Academic Unit of Gastroenterology, Royal Hallamshire Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK.,Department of Infection, Immunity and Cardiovascular Disease, Academic Unit of Gastroenterology, University of Sheffield, Sheffield, UK
| |
Collapse
|
91
|
Marietta E, Mangalam AK, Taneja V, Murray JA. Intestinal Dysbiosis in, and Enteral Bacterial Therapies for, Systemic Autoimmune Diseases. Front Immunol 2020; 11:573079. [PMID: 33193357 PMCID: PMC7655733 DOI: 10.3389/fimmu.2020.573079] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
Recent studies have shown that a number of common autoimmune diseases have perturbations of their intestinal microbiome (dysbiosis). These include: Celiac Disease (CeD), Multiple Sclerosis (MS), Rheumatoid Arthritis (RA), Sjogren’s Syndrome (SS), and Type 1 diabetes (T1D). All of these have intestinal microbiomes that are different from healthy controls. There have been numerous studies using animal models of single probiotics (monoclonal) or mixtures of probiotics (polyclonal) and even complete microbiota transfer (fecal microbial transfer-FMT) to inhibit or delay the onset of autoimmune diseases such as the aforementioned common ones. However, proportionally, fewer clinical trials have utilized monoclonal therapies or FMT than polyclonal therapies for treating autoimmune diseases, even though bacterial mono-therapies do inhibit the development of autoimmune diseases and/or delay the onset of autoimmune diseases in rodent models of those autoimmune diseases. In this review then, we review the previously completed and currently ongoing clinical trials that are testing bacterial therapies (FMT, monoclonal, and polyclonal) to treat common autoimmune dseases and discuss the successes in using bacterial monotherapies to treat rodent models of these common autoimmune diseases.
Collapse
Affiliation(s)
- Eric Marietta
- Department of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Dermatology, Mayo Clinic Rochester, Rochester, MN, United States
| | | | - Veena Taneja
- Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States
| | - Joseph A Murray
- Department of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, United States.,Department of Immunology, Mayo Clinic Rochester, Rochester, MN, United States
| |
Collapse
|
92
|
Ding JH, Jin Z, Yang XX, Lou J, Shan WX, Hu YX, Du Q, Liao QS, Xie R, Xu JY. Role of gut microbiota via the gut-liver-brain axis in digestive diseases. World J Gastroenterol 2020; 26:6141-6162. [PMID: 33177790 PMCID: PMC7596643 DOI: 10.3748/wjg.v26.i40.6141] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/29/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023] Open
Abstract
The gut-brain axis is a bidirectional information interaction system between the central nervous system (CNS) and the gastrointestinal tract, in which gut microbiota plays a key role. The gut microbiota forms a complex network with the enteric nervous system, the autonomic nervous system, and the neuroendocrine and neuroimmunity of the CNS, which is called the microbiota-gut-brain axis. Due to the close anatomical and functional interaction of the gut-liver axis, the microbiota-gut-liver-brain axis has attracted increased attention in recent years. The microbiota-gut-liver-brain axis mediates the occurrence and development of many diseases, and it offers a direction for the research of disease treatment. In this review, we mainly discuss the role of the gut microbiota in the irritable bowel syndrome, inflammatory bowel disease, functional dyspepsia, non-alcoholic fatty liver disease, alcoholic liver disease, cirrhosis and hepatic encephalopathy via the gut-liver-brain axis, and the focus is to clarify the potential mechanisms and treatment of digestive diseases based on the further understanding of the microbiota-gut- liver-brain axis.
Collapse
Affiliation(s)
- Jian-Hong Ding
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Zhe Jin
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Xiao-Xu Yang
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Jun Lou
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Wei-Xi Shan
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Yan-Xia Hu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Qian Du
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Qiu-Shi Liao
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Rui Xie
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| | - Jing-Yu Xu
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou Province, China
| |
Collapse
|
93
|
Smecuol E, Constante M, Temprano MP, Costa AF, Moreno ML, Pinto-Sanchez MI, Vázquez H, Stefanolo JP, Gonzalez AF, D'Adamo CR, Niveloni SI, Mauriño E, Verdu EF, Bai JC. Effect of Bifidobacterium infantis NLS super strain in symptomatic coeliac disease patients on long-term gluten-free diet - an exploratory study. Benef Microbes 2020; 11:527-534. [PMID: 33032471 DOI: 10.3920/bm2020.0016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bifidobacterium infantis NLS super strain (B. infantis NLS-SS) was previously shown to alleviate gastrointestinal symptoms in newly diagnosed coeliac disease (CD) patients consuming gluten. A high proportion of patients following a gluten-free diet experiences symptoms despite dietary compliance. The role of B. infantis in persistently symptomatic CD patients has not been explored. The aim of the study was to evaluate the effect of B. infantis NLS-SS on persistent gastrointestinal symptoms in patients with CD following a long-term GFD. We conducted a randomised, cross-over, double-blind, placebo-controlled trial in symptomatic adult CD patients on a GFD for at least two years. After one-week run-in, patients were randomised to B. infantis NLS-SS or placebo for 3 weeks with cross-over after a 2-week wash-out period. We estimated changes (Δ) in celiac symptom index (CSI) before and after treatment. Stool samples were collected for faecal microbiota analysis (16S rRNA sequencing). Gluten immunogenic peptide (GIP) excretion in stool and urine samples was measured at each study period. Eighteen patients were enrolled; six patients were excluded due violations in protocol. For patients with the highest clinical burden, CD symptoms were lower in probiotic than in placebo treatment (P=0.046). B. infantis and placebo treated groups had different microbiota profiles as assessed by beta diversity clustering. In probiotic treated groups, we observed an increase in abundance of B. infantis. Treatment with B. infantis was associated with decreased abundance of Ruminococcus sp. and Bifidobacterium adolescentis. GIP excretion in stools and urine was similar at each treatment period. There were no differences in adverse effects between the two groups. B. infantis NLS-SS improves specific CD symptoms in a subset of highly symptomatic treated patients (GFD). This is associated with a shift in stool microbiota profile. Larger studies are needed to confirm these findings. ClinicalTrials.gov: NCT03271138.
Collapse
Affiliation(s)
- E Smecuol
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - M Constante
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - M P Temprano
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - A F Costa
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - M L Moreno
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - M I Pinto-Sanchez
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - H Vázquez
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - J P Stefanolo
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - A F Gonzalez
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - C R D'Adamo
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - S I Niveloni
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - E Mauriño
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina
| | - E F Verdu
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - J C Bai
- Dr. C. Bonorino Udaondo Gastroenterology Hospital, Av. Caseros 2061, 1264 Buenos Aires, Argentina.,Research Institutes, School of Medicine; Universidad del Salvador, Buenos Aires, Argentina
| |
Collapse
|
94
|
Meng X, Zhang G, Cao H, Yu D, Fang X, de Vos WM, Wu H. Gut dysbacteriosis and intestinal disease: mechanism and treatment. J Appl Microbiol 2020; 129:787-805. [PMID: 32277534 PMCID: PMC11027427 DOI: 10.1111/jam.14661] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/14/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome functions like an endocrine organ, generating bioactive metabolites, enzymes or small molecules that can impact host physiology. Gut dysbacteriosis is associated with many intestinal diseases including (but not limited to) inflammatory bowel disease, primary sclerosing cholangitis-IBD, irritable bowel syndrome, chronic constipation, osmotic diarrhoea and colorectal cancer. The potential pathogenic mechanism of gut dysbacteriosis associated with intestinal diseases includes the alteration of composition of gut microbiota as well as the gut microbiota-derived signalling molecules. The many correlations between the latter and the susceptibility for intestinal diseases has placed a spotlight on the gut microbiome as a potential novel target for therapeutics. Currently, faecal microbial transplantation, dietary interventions, use of probiotics, prebiotics and drugs are the major therapeutic tools utilized to impact dysbacteriosis and associated intestinal diseases. In this review, we systematically summarized the role of intestinal microbiome in the occurrence and development of intestinal diseases. The potential mechanism of the complex interplay between gut dysbacteriosis and intestinal diseases, and the treatment methods are also highlighted.
Collapse
Affiliation(s)
- X Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - G Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - H Cao
- InnovHope Inc, Framingham, MA, USA
| | - D Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - X Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - W M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - H Wu
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
95
|
Probiotics for Celiac Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Am J Gastroenterol 2020; 115:1584-1595. [PMID: 32740074 DOI: 10.14309/ajg.0000000000000749] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Many patients with celiac disease (CD) experience persistent symptoms despite adhering to the gluten-free diet. Different studies have assessed the use of probiotics as an adjuvant treatment for CD. We performed a systematic review and meta-analysis to evaluate the efficacy of probiotics in improving gastrointestinal (GI) symptoms and quality of life (QOL) in patients with CD. METHODS We searched EMBASE, MEDLINE, CINAHL, Web of Science, CENTRAL, and DARE databases up to February 2019 for randomized controlled trials (RCTs) evaluating probiotics compared with placebo for treating CD. We collected data on GI symptoms, QOL, adverse events, serum tumor necrosis factor-α, intestinal permeability, and microbiota composition. RESULTS We screened 2,831 records and found that 7 articles describing 6 RCTs (n = 279 participants) were eligible for quantitative analysis. Probiotics improved GI symptoms when assessed by the GI Symptoms Rating Scale (mean difference symptom reduction: -28.7%; 95% confidence interval [CI] -43.96 to -13.52; P = 0.0002). There was no difference in GI symptoms after probiotics when different questionnaires were pooled. The levels of Bifidobacteria increased after probiotics (mean difference: 0.85 log colony-forming units (CFU) per gram; 95% CI 0.38-1.32 log CFU per gram; P = 0.0003). There were insufficient data on tumor necrosis factor-a levels or QOL for probiotics compared with placebo. No difference in adverse events was observed between probiotics and placebo. The overall certainty of the evidence ranged from very low to low. DISCUSSION Probiotics may improve GI symptoms in patients with CD. High-quality clinical trials are needed to improve the certainty in the evidence (see Visual abstract, Supplementary Digital Content 2, http://links.lww.com/AJG/B595).
Collapse
|
96
|
Marasco G, Cirota GG, Rossini B, Lungaro L, Di Biase AR, Colecchia A, Volta U, De Giorgio R, Festi D, Caio G. Probiotics, Prebiotics and Other Dietary Supplements for Gut Microbiota Modulation in Celiac Disease Patients. Nutrients 2020; 12:2674. [PMID: 32887325 PMCID: PMC7551848 DOI: 10.3390/nu12092674] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
To date, the only available treatment for celiac disease (CD) patients is a life-lasting gluten-free diet (GFD). Lack of adherence to the GFD leads to a significant risk of adverse health consequences. Food cross-contamination, nutritional imbalances, and persistent gastrointestinal symptoms are the main concerns related to GFD. Moreover, despite rigid compliance to GFD, patients struggle in achieving a full restoring of the gut microbiota, which plays a role in the nutritive compounds processing, and absorption. Pivotal studies on the supplementation of GFD with probiotics, such as Bifidobacterium and Lactobacilli, reported a potential to restore gut microbiota composition and to pre-digest gluten in the intestinal lumen, reducing the inflammation associated with gluten intake, the intestinal permeability, and the cytokine and antibody production. These findings could explain an improvement in symptoms and quality of life in patients treated with GFD and probiotics. On the other hand, the inclusion of prebiotics in GFD could also be easy to administer and cost-effective as an adjunctive treatment for CD, having the power to stimulate the growth of potentially health-promoting bacteria strains. However, evidence regarding the use of prebiotics and probiotics in patients with CD is still insufficient to justify their use in clinical practice.
Collapse
Affiliation(s)
- Giovanni Marasco
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.G.C.); (B.R.); (U.V.); (D.F.)
| | - Giovanna Grazia Cirota
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.G.C.); (B.R.); (U.V.); (D.F.)
| | - Benedetta Rossini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.G.C.); (B.R.); (U.V.); (D.F.)
| | - Lisa Lungaro
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (R.D.G.)
| | - Anna Rita Di Biase
- Department of Pediatrics, Policlinic Hospital, University of Modena, Via del Pozzo 71, 41126 Modena, Italy;
| | - Antonio Colecchia
- Gastroenterology Unit, Borgo Trento University Hospital of Verona, P.le Aristide Stefani 1, 37126 Verona, Italy;
| | - Umberto Volta
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.G.C.); (B.R.); (U.V.); (D.F.)
| | - Roberto De Giorgio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (R.D.G.)
| | - Davide Festi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy; (G.G.C.); (B.R.); (U.V.); (D.F.)
| | - Giacomo Caio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44124 Ferrara, Italy; (L.L.); (R.D.G.)
- Celiac Center and Mucosal Immunology and Biology Research, Massachusetts General Hospital-Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
97
|
|
98
|
Abstract
PURPOSE OF REVIEW This review summarizes recent progress in the diagnosis and management of irritable bowel syndrome, with a focus on dietary and microbiota aspects. RECENT FINDINGS From a pathophysiological point of view, IBS is a multifactorial condition with both peripheral (transit) as central (visceral hypersensitivity, anxiety, depression) contribution in a cumulative fashion to the symptom pattern and severity. More recently, the focus has shifted to diet and microbiota. The number of dietary options that can be used for IBS and the understanding of determinants of their efficacy is rapidly increasing. Several studies have confirmed the efficacy of the low fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAP) diet. Sucrose-isomaltase deficiency has emerged as pathogenetic mechanisms in a subset of patients, who do not respond to low FODMAP diet but may respond to starch and sucrose elimination. Herbal remedies, probiotics and secretagogues have been the topic of additional treatment trials. The efficacy of fecal microbiota transplantation in IBS is variable across studies, but donor selection is emerging as a critical factor. SUMMARY Irritable bowel syndrome has evolved into a disorder of interaction between dietary factors and gut microbiota, with impact on bowel symptoms as well as extra-intestinal, central, symptoms. Dietary adjustments and treatments targeting the gut microbiota are areas of active research and clinical progress.
Collapse
|
99
|
Cheng FW, Handu D. Nutrition Assessment, Interventions, and Monitoring for Patients with Celiac Disease: An Evidence Analysis Center Scoping Review. J Acad Nutr Diet 2020; 120:1381-1406. [PMID: 31953154 DOI: 10.1016/j.jand.2019.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Indexed: 11/21/2022]
Abstract
The objectives of this scoping review were to identify and characterize studies examining nutrition assessment, interventions, and measures to monitor gluten-free diet (GFD) adherence/compliance in patients with celiac disease (CD). An electronic literature search of four databases (Cochrane Database for systematic reviews, CINAHL, Embase, and Ovid MEDLINE) was conducted to identify articles examining nutrition care in CD individuals. Except for narrative review, grey literature, and case study/report, all types of peer-reviewed articles published between January 2007 and August 2018 were eligible. There were a total of 10,823 records; 10,368 were excluded during the first round of screening due to irrelevancy and/or duplication. Of the 455 full-text articles that were assessed, 292 met the criteria and were included. Most of the studies were observational studies (n=212), followed by experimental trials (n=50), evidence-based practice guideline (EBPG)/report/statement (n=16), and systematic review (SR) (n=14). Nine original studies examined assessment, focusing mainly on different tools/ways to assess GFD adherence. The majority of the included original articles (n=235) were in the nutrition intervention category with GFD, oats, and prebiotics/probiotics as the top-three most studied interventions. There were eight SRs on GFD and five on oats. One SR and 21 original studies investigated the effectiveness of different measures to monitor GFD adherence/compliance. Although recent CD EBPGs were identified, different methods with varying levels of rigor, in terms of literature search and assessment of evidence strength, were used. Based on this scoping review, interventions focused on gluten-free diet and oats have been significantly covered by either SRs or EBPGs. Studies related to prebiotics/probiotics and education program/counseling focused interventions, as well as assessment, in CD patients have increased in recent years. Thus, it might be beneficial to conduct SRs/EBPGs focused on these topics to guide practitioners.
Collapse
|
100
|
De Filippis F, Pasolli E, Ercolini D. The food-gut axis: lactic acid bacteria and their link to food, the gut microbiome and human health. FEMS Microbiol Rev 2020; 44:454-489. [PMID: 32556166 PMCID: PMC7391071 DOI: 10.1093/femsre/fuaa015] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Lactic acid bacteria (LAB) are present in foods, the environment and the animal gut, although fermented foods (FFs) are recognized as the primary niche of LAB activity. Several LAB strains have been studied for their health-promoting properties and are employed as probiotics. FFs are recognized for their potential beneficial effects, which we review in this article. They are also an important source of LAB, which are ingested daily upon FF consumption. In this review, we describe the diversity of LAB and their occurrence in food as well as the gut microbiome. We discuss the opportunities to study LAB diversity and functional properties by considering the availability of both genomic and metagenomic data in public repositories, as well as the different latest computational tools for data analysis. In addition, we discuss the role of LAB as potential probiotics by reporting the prevalence of key genomic features in public genomes and by surveying the outcomes of LAB use in clinical trials involving human subjects. Finally, we highlight the need for further studies aimed at improving our knowledge of the link between LAB-fermented foods and the human gut from the perspective of health promotion.
Collapse
Affiliation(s)
- Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, via Università, 100, 80055, Portici (NA)Italy
- Task Force on Microbiome Studies, Corso Umberto I, 40, 80100, Napoli, Italy
| |
Collapse
|