1
|
Akerele CA, Koralnik LR, Lafont E, Gilman C, Walsh-Messinger J, Malaspina D. Nutrition and brain health: Implications of Mediterranean diet elements for psychiatric disorders. Schizophr Res 2025; 281:30-44. [PMID: 40315757 DOI: 10.1016/j.schres.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 04/03/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
The Mediterranean diet is an anti-inflammatory diet now recognized for prevention and intervention against cardiometabolic disorders, although emerging literature also shows its benefits for mental health. This paper surveys literature pertinent to the Mediterranean diet with respect to schizophrenia, bipolar and unipolar depression and cognition. The National Library of Medicine database of literature was searched for publications relating to our topic through June 2024. Our results highlight the Mediterranean diet's potential role in mediating inflammation, potentially through the gut-brain axis, and its neuroprotective role against cognitive decline. Moreover, individual components of the Mediterranean diet are potentially therapeutic as well as protective, particularly fruits, vegetables, fatty fish, and whole grains. These dietary components reduce systemic inflammation, modulate gut microbiota and influence critical pathways such as moderating oxidative stress. Overall, this survey of recent literature highlights the potential of the Mediterranean diet to promote mental health and reduce the risk or severity of psychiatric and neurodegenerative disorders. The analysis underscores broader implications for the Mediterranean diet in advancing mental health outcomes and emphasizes the need for further studies and increased public education to encourage healthier eating habits and dietary interventions.
Collapse
Affiliation(s)
| | | | - Ezequiel Lafont
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA
| | - Caitlin Gilman
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Dolores Malaspina
- Icahn School of Medicine at Mount Sinai, Department of Psychiatry, New York, NY 10029, USA.
| |
Collapse
|
2
|
Zhao J, Guo Y, Jiang Q, Lan H, Hung WL, Lynch B. Bifidobacterium longum subsp. infantis YLGB-1496-Toxicological evaluation. J Appl Toxicol 2025; 45:230-244. [PMID: 39252460 DOI: 10.1002/jat.4688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/11/2024]
Abstract
Bifidobacterium infantis YLGB-1496, originally isolated from breast milk from a Taiwanese mother, is under study for use as a probiotic. As part of safety assessment, an Ames, in vivo mouse micronucleus, and in vivo mouse spermatocyte chromosome aberration assay were conducted along with a 13-week oral rat toxicity study. B. infantis YLGB-1496 had no activity in any of the genotoxicity assays. Administration of the bacteria to Sprague-Dawley rats at doses ranging from 0 to 1.5 g/kg bw/day had no treatment-related effects on any of the endpoints measured. There appear to be no concerns for translocation or pathogenicity of B. infantis YLGB-1496 based on extensive experience with the species in general. The results of the current investigations support potential use of B. infantis YLGB-1496 as a probiotic in infant formula.
Collapse
Affiliation(s)
- Jian Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yueyi Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qiuyue Jiang
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Barry Lynch
- Intertek Health Sciences Inc., Mississauga, ON, Canada
| |
Collapse
|
3
|
Mirfeizi Z, Mahmoudi M, Jokar MH, Sahebari M, Noori E, Mehrad-Majd H, Barati M, Faridzadeh A. Impact of synbiotics on disease activity in systemic lupus erythematosus: Results from a randomized clinical trial. J Food Sci 2024; 89:9835-9845. [PMID: 39437223 DOI: 10.1111/1750-3841.17460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that affects various organs in the body. In SLE, inflammatory cytokines play a crucial role in initiating and sustaining the inflammatory process. Synbiotics may help modulate these inflammatory cytokines. This randomized, double-blind, placebo-controlled clinical trial aimed to assess the impact of synbiotics intervention on interleukin-17A (IL-17A) levels, disease activity, and inflammatory factors in patients with SLE. Fifty SLE patients were randomly assigned to receive either standard therapy plus synbiotics (consisting of Streptococcus thermophilus, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus rhamnosus, Lactobacillus salivarius, Lactobacillus reuteri, Bifidobacterium lactis, Bifidobacterium longum, Bifidobacterium bifidum, and the prebiotic fructooligosaccharides) or standard therapy alone for 2 months. The results demonstrated a significant reduction in both protein and mRNA levels of IL-17A, as well as in the Systemic Lupus Erythematosus Disease Activity Index 2000 score, within the synbiotics group after the intervention compared to baseline. In contrast, the placebo group did not experience significant changes in IL-17A levels or disease activity. Synbiotic supplementation shows potential as an adjunctive therapeutic approach for SLE management; however, further research is needed to elucidate its underlying mechanisms. PRACTICAL APPLICATION: This study explores the use of synbiotics as a supplementary treatment for systemic lupus erythematosus, which is typically managed with immunosuppressive therapies.
Collapse
Affiliation(s)
- Zahra Mirfeizi
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Hassan Jokar
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Sahebari
- Rheumatology Department, Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Noori
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hasan Mehrad-Majd
- Clinical Research Development Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Barati
- Department of Laboratory Sciences, School of Paramedicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Arezoo Faridzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Immunology and Allergy, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Brunner JD, Chia N. Metabolic model-based ecological modeling for probiotic design. eLife 2024; 13:e83690. [PMID: 38380900 PMCID: PMC10942782 DOI: 10.7554/elife.83690] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 02/19/2024] [Indexed: 02/22/2024] Open
Abstract
The microbial community composition in the human gut has a profound effect on human health. This observation has lead to extensive use of microbiome therapies, including over-the-counter 'probiotic' treatments intended to alter the composition of the microbiome. Despite so much promise and commercial interest, the factors that contribute to the success or failure of microbiome-targeted treatments remain unclear. We investigate the biotic interactions that lead to successful engraftment of a novel bacterial strain introduced to the microbiome as in probiotic treatments. We use pairwise genome-scale metabolic modeling with a generalized resource allocation constraint to build a network of interactions between taxa that appear in an experimental engraftment study. We create induced sub-graphs using the taxa present in individual samples and assess the likelihood of invader engraftment based on network structure. To do so, we use a generalized Lotka-Volterra model, which we show has strong ability to predict if a particular invader or probiotic will successfully engraft into an individual's microbiome. Furthermore, we show that the mechanistic nature of the model is useful for revealing which microbe-microbe interactions potentially drive engraftment.
Collapse
Affiliation(s)
- James D Brunner
- Biosciences Division, Los Alamos National LaboratoryLos AlamosUnited States
- Center for Nonlinear Studies, Los Alamos National LaboratoryLos AlamosUnited States
| | - Nicholas Chia
- Data Science and Learning, Argonne National LaboratoryLemontUnited States
| |
Collapse
|
5
|
Maftei NM, Raileanu CR, Balta AA, Ambrose L, Boev M, Marin DB, Lisa EL. The Potential Impact of Probiotics on Human Health: An Update on Their Health-Promoting Properties. Microorganisms 2024; 12:234. [PMID: 38399637 PMCID: PMC10891645 DOI: 10.3390/microorganisms12020234] [Citation(s) in RCA: 89] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/25/2024] Open
Abstract
Probiotics, known to be live microorganisms, have been shown to improve or restore the gut microbiota, which in turn has been linked to improved health. It is believed that probiotics are the modern equivalent of a panacea, with claims that they may treat or prevent different diseases both in children and adults (e.g., from colic in babies to cardiovascular disease, respiratory infection, and cancer in adults). Ever since the early 2000s, probiotic-based fermented foods have had a resurgence in popularity, mostly due to claims made regarding their health benefits. Fermented foods have been associated with the prevention of irritable bowel syndrome, lactose intolerance, gastroenteritis, and obesity, but also other conditions such as chronic diarrhea, allergies, dermatitis, and bacterial and viral infections, all of which are closely related to an unhealthy lifestyle. Recent and ongoing developments in microbiome/microbiota science have given us new research directions for probiotics. The new types, mechanisms, and applications studied so far, and those currently under study, have a great potential to change scientific understanding of probiotics' nutritional applications and human health care. The expansion of fields related to the study of the microbiome and the involvement of probiotics in its improvement foreshadow an era of significant changes. An expanding range of candidate probiotic species is emerging that can address newly elucidated data-driven microbial niches and host targets. In the probiotic field, new variants of microbiome-modulating interventions are being developed, including prebiotics, symbiotics, postbiotics, microbial consortia, live biotherapeutic products, and genetically modified organisms, with renewed interest in polyphenols, fibers, and fermented foods to ensure human health. This manuscript aims to analyze recent, emerging, and anticipated trends in probiotics (sources, doses, mechanism of action, diseases for which probiotics are administered, side effects, and risks) and create a vision for the development of related areas of influence in the field.
Collapse
Affiliation(s)
- Nicoleta-Maricica Maftei
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Clinic Laboratory Department, Clinical Hospital of Children Hospital “Sf. Ioan”, 800487 Galati, Romania
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Cosmin Raducu Raileanu
- Department of Morphological and Functional Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (C.R.R.); (L.A.)
| | - Alexia Anastasia Balta
- Medical Department Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania;
| | - Lenuta Ambrose
- Department of Morphological and Functional Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (C.R.R.); (L.A.)
| | - Monica Boev
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Denisa Batîr Marin
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| | - Elena Lacramioara Lisa
- Department of Pharmaceutical Sciences, Faculty of Medicine, and Pharmacy, “Dunărea de Jos” University, 800010 Galati, Romania; (N.-M.M.); (E.L.L.)
- Research Centre in the Medical-Pharmaceutical Field, “Dunarea de Jos” University of Galati, 800010 Galati, Romania
| |
Collapse
|
6
|
Amieva-Balmori M, García-Mazcorro JF, Martínez-Conejo A, Hernández-Ramírez GA, García-Zermeño KR, Rodríguez-Aguilera O, Aja-Cadena M, Barradas-Cortés M, Quigley EMM, Remes-Troche JM. Fecal bacterial microbiota in constipated patients before and after eight weeks of daily Bifidobacterium infantis 35624 administration. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2023; 88:369-380. [PMID: 35810091 DOI: 10.1016/j.rgmxen.2022.06.014] [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: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION AND AIM In recent years, probiotics have been used in functional gastrointestinal disorders, including chronic constipation (CC). The effect of Bifidobacterium infantis strain 35624 on the gut microbiota of CC patients has not been previously studied. Our aim was to analyze the fecal microbiota of constipated patients, before and after consuming a single-strain probiotic (B. infantis strain 35624). MATERIALS AND METHODS We used 16S rRNA gene high-throughput sequencing to analyze the fecal microbiota of female patients (n=13) with CC. Patients were instructed to ingest one capsule of Alflorex® (containing 1×109 CFUs/g B. infantis strain 35624) daily for eight weeks. Fecal samples were obtained at the baseline and end (final) of probiotic administration. RESULTS Alpha diversity metrics did not differ between the baseline and final periods. The butyrate producer, Oscillospira, was the taxon most strongly correlated with amplicon sequence variants (R2=0.55, p<0.0001). Except for a few bacterial taxa, there were no significant differences in relative abundance between the baseline and final periods. Beta-diversity measures also showed limited evidence for the differences between the two time periods. CONCLUSIONS The results suggest that the fecal bacterial microbiota remains stable in constipated women consuming a single-strain probiotic. Those findings may be helpful in better understanding probiotic functioning in patients with digestive disorders.
Collapse
Affiliation(s)
- M Amieva-Balmori
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - J F García-Mazcorro
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - A Martínez-Conejo
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - G A Hernández-Ramírez
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - K R García-Zermeño
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - O Rodríguez-Aguilera
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - M Aja-Cadena
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - M Barradas-Cortés
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - E M M Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - J M Remes-Troche
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México.
| |
Collapse
|
7
|
Terra I, Santeiro C, da Fonseca Rua J. Supplementation With Bifidobacterium and Symptomatic Control in Irritable Bowel Syndrome: An Evidence-Based Review. Cureus 2023; 15:e44447. [PMID: 37791169 PMCID: PMC10544091 DOI: 10.7759/cureus.44447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/05/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder, with a global prevalence of around 11%. Family doctors should be aware of the diagnosis and treatment of this pathology. The benefit of using probiotics is questionable. The purpose of this review is to establish the evidence of the association between Bifidobacterium supplementation and symptomatic control in patients with IBS. The research was conducted using the National Guideline Clearinghouse, National Electronic Library for Health of the British NHS, Canadian Medical Association Practice Guidelines InfoBase, Cochrane Library, Database of Abstracts of Reviews of Effectiveness, Bandolier, Evidence-Based Medicine Online, and PubMed. Articles published between March 2017 and March 2022 in humans and written in Portuguese, Spanish, and English using the terms IBS and Bifidobacterium were included. To stratify the level of evidence (LOE), the Strength of Recommendation Taxonomy (SORT), from the American Academy of Family Physicians, was used. Thirty-seven articles were found corresponding to the search terms, and a total of seven articles were selected. Three clinical trials and a simple review have demonstrated improvement in symptoms, although further studies are needed. The guideline and the systematic review did not demonstrate superiority in symptomatic relief when compared to other species of probiotics. The meta-analysis did not show the efficacy of the isolated use of Bifidobacterium. The evidence of an association between supplementation with Bifidobacterium and symptomatic control in patients with IBS is not clear. Some studies seem to demonstrate benefits in improving symptoms (SORT C).
Collapse
Affiliation(s)
- Inês Terra
- Family Medicine, Unidades de Saúde Familiar (USF) Casa dos Pescadores, Póvoa de Varzim, PRT
| | - Catarina Santeiro
- Family Medicine, Unidades de Saúde Familiar (USF) Lusitânia, Évora, PRT
| | | |
Collapse
|
8
|
Tsounis EP, Triantos C, Konstantakis C, Marangos M, Assimakopoulos SF. Intestinal barrier dysfunction as a key driver of severe COVID-19. World J Virol 2023; 12:68-90. [PMID: 37033148 PMCID: PMC10075050 DOI: 10.5501/wjv.v12.i2.68] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/08/2022] [Accepted: 01/16/2023] [Indexed: 03/21/2023] Open
Abstract
The intestinal lumen harbors a diverse consortium of microorganisms that participate in reciprocal crosstalk with intestinal immune cells and with epithelial and endothelial cells, forming a multi-layered barrier that enables the efficient absorption of nutrients without an excessive influx of pathogens. Despite being a lung-centered disease, severe coronavirus disease 2019 (COVID-19) affects multiple systems, including the gastrointestinal tract and the pertinent gut barrier function. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can inflict either direct cytopathic injury to intestinal epithelial and endothelial cells or indirect immune-mediated damage. Alternatively, SARS-CoV-2 undermines the structural integrity of the barrier by modifying the expression of tight junction proteins. In addition, SARS-CoV-2 induces profound alterations to the intestinal microflora at phylogenetic and metabolomic levels (dysbiosis) that are accompanied by disruption of local immune responses. The ensuing dysregulation of the gut-lung axis impairs the ability of the respiratory immune system to elicit robust and timely responses to restrict viral infection. The intestinal vasculature is vulnerable to SARS-CoV-2-induced endothelial injury, which simultaneously triggers the activation of the innate immune and coagulation systems, a condition referred to as “immunothrombosis” that drives severe thrombotic complications. Finally, increased intestinal permeability allows an aberrant dissemination of bacteria, fungi, and endotoxin into the systemic circulation and contributes, to a certain degree, to the over-exuberant immune responses and hyper-inflammation that dictate the severe form of COVID-19. In this review, we aim to elucidate SARS-CoV-2-mediated effects on gut barrier homeostasis and their implications on the progression of the disease.
Collapse
Affiliation(s)
- Efthymios P Tsounis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Christos Konstantakis
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University Hospital of Patras, Patras 26504, Greece
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| | - Stelios F Assimakopoulos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Patras 26504, Greece
| |
Collapse
|
9
|
Comorbidity of functional bowel disorders and obesity in terms of microbiome. ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.5-2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Bacterial ecosystem of the gut plays a fundamental role in the normal functioning of the metabolic and immune systems. Functional bowel disease and obesity are highly prevalent in the population and place a heavy burden on healthcare system. Both comorbidity and multimorbidity are considered to be common for obesity and intestinal functional disorders. Changes in the microbiota can be both the cause and consequence of each disease: intestinal functional disorder changes the composition of the microbiota, resulting in obesity, and vice versa. Intestinal functional disorders and obesity are characterized by a similar type of dysbiosis.The aim of the review is to analyze the research findings available to date in order to establish the relationship between the gut microbiome, functional bowel disease and obesity. The researches have shown that patients with intestinal functional disorders have a different gut microbiome than healthy individuals. For intestinal functional disorders, the general patterns of the intestinal microbiota composition were described, and the characteristic taxonomic groups of bacteria were identified. On the other side, it must be noted that there is no clear correlation between intestinal functional disorders and obesity in terms of the microbiota. This can be explained by the high heterogeneity of intestinal functional disorders, as well as by the lack of a unified approach to creating a study design, by different sizes of population samples and also by different diagnostic criteria. The necessity to determine the criteria in the development of the design of future studies is discussed.
Collapse
|
10
|
Shin SY, Park S, Moon JM, Kim K, Kim JW, Chun J, Lee TH, Choi CH. Compositional Changes in the Gut Microbiota of Responders and Non-responders to Probiotic Treatment Among Patients With Diarrhea-predominant Irritable Bowel Syndrome: A Post Hoc Analysis of a Randomized Clinical Trial. J Neurogastroenterol Motil 2022; 28:642-654. [PMID: 36250371 PMCID: PMC9577570 DOI: 10.5056/jnm21202] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/08/2022] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Background/Aims We aim to evaluate the differences in the microbiome of responders and non-responders, as well as predict the response to probiotic therapy, based on fecal microbiome data in patients with diarrhea-predominant irritable bowel syndrome (IBS-D). Methods A multi-strain probiotics that contains Lactobacillus acidophilus (KCTC 11906BP), Lactobacillus plantarum (KCTC11867BP), Lactobacillus rhamnosus (KCTC 11868BP), Bifidobacterium breve (KCTC 11858BP), Bifidobacterium lactis (KCTC 11903BP), Bifidobacterium longum (KCTC 11860BP), and Streptococcus thermophilus (KCTC 11870BP) were used. Patients were categorized into probiotic and placebo groups, and fecal samples were collected from all patients before and at the end of 8 weeks of treatment. The probiotic group was further divided into responders and non-responders. Responders were defined as patients who experienced adequate relief of overall irritable bowel syndrome symptoms after probiotic therapy. Fecal microbiota were investigated using Illumina MiSeq and analyzed using the EzBioCloud 16S database and microbiome pipeline (https://www.EZbiocloud.net). Results There was no significant difference in the alpha and beta diversity between the responder and non-responder groups. The abundances of the phylum Proteobacteria and genus Bacteroides significantly decreased after probiotic treatment. Bifidobacterium bifidum, Pediococcus acidilactici, and Enterococcus faecium showed a significantly higher abundance in the probiotic group after treatment compared to the placebo group. Enterococcus faecalis and Lactococcus lactis were identified as biomarkers of non-response to probiotics. The abundance of Fusicatenibacter saccharivorans significantly increased in the responders after treatment. Conclusions Probiotic treatment changes some composition of fecal bacteria in patients with IBS-D. E. faecalis and L. lactis may be prediction biomarkers for non-response to probiotics. Increased abundance of F. sccharivorans is correlated to symptom improvement by probiotics in patients with IBS-D.
Collapse
Affiliation(s)
- Seung Yong Shin
- Chung-Ang University College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Sein Park
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea
| | - Jung Min Moon
- Chung-Ang University College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Kisung Kim
- Chung-Ang University College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Jeong Wook Kim
- Chung-Ang University College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Jongsik Chun
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Korea.,Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Korea.,School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Tae Hee Lee
- Institute for Digestive Research, Digestive Disease Center Soonchunhyang University College of Medicine, Seoul, Korea
| | - Chang Hwan Choi
- Chung-Ang University College of Medicine, Department of Internal Medicine, Seoul, Korea
| | | |
Collapse
|
11
|
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
|
12
|
Bifidobacterium infantis 35624 efficacy in patients with uncontrolled asthma: A randomized placebo-controlled trial. Ann Allergy Asthma Immunol 2022; 129:790-792. [PMID: 36089255 DOI: 10.1016/j.anai.2022.08.1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/20/2022]
|
13
|
Chang C, Yuan X, Zhang X, Chen X, Li K. Gastrointestinal Microbiome and Multiple Health Outcomes: Umbrella Review. Nutrients 2022; 14:3726. [PMID: 36145102 PMCID: PMC9505003 DOI: 10.3390/nu14183726] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, there has been growing concern about the impact of the gastrointestinal microbiome on human health outcomes. To clarify the evidence for a link between the gastrointestinal microbiome and a variety of health outcomes in humans, we conducted an all-encompassing review of meta-analyses and systematic reviews that included 195 meta-analyses containing 950 unique health outcomes. The gastrointestinal microbiome is related to mortality, gastrointestinal disease, immune and metabolic outcomes, neurological and psychiatric outcomes, maternal and infant outcomes, and other outcomes. Existing interventions for intestinal microbiota (such as probiotics, fecal microbiota transplant, etc.) are generally safe and beneficial to a variety of human health outcomes, but the quality of evidence is not high, and more detailed and well-designed randomized controlled trials are necessary.
Collapse
Affiliation(s)
- Chengting Chang
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingzhu Yuan
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingxia Zhang
- Department of Organization, West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xinrong Chen
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Ka Li
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| |
Collapse
|
14
|
Abstract
Chronic low-level inflammation is a causative factor in many of our common diseases. Switching to an anti-inflammatory diet is an important step that patients can take in for rectifying this risk factor. In this review, the author discusses the essential components of an anti-inflammatory diet and its contribution not only to the overall well-being but also to the body's defense against disease. The human microbiome is reviewed in detail and dietary connections and recommendations are explained for several otolaryngologic conditions.
Collapse
Affiliation(s)
- Agnes Czibulka
- Yale University, Quinnipiac University, 6 Burgis Lane, Guilford 06437, USA.
| |
Collapse
|
15
|
Piazzesi A, Putignani L. Extremely small and incredibly close: Gut microbes as modulators of inflammation and targets for therapeutic intervention. Front Microbiol 2022; 13:958346. [PMID: 36071979 PMCID: PMC9441770 DOI: 10.3389/fmicb.2022.958346] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/25/2022] [Indexed: 11/15/2022] Open
Abstract
Chronic inflammation is a hallmark for a variety of disorders and is at least partially responsible for disease progression and poor patient health. In recent years, the microbiota inhabiting the human gut has been associated with not only intestinal inflammatory diseases but also those that affect the brain, liver, lungs, and joints. Despite a strong correlation between specific microbial signatures and inflammation, whether or not these microbes are disease markers or disease drivers is still a matter of debate. In this review, we discuss what is known about the molecular mechanisms by which the gut microbiota can modulate inflammation, both in the intestine and beyond. We identify the current gaps in our knowledge of biological mechanisms, discuss how these gaps have likely contributed to the uncertain outcome of fecal microbiota transplantation and probiotic clinical trials, and suggest how both mechanistic insight and -omics-based approaches can better inform study design and therapeutic intervention.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
- *Correspondence: Lorenza Putignani,
| |
Collapse
|
16
|
Trukhan DI. Disorders of intestinal microbiocenosis: expanding the application of probiotics. MEDITSINSKIY SOVET = MEDICAL COUNCIL 2022:132-143. [DOI: 10.21518/2079-701x-2022-16-7-132-143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The problem of interaction between a person and the intestinal microbiome is surrounded by many secrets and mysteries. The bacterial flora of the gastrointestinal tract has a local and systemic effect not only on the digestive system, but also on the entire body as a whole. Numerous studies have proved the pathogenetic relationship of the state of the intestinal biocenosis not only with diseases of the gastrointestinal tract, but also with pathological processes from other organs and systems of the body. In terms of its role in maintaining homeostasis, the intestinal microflora is not inferior to any other vital organ. In the presented review, the current aspects of the terminology and clinic of disorders of intestinal microbiocenosis are considered. Probiotics occupy an important place in the complex therapy of intestinal microbiocenosis disorders and the corresponding clinical manifestations. The review considers the main mechanisms of probiotic / host interaction, non-immunological and immunological effects of probiotics and the requirements for them, the main directions of use of representatives of the normal microflora Bifidobacterium and Lactobacillus. The data of meta-analyzes and systematic reviews, testifying to the expansion of indications for the appointment of probiotics, are considered the possibilities of probiotics in the complex therapy of Helicobacter pylori infection, syndrome of increased epithelial intestinal permeability, and the prevention of respiratory infections.The review concludes with the results of a search in the PubMed database on the possibility of using probiotics in the prevention and treatment of a new coronavirus infection COVID-19. The availability of modern, effective and safe probiotics in the arsenal of a practical doctor (primarily a general practitioner and general practitioner), and their use, contributes to the optimization of drug therapy not only in gastroenterological patients, but also in patients with other somatic pathologies, including those with new coronavirus infection COVID-19.
Collapse
|
17
|
Kioukis A, Pourjam M, Neuhaus K, Lagkouvardos I. Taxonomy Informed Clustering, an Optimized Method for Purer and More Informative Clusters in Diversity Analysis and Microbiome Profiling. FRONTIERS IN BIOINFORMATICS 2022; 2:864597. [PMID: 36304326 PMCID: PMC9580952 DOI: 10.3389/fbinf.2022.864597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Bacterial diversity is often analyzed using 16S rRNA gene amplicon sequencing. Commonly, sequences are clustered based on similarity cutoffs to obtain groups reflecting molecular species, genera, or families. Due to the amount of the generated sequencing data, greedy algorithms are preferred for their time efficiency. Such algorithms rely only on pairwise sequence similarities. Thus, sometimes sequences with diverse phylogenetic background are clustered together. In contrast, taxonomic classifiers use position specific taxonomic information in assigning a probable taxonomy to a given sequence. Here we introduce Taxonomy Informed Clustering (TIC), a novel approach that utilizes classifier-assigned taxonomy to restrict clustering to only those sequences that share the same taxonomic path. Based on this concept, we offer a complete and automated pipeline for processing of 16S rRNA amplicon datasets in diversity analyses. First, raw reads are processed to form denoised amplicons. Next, the denoised amplicons are taxonomically classified. Finally, the TIC algorithm progressively assigning clusters at molecular species, genus and family levels. TIC outperforms greedy clustering algorithms like USEARCH and VSEARCH in terms of clusters’ purity and entropy, when using data from the Living Tree Project as test samples. Furthermore, we applied TIC on a dataset containing all Bifidobacteriaceae-classified sequences from the IMNGS database. Here, TIC identified evidence for 1000s of novel molecular genera and species. These results highlight the straightforward application of the TIC pipeline and superior results compared to former methods in diversity studies. The pipeline is freely available at: https://github.com/Lagkouvardos/TIC.
Collapse
Affiliation(s)
| | - Mohsen Pourjam
- Core Facility Microbiome, ZIEL – Institute for Food & Health, Technical University Munich, Freising, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome, ZIEL – Institute for Food & Health, Technical University Munich, Freising, Germany
| | - Ilias Lagkouvardos
- Core Facility Microbiome, ZIEL – Institute for Food & Health, Technical University Munich, Freising, Germany
- Institute of Marine Biology, Biotechnology and Aquaculture, Hellenic Centre for Marine Research, Heraklion, Greece
- *Correspondence: Ilias Lagkouvardos,
| |
Collapse
|
18
|
Probiotics and Phytochemicals: Role on Gut Microbiota and Efficacy on Irritable Bowel Syndrome, Functional Dyspepsia, and Functional Constipation. GASTROINTESTINAL DISORDERS 2022. [DOI: 10.3390/gidisord4010005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Functional gastrointestinal disorders (FGIDs), such as irritable bowel syndrome, functional constipation, and functional dyspepsia, have had a high prevalence over the past few years. Recent evidence suggests that functional foods and bioactive compounds, such as probiotics and phytochemicals, may have a positive effect in treating the symptoms of the above diseases. In this systematic review study, 32 published studies were selected with the use of comprehensive scientific databases, according to PRISMA guidelines, with emphasis on recent interventional studies that reflect the effect of probiotics and selected phytochemicals on the improvement of FGID symptoms. The bioactive compounds in the selected studies were administered to patients either in capsule form or in enriched food products (yogurt, juice, etc.). According to the results, there is a correlation between the consumption of probiotics and phytochemicals, such as polyphenols, and the relief of symptoms in selected gastrointestinal disorders. Enriching foods that are regularly consumed by the population, such as fruit juices, yogurt, and cheese, with ingredients that may have a positive effect on gastrointestinal disorders, could be a possible novel goal for the management of these diseases. However, further evidence is required for the role of probiotics and phytochemicals in FGIDs to be fully understood.
Collapse
|
19
|
Bacteria and bacterial derivatives as delivery carriers for immunotherapy. Adv Drug Deliv Rev 2022; 181:114085. [PMID: 34933064 DOI: 10.1016/j.addr.2021.114085] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
There is growing interest in the role of microorganisms in human health and disease, with evidence showing that new types of biotherapy using engineered bacterial therapeutics, including bacterial derivatives, can address specific mechanisms of disease. The complex interactions between microorganisms and metabolic/immunologic pathways underlie many diseases with unmet medical needs, suggesting that targeting these interactions may improve patient treatment. Using tools from synthetic biology and chemical engineering, non-pathogenic bacteria or bacterial products can be programmed and designed to sense and respond to environmental signals to deliver therapeutic effectors. This review describes current progress in biotherapy using live bacteria and their derivatives to achieve therapeutic benefits against various diseases.
Collapse
|
20
|
Galica AN, Galica R, Dumitrașcu DL. Diet, fibers, and probiotics for irritable bowel syndrome. J Med Life 2022; 15:174-179. [PMID: 35419092 PMCID: PMC8999090 DOI: 10.25122/jml-2022-0028] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/21/2022] [Indexed: 11/20/2022] Open
Abstract
Many aspects make irritable bowel syndrome (IBS) challenging for both patients and physicians. The unclear pathogenesis with many pathways to be explored, bothering symptoms that affect the quality of life, and many subtypes of the condition are only a few reasons that make IBS difficult to control and obtain satisfactory results. Treatment options start with general advice for lifestyle, continue with non-pharmaceutical treatments, and finally touch classic treatments. In this review, pharmaceutical treatment options are not accounted for. Consensus groups and meta-analyses have concluded guidelines that overall are the same, with variations in the strength of recommendations and some cultural and geographical particularities. Dietary interventions, probiotics, and fibers can be seen as non-pharmaceutical treatments that coexist in various protocols because of the relevant evidence regarding their efficacy in treating IBS symptoms.
Collapse
Affiliation(s)
- Adelina Nicoleta Galica
- 2 Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Nursing, Faculty of Natural and Human Sciences, Fan S. Noli University, Korçe, Albania
| | - Reitano Galica
- Department of Obstetrics and Gynecology, Regional Hospital Korçe, Korçe, Albania
| | - Dan Lucian Dumitrașcu
- 2 Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
21
|
Evaluation of Bacterial Diversity and Evolutionary Dynamics of Gut Bifidobacterium longum Isolates Obtained from Older Individuals in Hubei Province, China. Microbiol Spectr 2022; 10:e0144221. [PMID: 35044201 PMCID: PMC8768838 DOI: 10.1128/spectrum.01442-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bifidobacterium longum predominates in the human gut throughout the life span, from birth to old age, and could alter the intestinal microbial population and immune function in the elderly. We investigated the intestinal bacterial diversity in the elderly, and further evaluated the genetic diversity and population structure of B. longum. The results revealed a distinct difference in gut bacterial populations between the elderly from Xiangyang and its neighboring region, Enshi city. A total of 62 bifidobacterial strains were isolated, 30 of which were found to be B. longum. The multilocus sequence typing (MLST) analysis also revealed that 437 B. longum isolates from diverse regions worldwide, including the 30 isolated in this study, could be classified into 341 sequence types (STs). They could be further clustered into 10 clonal complexes and 127 singleton STs, indicating a highly genetic diversity among B. longum isolates. Two putative clone complexes (CCs) containing the isolates from Xiangyang were found to be geographically specific, and a 213-bp recombination fragment was detected. Phylogenetic trees divided these 437 isolates into three lineages, corresponding to the three subspecies of B. longum. It is noteworthy that two isolates from the elderly were identified to be B. longum subsp. suis, while the others were B. longum subsp. longum. Together, our study characterized the intestinal bacterial diversity and evolution of B. longum in the elderly, and it could contribute to further studies on the genotyping and discrimination of B. longum. IMPORTANCEBifidobacterium longum are common inhabitants of the human gut throughout the life span, and have been associated with health-promoting effects, yet little is known about the genotype profile and evolution of these isolates. Our study showed that there was significant difference in gut bacterial community and abundance of B. longum between the elderly from two neighboring cities. Furthermore, the possible geographically specific STs, CCs, and intraspecies recombination fragment were found among the B. longum isolates from elderly.
Collapse
|
22
|
Hao S, Wang J, Wang Y. Effectiveness and safety of Bifidobacterium in preventing dental caries: a systematic review and meta-analysis. Acta Odontol Scand 2021; 79:613-622. [PMID: 33956564 DOI: 10.1080/00016357.2021.1921259] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES The effectiveness and safety of Bifidobacterium in dental caries prevention are controversial. Thus, we performed this systematic review and meta-analysis to explore the preventive value of Bifidobacterium. METHODS Eligible studies were identified from several databases, including PubMed, the Cochrane Library, Embase, Web of Science, and Scopus. Hand searches were also conducted in relevant bibliographies. We then extracted and pooled standardized mean difference (SMD) and risk ratio (RR) to analyze the anti-caries effect of Bifidobacterium with Stata 16.0 software. If the data obtained was not suitable for meta-analysis, qualitative descriptions were performed. RESULTS Compared with the placebo control group, there was no statistically significant reduction in Streptococcus mutans and Lactobacilli counts in saliva in the test group. Also, there were no significant differences in Streptococcus mutans and Lactobacillus counts in dental plaque and no significant difference in caries incidence in deciduous teeth. There was no significant difference in the incidence of adverse events between the Bifidobacterium and control groups. CONCLUSIONS Available evidence demonstrates that Bifidobacterium is neither effective in reducing Streptococcus mutans and Lactobacillus counts in the saliva or dental plaque nor in reducing the occurrence of caries in deciduous teeth. Evaluation of its safety requires further investigations. Therefore, Bifidobacterium is not a competent probiotic candidate to prevent dental caries.
Collapse
Affiliation(s)
- Siyuan Hao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Jiahe Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| | - Yan Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, P. R. China
| |
Collapse
|
23
|
McFarland LV, Karakan T, Karatas A. Strain-specific and outcome-specific efficacy of probiotics for the treatment of irritable bowel syndrome: A systematic review and meta-analysis. EClinicalMedicine 2021; 41:101154. [PMID: 34712929 PMCID: PMC8529205 DOI: 10.1016/j.eclinm.2021.101154] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Consistent guidance for choosing an appropriate probiotic for the treatment of irritable bowel syndrome is lacking. METHODS Literature databases searched included: PubMed, Google Scholar and NIH registry of clinical trials from inception to June 2021. Inclusion criteria: randomized controlled trials (RCTs) enrolling adult or pediatric IBS patients comparing probiotics against controls and ≥ 2 RCTs with common IBS outcome measures within each type of probiotic. Five common measures of IBS symptoms (changes in global Irritable Bowel Syndrome Severity Scoring System or IBS-SSS scores, frequency of global responders, changes in bloating or abdominal pain scores and frequency of abdominal pain relief) were used. This study was registered at Prospero (#CRD42018109169). FINDINGS We screened 521 studies and included 42 randomized controlled trials (45 treatment arms, N = 3856). Four probiotics demonstrated significant reduction in abdominal pain relief: B. coagulans MTCC5260 (RR= 4.9, 95% C.I. 3.3, 7.3), L. plantarum 299v (RR= 4.6, 95% CI 1.9, 11.0), S. boulardii CNCM I-745 (RR= 1.5, 95% C.I. 1.1, 2.1) and S. cerevisiae CNCM I-3856 (RR= 1.3, 95% C.I. 1.04, 1.6). Mild-moderate adverse events were reported in 51% of the trials, none were more associated with the probiotic compared to controls. INTERPRETATION Although the analysis of probiotic efficacy was limited by the diversity of IBS outcomes used in trials and lack of confirmatory trials for some strains, six single-strain probiotics and three different types of probiotic mixtures showed significant efficacy for at least one IBS outcome measure. These results might be relevant to clinical practice and policy.
Collapse
Affiliation(s)
- Lynne V. McFarland
- Department Medicinal Chemistry, School of Pharmacy, University of Washington, 6047 38th Avenue NE, Seattle, WA 98195, United States
| | - Tarkan Karakan
- Department of Gastroenterology, Gazi University Faculty of Medicine, Beşevler, Ankara 06500, Turkey
| | - Ali Karatas
- Department of Gastroenterology, Gazi University Faculty of Medicine, Beşevler, Ankara 06500, Turkey
| |
Collapse
|
24
|
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: 42] [Impact Index Per Article: 10.5] [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
|
25
|
Wassenaar TM, Juncos VA, Zimmermann K. Interactions between the Gut Microbiome, Lung Conditions, and Coronary Heart Disease and How Probiotics Affect These. Int J Mol Sci 2021; 22:ijms22189700. [PMID: 34575864 PMCID: PMC8472021 DOI: 10.3390/ijms22189700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022] Open
Abstract
The importance of a healthy microbiome cannot be overemphasized. Disturbances in its composition can lead to a variety of symptoms that can extend to other organs. Likewise, acute or chronic conditions in other organs can affect the composition and physiology of the gut microbiome. Here, we discuss interorgan communication along the gut–lung axis, as well as interactions between lung and coronary heart diseases and between cardiovascular disease and the gut microbiome. This triangle of organs, which also affects the clinical outcome of COVID-19 infections, is connected by means of numerous receptors and effectors, including immune cells and immune-modulating factors such as short chain fatty acids (SCFA) and trimethlamine–N–oxide (TMAO). The gut microbiome plays an important role in each of these, thus affecting the health of the lungs and the heart, and this interplay occurs in both directions. The gut microbiome can be influenced by the oral uptake of probiotics. With an improved understanding of the mechanisms responsible for interorgan communication, we can start to define what requirements an ‘ideal’ probiotic should have and its role in this triangle.
Collapse
Affiliation(s)
- Trudy M. Wassenaar
- Molecular Microbiology and Genomics Consultants, Tannenstrasse 7, 55576 Zotzenheim, Germany
- Correspondence:
| | - Valentina A. Juncos
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72209, USA;
| | | |
Collapse
|
26
|
Aziz MNM, Kumar J, Muhammad Nawawi KN, Raja Ali RA, Mokhtar NM. Irritable Bowel Syndrome, Depression, and Neurodegeneration: A Bidirectional Communication from Gut to Brain. Nutrients 2021; 13:nu13093061. [PMID: 34578939 PMCID: PMC8468817 DOI: 10.3390/nu13093061] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with irritable bowel syndrome (IBS) are increasingly presenting with a wide range of neuropsychiatric symptoms, such as deterioration in gastroenteric physiology, including visceral hypersensitivity, altered intestinal membrane permeability, and gastrointestinal motor dysfunction. Functional imaging of IBS patients has revealed several abnormalities in various brain regions, such as significant activation of amygdala, thinning of insular and anterior cingulate cortex, and increase in hypothalamic gray matter, which results in poor psychiatric and cognitive outcomes. Interrelations between the enteric and central events in IBS-related gastrointestinal, neurological, and psychiatric pathologies have compelled researchers to study the gut-brain axis-a bidirectional communication that maintains the homeostasis of the gastrointestinal and central nervous system with gut microbiota as the protagonist. Thus, it can be disrupted by any alteration owing to the gut dysbiosis or loss of diversity in microbial composition. Available evidence indicates that the use of probiotics as a part of a balanced diet is effective in the management of IBS and IBS-associated neurodegenerative and psychiatric comorbidities. In this review, we delineate the pathogenesis and complications of IBS from gastrointestinal and neuropsychiatric standpoints while also discussing the neurodegenerative events in enteric and central nervous systems of IBS patients and the therapeutic potential of gut microbiota-based therapy established on clinical and preclinical data.
Collapse
Affiliation(s)
- Muhammad Nazirul Mubin Aziz
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.N.M.A.); (J.K.)
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.N.M.A.); (J.K.)
- Gut Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (K.N.M.N.); (R.A.R.A.)
| | - Khairul Najmi Muhammad Nawawi
- Gut Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (K.N.M.N.); (R.A.R.A.)
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Raja Affendi Raja Ali
- Gut Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (K.N.M.N.); (R.A.R.A.)
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Norfilza M. Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (M.N.M.A.); (J.K.)
- Gut Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia; (K.N.M.N.); (R.A.R.A.)
- Correspondence:
| |
Collapse
|
27
|
Ivashkin KV, Grechishnikova VR, Reshetova MS, Ivashkin VT. Irritable Bowel and Bacterial Overgrowth Syndromes: a Bacterial Link Hypothesis of Functional Disease. RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2021; 31:54-63. [DOI: 10.22416/1382-4376-2021-31-1-54-63] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Aim. Assessment of the irritable bowel syndrome (IBS) and small intestinal bacterial overgrowth syndrome (SIBO) interlinkage.Key points. SIBO may represent a "peripheral" mechanism of IBS, aside to nonspecific inflammation, increased epithelial permeability and local immune system activation. In various assays, the SIBO rate in IBS patients was 4-46% vs. 0-13% in an intact cohort. A limited diagnosability of SIBO obscures the SIBO-IBS causal interplay. Impaired motility in IBS may predispose to the SIBO development. Proinflammatory cytokines and mediators in SIBO, in turn, provoke visceral hypersensitivity and intense motility, the key IBS factors. Both conditions relate to qualitative and quantitative changes in microbiota, which warrants the application of probiotic Lactobacillus and Bifidobacterium strains.Conclusion. Further research into the SIBO-IBS interface is required for developing optimal probiotic-based therapies.
Collapse
Affiliation(s)
- K. V. Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | - M. S. Reshetova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V. T. Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
28
|
|
29
|
Li Y, Hong G, Yang M, Li G, Jin Y, Xiong H, Qian W, Hou X. Fecal bacteria can predict the efficacy of rifaximin in patients with diarrhea-predominant irritable bowel syndrome. Pharmacol Res 2020; 159:104936. [PMID: 32470562 DOI: 10.1016/j.phrs.2020.104936] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/15/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Rifaximin for treating diarrhea-predominant irritable bowel syndrome (IBS-D) by regulating intestinal microbiota has been studied and recommended. In this study, we tried to investigate the effect of rifaximin on different components of intestinal microbiota and explore which component of gut microbiota can predict the efficacy of rifaximin in IBS-D. METHODS Healthy controls (HC) and IBS-D patients meeting the Rome III criteria were recruited, and IBS-D patients were orally administered 400 mg rifaximin three times daily for 2 weeks. Subjects were tested for small intestinal bacterial overgrowth (SIBO), their symptoms were recorded, and fecal and rectal mucosal samples were collected before and after treatment. Fecal and rectal mucosal bacterial data were obtained via 16S rRNA sequencing, and fecal fungal data were obtained via ITS2 sequencing. RESULTS IBS-D patients were divided into two subgroups based on fecal bacterial composition, IBS1 (patients whose fecal bacterial composition were different from HC) and IBS0 (patients whose fecal bacterial profiles were similar to HC). Rifaximin increased fecal Bifidobacterium and decreased E. coli and Enterobacter in IBS1 patients. Although rectal mucosal bacteria and fecal fungi were not significantly altered in all patients after rifaximin intervention, rifaximin enhanced the connections among fecal bacteria, mucosal bacteria and fecal fungi in IBS1 patients. Compared with IBS0, we surprisingly found rifaximin ameliorated abdominal symptoms of IBS1 much better. Receiver operating curve analysis revealed patients whose fecal microbial dysbiosis indices (MDI) were higher than -3.006 could be diagnosed as IBS1. CONCLUSION Fecal bacterial dysbiosis could be a biomarker for rifaximin treatment for IBS-D.
Collapse
Affiliation(s)
- Ying Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gaichao Hong
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Min Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gangping Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Jin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hanhua Xiong
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
30
|
Bai T, Zeng H, Long Y, Li X, Sun X, Lan Y, Gao L, Zhang L, Feng Z, Hou X. The efficacy of Bifidobacterium quadruple viable tablet in the treatment of diarrhea predominant irritable bowel syndrome: protocol for a randomized, double-blind, placebo-controlled, multicenter trial. Trials 2020; 21:597. [PMID: 32605578 PMCID: PMC7329396 DOI: 10.1186/s13063-020-04490-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/08/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal disorders characterized by recurrent abdominal pain associated with defecation or a change in bowel habits. Leading to significant negative effect on patients' quality of life and huge financial burden to health system, the management of IBS is a great challenge. Probiotics are considered as an effective therapy; however, in a lack of high-quality evidence of efficacy, no strain- and dose-specific probiotics were recommended in clinical guidelines. This study aims to evaluate the efficacy of the Bifidobacterium quadruple viable tablet in the treatment of IBS-D. METHODS/DESIGN A multicenter randomized controlled trial will be performed in fourteen hospitals. A total of three hundred patients who fulfill the eligibility criteria will be stratified divided into an experimental group and a control group randomly in a ratio of 1:1. The experimental group is treated with the Bifidobacterium quadruple viable tablet while the control group is treated with placebo. All the patients will receive a 4-week treatment and a 2-week follow-up. The primary outcome is the effectiveness in improving abdominal pain and stool consistency; the secondary outcome includes evaluation of overall symptom relief, frequency of defecation, bloating, urgency of defecation, remedial medication, score of IBS-QOL, and changes of microbiota and metabonomics. Physical examination, vital signs, laboratory tests, adverse events, and concomitant medication will be taken into account for intervention safety assessment during the trial. DISCUSSION This multicenter randomized controlled trial may provide high-quality evidence on the efficacy of the Bifidobacterium quadruple viable tablet for IBS-D on both physical and mental dimensions in China. To fill the gap of previous probiotic intervention studies, in addition, this study will also present safety assessment which will be a significant emphasis. TRIAL REGISTRATION ChiCTR1800017721 . Registered on 10 August 2018.
Collapse
Affiliation(s)
- Tao Bai
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, China
| | - Haoyu Zeng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, China
| | - Yanqin Long
- Division of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang, China
| | - Xiaoqing Li
- Division of Gastroenterology, Peking Union Medical College Hospital, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Xiaohong Sun
- Division of Gastroenterology, Peking Union Medical College Hospital, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, China
| | - Yu Lan
- Division of Gastroenterology, Beijing Jishuitan Hospital, Xicheng District Xinjiekou No. 31 East Street, Beijing, China
| | - Lingling Gao
- Peking University Clinical Research Institute, No.38 Xueyuan Road, Haidian District, Beijing, China
| | - Lu Zhang
- Hangzhou Grand Biologic Pharmaceutical. INC, 63 Jiuhuan Road, Jianggan District, Hangzhou, Zhejiang, China
| | - Zenghui Feng
- Hangzhou Grand Biologic Pharmaceutical. INC, 63 Jiuhuan Road, Jianggan District, Hangzhou, Zhejiang, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei, China.
| |
Collapse
|
31
|
Kim MJ, Min YW, Lee C, Jang YS, Kim CH, Chang DK. The effects of three novel probiotics isolated from the Korean fermented food Kimchi on the stress-induced defecation of rats. PRECISION AND FUTURE MEDICINE 2020. [DOI: 10.23838/pfm.2019.00121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
32
|
Mogilnicka I, Ufnal M. Gut Mycobiota and Fungal Metabolites in Human Homeostasis. Curr Drug Targets 2020; 20:232-240. [PMID: 30047327 DOI: 10.2174/1389450119666180724125020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 05/13/2018] [Accepted: 07/19/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Accumulating evidence suggests that microbiota play an important role in host's homeostasis. Thus far, researchers have mostly focused on the role of bacterial microbiota. However, human gut is a habitat for several fungal species, which produce numerous metabolites. Furthermore, various types of food and beverages are rich in a wide spectrum of fungi and their metabolites. METHODS We searched PUBMED and Google Scholar databases to identify clinical and pre-clinical studies on fungal metabolites, composition of human mycobiota and fungal dysbiosis. RESULTS Fungal metabolites may serve as signaling molecules and exert significant biological effects including trophic, anti-inflammatory or antibacterial actions. Finally, research suggests an association between shifts in gut fungi composition and human health. Changes in mycobiota composition have been found in obesity, hepatitis and inflammatory bowel diseases. CONCLUSION The influence of mycobiota and dietary fungi on homeostasis in mammals suggests a pharmacotherapeutic potential of modulating the mycobiota which may include treatment with probiotics and fecal transplantation. Furthermore, antibacterial action of fungi-derived molecules may be considered as a substitution for currently used antibacterial agents and preservatives in food industry.
Collapse
Affiliation(s)
- Izabella Mogilnicka
- Department of Experimental Physiology and Pathophysiology, Laboratory of the Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Ufnal
- Department of Experimental Physiology and Pathophysiology, Laboratory of the Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
33
|
Kumar R, Sood U, Gupta V, Singh M, Scaria J, Lal R. Recent Advancements in the Development of Modern Probiotics for Restoring Human Gut Microbiome Dysbiosis. Indian J Microbiol 2020; 60:12-25. [PMID: 32089570 PMCID: PMC7000592 DOI: 10.1007/s12088-019-00808-y] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/17/2019] [Indexed: 12/17/2022] Open
Abstract
A healthy gut is predominantly occupied by bacteria which play a vital role in nutrition and health. Any change in normal gut homeostasis imposes gut dysbiosis. So far, efforts have been made to mitigate the gastrointestinal symptoms using modern day probiotics. The majority of the probiotics strains used currently belong to the genera Lactobacillus, Clostridium, Bifidobacterium and Streptococcus. Recent advancements in culturomics by implementing newer techniques coupled with the use of gnotobiotic animal models provide a subtle ground to develop novel host specific probiotics therapies. In this review article, the recent advances in the development of microbe-based therapies which can now be implemented to treat a wide spectrum of diseases have been discussed. However, these probiotics are not classified as drugs and there is a lack of stringent law enforcement to protect the end users against the pseudo-probiotic products. While modern probiotics hold strong promise for the future, more rigorous regulations are needed to develop genuine probiotic products and characterize novel probiotics using the latest research and technology. This article also highlights the possibility of reducing antibiotic usage by utilizing probiotics developed using the latest concepts of syn and ecobiotics.
Collapse
Affiliation(s)
- Roshan Kumar
- PhiXGen Private Limited, Gurugram, Haryana 122001 India
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD USA
- South Dakota Centre for Biologics Research and Commercialization, Brookings, SD USA
| | - Utkarsh Sood
- PhiXGen Private Limited, Gurugram, Haryana 122001 India
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Vipin Gupta
- PhiXGen Private Limited, Gurugram, Haryana 122001 India
- Department of Zoology, University of Delhi, Delhi, 110007 India
| | - Mona Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi 110067 India
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD USA
- South Dakota Centre for Biologics Research and Commercialization, Brookings, SD USA
| | - Rup Lal
- PhiXGen Private Limited, Gurugram, Haryana 122001 India
- Department of Zoology, University of Delhi, Delhi, 110007 India
| |
Collapse
|
34
|
Rahmani P, Moradzadeh A, Farahmand F. Giving probiotics to your children for gastrointestinal problems: In the light of scientific findings. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100164] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Nelkowska DD. Treating irritable bowel syndrome through an interdisciplinary approach. Ann Gastroenterol 2019; 33:1-8. [PMID: 31892791 PMCID: PMC6928481 DOI: 10.20524/aog.2019.0441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/15/2019] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional disorder with a multifactorial etiology and a complex clinical picture. The recent discovery of the dysregulation of the gut-brain axis as an important pathogenetic mechanism for the development of IBS is a kind of breakthrough in the understanding of IBS and prevalent comorbidities. Nevertheless, IBS treatment still causes many problems and often turns out to be ineffective or brings only short-term effects in reducing symptom severity. In reference to the characteristics of IBS, including new findings regarding etiopathogenesis, an interdisciplinary treatment approach is proposed and the roles of medical and psychological interventions are underlined. The literature search was conducted using electronic databases with a focus on the latest publications. The review may be useful for matching the best strategy of IBS management.
Collapse
|
36
|
van Thiel IAM, Botschuijver S, de Jonge WJ, Seppen J. Painful interactions: Microbial compounds and visceral pain. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165534. [PMID: 31634534 DOI: 10.1016/j.bbadis.2019.165534] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/18/2022]
Abstract
Visceral pain, characterized by abdominal discomfort, originates from organs in the abdominal cavity and is a characteristic symptom in patients suffering from irritable bowel syndrome, vulvodynia or interstitial cystitis. Most organs in which visceral pain originates are in contact with the external milieu and continuously exposed to microbes. In order to maintain homeostasis and prevent infections, the immune- and nervous system in these organs cooperate to sense and eliminate (harmful) microbes. Recognition of microbial components or products by receptors expressed on cells from the immune and nervous system can activate immune responses but may also cause pain. We review the microbial compounds and their receptors that could be involved in visceral pain development.
Collapse
Affiliation(s)
- I A M van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - S Botschuijver
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands
| | - J Seppen
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, location AMC, Meibergdreef 69, 1105 BK Amsterdam, the Netherlands.
| |
Collapse
|
37
|
Gaucher F, Kponouglo K, Rabah H, Bonnassie S, Ossemond J, Pottier S, Jardin J, Briard-Bion V, Marchand P, Blanc P, Jeantet R, Jan G. Propionibacterium freudenreichii CIRM-BIA 129 Osmoadaptation Coupled to Acid-Adaptation Increases Its Viability During Freeze-Drying. Front Microbiol 2019; 10:2324. [PMID: 31681198 PMCID: PMC6797830 DOI: 10.3389/fmicb.2019.02324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022] Open
Abstract
Propionibacterium freudenreichii is a beneficial bacterium with documented effects on the gut microbiota and on inflammation. Its presence within the animal and human intestinal microbiota was correlated with immunomodulatory effects, mediated by both propionibacterial surface components and by secreted metabolites. It is widely implemented, both in the manufacture of fermented dairy products such as Swiss-type cheeses, and in the production of probiotic food complements, under the form of freeze-dried powders. The bottleneck of this drying process consists in the limited survival of bacteria during drying and storage. Protective pre-treatments have been applied to other bacteria and may, in a strain-dependent manner, confer enhanced resistance. However, very little information was yet published on P. freudenreichii adaptation to freeze-drying. In this report, an immunomodulatory strain of this probiotic bacterium was cultured under hyperosmotic constraint in order to trigger osmoadaptation. This adaptation was then combined with acid or thermal pre-treatment. Such combination led to accumulation of key stress proteins, of intracellular compatible solute glycine betaine, to modulation of the propionibacterial membrane composition, and to enhanced survival upon freeze-drying. This work opens new perspectives for efficient production of live and active probiotic propionibacteria.
Collapse
Affiliation(s)
- Floriane Gaucher
- UMR STLO, Agrocampus Ouest, INRA, Rennes, France
- Bioprox, Levallois-Perret, France
| | | | - Houem Rabah
- UMR STLO, Agrocampus Ouest, INRA, Rennes, France
- Bba, Pôle Agronomique Ouest, Régions Bretagne et Pays de la Loire, Rennes, France
| | - Sylvie Bonnassie
- UMR STLO, Agrocampus Ouest, INRA, Rennes, France
- Université de Rennes I, Rennes, France
| | | | - Sandrine Pottier
- CNRS, ISCR – UMR 6226, PRISM, BIOSIT – UMS 3480 Université de Rennes I, Rennes, France
| | | | | | | | | | | | - Gwénaël Jan
- UMR STLO, Agrocampus Ouest, INRA, Rennes, France
| |
Collapse
|
38
|
Effectiveness of probiotics in irritable bowel syndrome: methodological quality of meta-analyses and systematic reviews. FRONTIERS OF NURSING 2019. [DOI: 10.2478/fon-2019-0018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Objective
This study assessed the methodological quality of systematic reviews/meta-analysis of the effectiveness of probiotics against irritable bowel syndrome (IBS) using the accepted methodological quality assessment scale AMSTAR and explored the factors that influenced the quality of methodology. It was designed to provide a reference for future research and systematic reviews/metaanalysis.
Methods
The methodological quality of existing systematic reviews/meta-analysis was evaluated using the AMSTAR scale. Influencing factors of methodological quality were statistically analyzed using RevMan 5.3 software. The included systematic reviews/metaanalysis must include the following characteristics: (1) methods using systematic evaluation/meta-analysis, (2) probiotic intervention, and (3) language limitation to Chinese and English.
Results
The AMSTAR score was 5–9 (7.42 ± 1.22), and the quality is above average. The factors affecting the methodological quality are the number of authors and whether they cooperate with the institution.
Conclusions
Studies have shown that current systematic reviews/meta-analysis of the effectiveness of probiotics on IBS does not fully comply with methodological quality standards, and therefore the methodological quality of research in this area needs to be strengthened. To better clarify how probiotics affect IBS, future systematic reviews and meta-analyses should not only follow methodological quality standards but also include more effective outcome measures, and they should focus more on the discussion of research results. We look forward to the development of higher-quality randomized controlled trials in the future.
Collapse
|
39
|
Chong PP, Chin VK, Looi CY, Wong WF, Madhavan P, Yong VC. The Microbiome and Irritable Bowel Syndrome - A Review on the Pathophysiology, Current Research and Future Therapy. Front Microbiol 2019; 10:1136. [PMID: 31244784 PMCID: PMC6579922 DOI: 10.3389/fmicb.2019.01136] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 05/06/2019] [Indexed: 11/16/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional disorder which affects a large proportion of the population globally. The precise etiology of IBS is still unknown, although consensus understanding proposes IBS to be of multifactorial origin with yet undefined subtypes. Genetic and epigenetic factors, stress-related nervous and endocrine systems, immune dysregulation and the brain-gut axis seem to be contributing factors that predispose individuals to IBS. In addition to food hypersensitivity, toxins and adverse life events, chronic infections and dysbiotic gut microbiota have been suggested to trigger IBS symptoms in tandem with the predisposing factors. This review will summarize the pathophysiology of IBS and the role of gut microbiota in relation to IBS. Current methodologies for microbiome studies in IBS such as genome sequencing, metagenomics, culturomics and animal models will be discussed. The myriad of therapy options such as immunoglobulins (immune-based therapy), probiotics and prebiotics, dietary modifications including FODMAP restriction diet and gluten-free diet, as well as fecal transplantation will be reviewed. Finally this review will highlight future directions in IBS therapy research, including identification of new molecular targets, application of 3-D gut model, gut-on-a-chip and personalized therapy.
Collapse
Affiliation(s)
- Pei Pei Chong
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Voon Kin Chin
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Chung Yeng Looi
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Priya Madhavan
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Voon Chen Yong
- School of Biosciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
40
|
Abstract
In recent years, interest in the relationship between gut microbiota and disease states has grown considerably. Indeed, several strategies have been employed to modify the microbiome through the administration of different diets, by the administration of antibiotics or probiotics, or even by transplantation of feces. In the present manuscript, we focus specifically on the potential application of probiotics, which seem to be a safe strategy, in the management of digestive, pain, and emotional disorders. We present evidence from animal models and human studies, notwithstanding that translation to clinic still deserves further investigation. The microbiome influences gut functions as well as neurological activity by a variety of mechanisms, which are also discussed. The design and performance of larger trials is urgently needed to verify whether these new strategies might be useful not only for the treatment of disorders affecting the gastrointestinal tract but also in the management of emotional and pain disorders not directly related to the gut.
Collapse
|
41
|
Shapiro J, Bernica J, Hernaez R. Risk of Bias Analysis of Systematic Reviews of Probiotics for Treatment of Irritable Bowel Syndrome. Clin Gastroenterol Hepatol 2019; 17:784-785. [PMID: 30012436 DOI: 10.1016/j.cgh.2018.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Global prevalence of irritable bowel syndrome (IBS) is between 7% and 21%.1 Pathogenesis of IBS symptoms such as pain, altered bowel habits, and bloating is multifactorial, with central and peripheral mechanisms serving as targets for treatments.2 Probiotics have been studied for the treatment of IBS and are well tolerated and safe.3.
Collapse
Affiliation(s)
- Jordan Shapiro
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, Texas.
| | - Jessica Bernica
- Internal Medicine Residency Program, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Ruben Hernaez
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas; Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey VA Medical Center, Houston, Texas; Section of Gastroenterology, Michael E. DeBakey VA Medical Center, Houston, Texas
| |
Collapse
|
42
|
Ooi SL, Correa D, Pak SC. Probiotics, prebiotics, and low FODMAP diet for irritable bowel syndrome - What is the current evidence? Complement Ther Med 2019; 43:73-80. [PMID: 30935559 DOI: 10.1016/j.ctim.2019.01.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/15/2019] [Indexed: 11/26/2022] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal disorders worldwide. While the pathogenesis is not clearly understood, current research points to the role of the gut microbiome and alterations in the diversity of the microbiota. Probiotics, prebiotics, and low FODMAP diet are therapeutic means associated with modification of the gut microbiome for the alleviation of IBS symptoms. This narrative review assesses the current evidence on the efficacy of these treatment options based on findings from recent systematic reviews and meta-analyses published from October 2013 to October 2018. There is a general agreement in the 11 included systematic reviews and meta-analyses that probiotic therapy is safe and can be effective in improving overall IBS symptom scores and abdominal pain in the general IBS population. Nonetheless, conflicting findings remain and no recommendation on the specific species/strains or combination can be made. Short-term restriction of FODMAP in the diet can improve IBS symptoms as per the findings of 7 systematic reviews and meta-analyses, even though the quality of the evidence remains questionable. Inappropriate use of the low FODMAP diet can potentially impact health negatively. As such, a low FODMAP diet is only recommended as a second line treatment guided by qualified clinicians with specialized training. Despite preclinical studies of some prebiotics demonstrated the potential use in improving gut microbiome and intestinal inflammatory response, the beneficial effect of prebiotics for IBS remains theoretical. Two systematic reviews found no evidence to support the clinical use of prebiotics for IBS.
Collapse
Affiliation(s)
- Soo Liang Ooi
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Dianne Correa
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia
| | - Sok Cheon Pak
- School of Biomedical Sciences, Charles Sturt University, Bathurst, NSW 2795, Australia.
| |
Collapse
|
43
|
O'Malley D. Endocrine regulation of gut function - a role for glucagon-like peptide-1 in the pathophysiology of irritable bowel syndrome. Exp Physiol 2019; 104:3-10. [PMID: 30444291 DOI: 10.1113/ep087443] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the topic of this review? Pathophysiological changes linked to irritable bowel syndrome (IBS) include stress and immune activation, changes in gastrointestinal microbial and bile acid profiles and sensitization of extrinsic and intrinsic gut neurons. This review explores the potential role for L-cells in these pathophysiological changes. What advances does it highlight? L-cells, which secrete glucagon-like peptide-1 in response to nutrients, microbial factors, bile acids and short-chain fatty acids, may sense IBS-related changes in the luminal environment. Glucagon-like peptide-1 can act as a hormone, a paracrine factor or a neuromodulatory factor and, through its actions on central or peripheral neurons, may play a role in gastrointestinal dysfunction. ABSTRACT The prevalent and debilitating functional bowel disorder, irritable bowel syndrome (IBS), is characterized by symptoms that include abdominal pain, bloating, diarrhoea and/or constipation. The heterogeneity of IBS underscores a complex multifactorial pathophysiology, which is not completely understood but involves dysfunction of the bi-directional signalling axis between the brain and the gut. This axis incorporates efferent and afferent branches of the autonomic nervous system, circulating endocrine hormones and immune factors, local paracrine and neurocrine factors and microbial metabolites. L-cells, which are electrically excitable biosensors embedded in the gastrointestinal epithelium, secrete glucagon-like peptide-1 (GLP-1) in response to nutrients in the small intestine. However, they appear to function in a different manner more distally in the gastrointestinal tract, where they are activated by luminal factors including short-chain fatty acids, bile acids and microbial metabolic products, all of which are altered in IBS patients. Glucagon-like peptide-1 can also interact with the hypothalamic-pituitary-adrenal stress axis and the immune system, both of which are activated in IBS. Given that a GLP-1 mimetic has been found to alleviate acute pain symptoms in IBS patients, GLP-1 might be important in the manifestation of IBS symptoms. This review assesses the current knowledge about the role of GLP-1 in IBS pathophysiology and its potential role as a signal transducer in the microbiome-gut-brain signalling axis.
Collapse
Affiliation(s)
- Dervla O'Malley
- Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
44
|
Camilleri M. Management Options for Irritable Bowel Syndrome. Mayo Clin Proc 2018; 93:1858-1872. [PMID: 30522596 PMCID: PMC6314474 DOI: 10.1016/j.mayocp.2018.04.032] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 04/23/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023]
Abstract
Irritable bowel syndrome (IBS) is associated with diverse pathophysiologic mechanisms. These mechanisms include increased abnormal colonic motility or transit, intestinal or colorectal sensation, increased colonic bile acid concentration, and superficial colonic mucosal inflammation, as well as epithelial barrier dysfunction, neurohormonal up-regulation, and activation of secretory processes in the epithelial layer. Novel approaches to treatment include lifestyle modification, changes in diet, probiotics, and pharmacotherapy directed to the motility, sensation, and intraluminal milieu of patients with IBS. Despite recent advances, there is a need for development of new treatments to relieve pain in IBS without deleterious central or other adverse effects.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.) Program and Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN.
| |
Collapse
|
45
|
Liu Y, Tran DQ, Rhoads JM. Probiotics in Disease Prevention and Treatment. J Clin Pharmacol 2018; 58 Suppl 10:S164-S179. [PMID: 30248200 PMCID: PMC6656559 DOI: 10.1002/jcph.1121] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/17/2018] [Indexed: 12/17/2022]
Abstract
Few treatments for human diseases have received as much investigation in the past 20 years as probiotics. In 2017, English-language meta-analyses totaling 52 studies determined the effect of probiotics on conditions ranging from necrotizing enterocolitis and colic in infants to constipation, irritable bowel syndrome, and hepatic encephalopathy in adults. The strongest evidence in favor of probiotics lies in the prevention or treatment of 5 disorders: necrotizing enterocolitis, acute infectious diarrhea, acute respiratory tract infections, antibiotic-associated diarrhea, and infant colic. Probiotic mechanisms of action include the inhibition of bacterial adhesion; enhanced mucosal barrier function; modulation of the innate and adaptive immune systems (including induction of tolerogenic dendritic cells and regulatory T cells); secretion of bioactive metabolites; and regulation of the enteric and central nervous systems. Future research is needed to identify the optimal probiotic and dose for specific diseases, to address whether the addition of prebiotics (to form synbiotics) would enhance activity, and to determine if defined microbial communities would provide benefit exceeding that of single-species probiotics.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, and the Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Dat Q Tran
- Department of Pediatrics, Division of Gastroenterology, and the Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - J Marc Rhoads
- Department of Pediatrics, Division of Gastroenterology, and the Pediatric Research Center, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| |
Collapse
|
46
|
Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features. Cell 2018; 174:1388-1405.e21. [DOI: 10.1016/j.cell.2018.08.041] [Citation(s) in RCA: 946] [Impact Index Per Article: 135.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 06/05/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022]
|
47
|
McFarland LV, Evans CT, Goldstein EJC. Strain-Specificity and Disease-Specificity of Probiotic Efficacy: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2018; 5:124. [PMID: 29868585 PMCID: PMC5949321 DOI: 10.3389/fmed.2018.00124] [Citation(s) in RCA: 317] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
Background As the use and diversity of probiotic products expands, the choice of an appropriate type of probiotic is challenging for both medical care professionals and the public alike. Two vital factors in choosing the appropriate probiotic are often ignored, namely, the probiotic strain-specificity and disease-specificity for efficacy. Reviews and meta-analyses often pool together different types of probiotics, resulting in misleading conclusions of efficacy. Methods A systematic review of the literature (1970-2017) assessing strain-specific and disease-specific probiotic efficacy was conducted. Trials were included for probiotics with an identifiable strain (either single strain or mixtures of strains) that had at least two randomized, controlled trials for each type of disease indication. The goal was to determine if probiotic strains have strain and/or disease-specific efficacy. Results We included 228 trials and found evidence for both strain specificity and disease specificity for the efficacy of specific probiotic strains. Significant efficacy evidence was found for 7 (70%) of probiotic strain(s) among four preventive indications and 11 (65%) probiotic strain(s) among five treatment indications. Strain-specific efficacy for preventing adult antibiotic-associated diarrhea was clearly demonstrated within the Lactobacillus species [e.g., by the mixture of Lactobacillus acidophilus CL1285, Lactobacillus casei LBC80R, and Lactobacillus rhamnosus CLR2 (Bio-K+®), by L. casei DN114001 (Actimel®) and by Lactobacillus reuteri 55730], while other Lactobacillus strains did not show efficacy. Significant disease-specific variations in efficacy was demonstrated by L. rhamnosus GG and Saccharomyces boulardii CNCM I-745, as well as other probiotic strains. Conclusion Strong evidence was found supporting the hypothesis that the efficacy of probiotics is both strain-specific and disease-specific. Clinical guidelines and meta-analyses need to recognize the importance of reporting outcomes by both specific strain(s) of probiotics and the type of disease. The clinical relevance of these findings indicates that health-care providers need to take these two factors into consideration when recommending the appropriate probiotic for their patient.
Collapse
Affiliation(s)
- Lynne V McFarland
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington Medical Center, Seattle, WA, United States
| | - Charlesnika T Evans
- Department of Preventive Medicine and Center for Healthcare Studies, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.,Department of Veterans Affairs (VA), Center of Innovation of Complex Chronic Healthcare (CINCCH), Edward Hines Jr VA Hospital, Hines, IL, United States
| | - Ellie J C Goldstein
- RM Alden Research Laboratory, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|
48
|
Hungin APS, Mitchell CR, Whorwell P, Mulligan C, Cole O, Agréus L, Fracasso P, Lionis C, Mendive J, Philippart de Foy J, Seifert B, Wensaas K, Winchester C, de Wit N, the European Society for Primary Care Gastroenterology. Systematic review: probiotics in the management of lower gastrointestinal symptoms - an updated evidence-based international consensus. Aliment Pharmacol Ther 2018; 47:1054-1070. [PMID: 29460487 PMCID: PMC5900870 DOI: 10.1111/apt.14539] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/15/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND In 2013, a systematic review and Delphi consensus reported that specific probiotics can benefit adult patients with irritable bowel syndrome (IBS) and other gastrointestinal (GI) problems. AIM To update the consensus with new evidence. METHODS A systematic review identified randomised, placebo-controlled trials published between January 2012 and June 2017. Evidence was graded, previously developed statements were reassessed by an 8-expert panel, and agreement was reached via Delphi consensus. RESULTS A total of 70 studies were included (IBS, 34; diarrhoea associated with antibiotics, 13; diarrhoea associated with Helicobacter pylori eradication therapy, 7; other conditions, 16). Of 15 studies that examined global IBS symptoms as a primary endpoint, 8 reported significant benefits of probiotics vs placebo. Consensus statements with 100% agreement and "high" evidence level indicated that specific probiotics help reduce overall symptom burden and abdominal pain in some patients with IBS and duration/intensity of diarrhoea in patients prescribed antibiotics or H. pylori eradication therapy, and have favourable safety. Statements with 70%-100% agreement and "moderate" evidence indicated that, in some patients with IBS, specific probiotics help reduce bloating/distension and improve bowel movement frequency/consistency. CONCLUSIONS This updated review indicates that specific probiotics are beneficial in certain lower GI problems, although many of the new publications did not report benefits of probiotics, possibly due to inclusion of new, less efficacious preparations. Specific probiotics can relieve lower GI symptoms in IBS, prevent diarrhoea associated with antibiotics and H. pylori eradication therapy, and show favourable safety. This study will help clinicians recommend/prescribe probiotics for specific symptoms.
Collapse
|
49
|
Rodiño-Janeiro BK, Vicario M, Alonso-Cotoner C, Pascua-García R, Santos J. A Review of Microbiota and Irritable Bowel Syndrome: Future in Therapies. Adv Ther 2018; 35:289-310. [PMID: 29498019 PMCID: PMC5859043 DOI: 10.1007/s12325-018-0673-5] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Indexed: 12/01/2022]
Abstract
Irritable bowel syndrome (IBS), one of the most frequent digestive disorders, is characterized by chronic and recurrent abdominal pain and altered bowel habit. The origin seems to be multifactorial and is still not well defined for the different subtypes. Genetic, epigenetic and sex-related modifications of the functioning of the nervous and immune-endocrine supersystems and regulation of brain-gut physiology and bile acid production and absorption are certainly involved. Acquired predisposition may act in conjunction with infectious, toxic, dietary and life event-related factors to enhance epithelial permeability and elicit mucosal microinflammation, immune activation and dysbiosis. Notably, strong evidence supports the role of bacterial, viral and parasitic infections in triggering IBS, and targeting microbiota seems promising in view of the positive response to microbiota-related therapies in some patients. However, the lack of highly predictive diagnostic biomarkers and the complexity and heterogeneity of IBS patients make management difficult and unsatisfactory in many cases, reducing patient health-related quality of life and increasing the sanitary burden. This article reviews specific alterations and interventions targeting the gut microbiota in IBS, including prebiotics, probiotics, synbiotics, non-absorbable antibiotics, diets, fecal transplantation and other potential future approaches useful for the diagnosis, prevention and treatment of IBS.
Collapse
Affiliation(s)
- Bruno K Rodiño-Janeiro
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.
| | - María Vicario
- Translational Mucosal Immunology Group, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain
| | - Carmen Alonso-Cotoner
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain
| | | | - Javier Santos
- Laboratory of Neuro-Immuno-Gastroenterology, Digestive System Research Unit, Vall d'Hebron Institut de Recerca, Department of Gastroenterology, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona (Facultat de Medicina), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Subdirección General de Investigación Sanitaria, Ministerio de Economía, Industria y Competitividad, Madrid, Spain.
| |
Collapse
|
50
|
Zhang J, Ren FG, Liu P, Zhang HK, Zhu HY, Feng Z, Zhang XF, Wang B, Liu XM, Zhang XG, Wu RQ, Lv Y. Characteristics of fecal microbial communities in patients with non-anastomotic biliary strictures after liver transplantation. World J Gastroenterol 2017; 23:8217-8226. [PMID: 29290658 PMCID: PMC5739928 DOI: 10.3748/wjg.v23.i46.8217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/13/2017] [Accepted: 11/07/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the possible relationship between fecal microbial communities and non-anastomotic stricture (NAS) after liver transplantation (LT). METHODS A total of 30 subjects including 10 patients with NAS, 10 patients with no complications after LT, and 10 non-LT healthy individuals were enrolled. Fecal microbial communities were assessed by the 16S rRNA gene sequencing technology. RESULTS Different from the uncomplicated and healthy groups, unbalanced fecal bacterium ratio existed in patients with NAS after LT. The results showed that NAS patients were associated with a decrease of Firmicutes and Bacteroidetes and an increase of Proteobacteria at the phylum level, with the proportion-ratio imbalance between potential pathogenic families including Enterococcaceae, Streptococcaceae, Enterobacteriaceae, Pseudomonadaceae and dominant families including Bacteroidaceae. CONCLUSION The compositional shifts of the increase of potential pathogenic bacteria as well as the decrease of dominant bacteria might contribute to the incidence of NAS.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Feng-Gang Ren
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Peng Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Hong-Ke Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Hao-Yang Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Zhe Feng
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Xue-Ming Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Xiao-Gang Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Rong-Qian Wu
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| | - Yi Lv
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Institute of Advanced Surgical Technology and Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
- Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, China
| |
Collapse
|