1
|
Santamarina AB, Filho VN, de Freitas JA, Franco LAM, Martins RC, Fonseca JV, Orellana Turri JA, Hufnagel MT, Demarque DP, da Silva BFRB, Gusmão AF, Olivieri EHR, de Souza E, de Souza EA, Otoch JP, Pessoa AFM. Nutraceutical Blends Promote Weight Loss, Inflammation Reduction, and Better Sleep: The Role of Faecalibacterium prausnitzii in Overweight Adults-A Double-Blind Trial. Mol Nutr Food Res 2025:e202400806. [PMID: 39981988 DOI: 10.1002/mnfr.202400806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/02/2024] [Accepted: 01/14/2025] [Indexed: 02/22/2025]
Abstract
This study explores the effects of a nutraceutical blend with prebiotics, β-glucans, essential minerals, and silymarin on gut microbiota, inflammation, and sleep quality in obesity through microbiota reshaping and metabolic improvements over 90 days. A double-blind, randomized trial was conducted on 77 participants divided into two groups receiving either a standard nutraceutical blend (NSupple) or a silymarin-enriched blend (NSupple_Silybum). Fecal and plasma samples were collected at baseline and post-supplementation for gut microbiota, metabolic, and inflammatory marker analysis. The results showed a reduction in body weight, waist-to-height ratio, total cholesterol, and fractions in the NSupple_Silybum group. There was a dysbiosis recovery shown by the increase in beneficial gut bacteria, such as Lentisphaerae phylum, Lactobacillus and Faecalibacterium genera, and Faecalibacterium prausnitzii in the NSupple group, with a concurrent reduction in Adlercreutzia and Sutterella in the NSupple_Silybum group. Both groups demonstrated improved inflammatory profiles by the reduced TNF-α/IL-10 ratio, reduced cortisol levels, and reduced Firmicutes/Bacteroides ratio. Additionally, improvements in sleep quality were associated with reductions in pro-inflammatory cytokines and improved microbiota composition. The nutraceutical blend reshaped gut microbiota, enhanced anti-inflammatory species, and improved metabolic and sleep parameters, highlighting its potential as a nutritional strategy for managing obesity and reducing inflammation.
Collapse
Affiliation(s)
- Aline Boveto Santamarina
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
| | - Victor Nehmi Filho
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
| | - Jéssica Alves de Freitas
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
| | - Lucas Augusto Moysés Franco
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Roberta Cristina Martins
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Joyce Vanessa Fonseca
- Laboratório de Investigação Médica em Protozoologia, Bacteriologia e Resistência Antimicrobiana (LIM-49), Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - José Antônio Orellana Turri
- Departamento de Ginecologia e Obstetrícia, Universidade de São Paulo Faculdade de Medicina, São Paulo, Brazil
| | - Mariana Tedesco Hufnagel
- Laboratório de Farmacognosia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Daniel Pecoraro Demarque
- Laboratório de Farmacognosia, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Bruna Fernanda Rio Branco da Silva
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório Interdisciplinar em Fisiologia e Exercício, Universidade Federal de São Paulo (UNIFESP), Santos, Brazil
| | | | | | - Erica de Souza
- Ambulatório Médico Monte Azul, Associação Comunitária Monte Azul, São Paulo, Brazil
| | - Esther Alves de Souza
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
| | - José Pinhata Otoch
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
- Hospital Universitário da Universidade de São Paulo, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Ana Flávia Marçal Pessoa
- Laboratório de Produtos e Derivados Naturais, Laboratório de Investigação Médica-26 (LIM-26), Departamento de Cirurgia, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Pesquisa e Desenvolvimento, Efeom Nutrição S/A, São Paulo, Brazil
- Laboratório de Parasitologia Médica (LIM-46), Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Li Y, Gong Q, He W, Ke J. Dietary intake of live microbes and its association with frailty in older adults: a NHANES analysis (1999-2018). BMC Geriatr 2025; 25:91. [PMID: 39934741 DOI: 10.1186/s12877-025-05725-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/21/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Diet plays a crucial part in maintaining a healthy body, and microbes, as an essential dietary component, have attracted more attention in recent years. In this study, we will explore the link between dietary intake of live microbes and frailty in the elderly. METHODS Older participants from the 1999-2018 National Health and Nutrition Examination Survey (NHANES) who were not less than 60 years of age were enrolled in this research. Participants' dietary microbe intake was assessed by a self-report questionnaire. Participants were stratified into low, medium, and high intake groups according to their consumption of foods with varying microbial content. The frailty index was assessed by 49 frailty indicators. Frailty was defined as a frailty index > 0.21. Logistic regression was used to analyze the link between dietary intake of live microbes and frailty in older adults. RESULTS A total of 15,179 older adults' basic information was collected for this study. The frailty index was higher than 0.21 in 32.8% of participants. Older adults with low, medium, and high dietary microbe intake accounted for 33.5%, 47.0%, and 19.5%, respectively. In models adjusted for confounders, the prevalence of frailty was lower in participants with the medium (OR = 0.825, 95%CI: 0.749-0.908) and high (OR = 0.779, 95%CI: 0.679-0.894) dietary microbe intake groups, compared with in participants with the lowest intake group. The RCS analysis revealed a significant non-linear association between dietary live microbe intake and frailty prevalence, with the strongest reduction observed below an inflection point of 161.82 (P for non-linearity < 0.05). In addition, stratified analyses did not reveal interactions between the study variables. CONCLUSION This study demonstrates a negative and non-linear association between dietary live microbe intake and frailty in older adults. These findings provide evidence of a potential link between live microbe intake and frailty, warranting further longitudinal and interventional studies to explore this relationship and its implications for healthy aging.
Collapse
Affiliation(s)
- Yuan Li
- Department of Comprehensive Rehabilitation, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350003, China
| | - Qin Gong
- Department of Comprehensive Rehabilitation, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350003, China
| | - Weixiu He
- Department of Orthopedic Rehabilitation, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, 350003, China
| | - Junhua Ke
- Department of Geriatric Rehabilitation, Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, 13 Hudong branch Road, Fuzhou, 350003, Fujian, China.
| |
Collapse
|
3
|
Damianos J, Abdelnaem N, Camilleri M. Gut Goo: Physiology, Diet, and Therapy of Intestinal Mucus and Biofilms in Gastrointestinal Health and Disease. Clin Gastroenterol Hepatol 2025; 23:205-215. [PMID: 39426645 PMCID: PMC11761393 DOI: 10.1016/j.cgh.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
The gastrointestinal tract has remarkable capacity to withstand considerable insults from exposure to abrasive food particles, chemicals, allergens, and pathogenic microbes. Maintaining a robust epithelial barrier sequesters these potentially harmful substances in the lumen, preventing absorption into the systemic circulation. Normal functioning of this barrier is central in diverse physiological processes including digestion, immunity, inflammation, and gut-brain signaling. One crucial component of the barrier is the mucus layer covering the epithelium. There is increased appreciation of the importance of mucus in maintenance of the gut barrier, and how dysregulation of the mucus layer contributes to several common gastrointestinal pathologies. This manuscript reviews the physical and chemical properties of mucus, its maintenance and turnover, and its role in maintaining gut barrier integrity. The dynamic interactions of the mucus layer within the gut ecosystem are illustrated by highlighting how a weakened mucus layer or defective mucus production facilitate pathogenic microbial colonization and mucosal biofilm formation. These may potentially contribute to the pathogenesis of gastrointestinal diseases such as inflammatory bowel diseases or result in secretion and mucosal damage and inflammation in bile acid diarrhea. A final goal is to review how certain dietary factors, especially low-fiber diets and emulsifiers common in Western diets, can harm the mucus layer. This report summarizes evidence from preclinical and human studies that document damage to the mucus layer, and reviews approaches, including diets and probiotics, that promote a healthy mucus layer and break down pathogenic biofilms, thereby potentially preventing and/or treating gastrointestinal diseases that impact mucosal integrity.
Collapse
Affiliation(s)
- John Damianos
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Nada Abdelnaem
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
4
|
Liu M, Fan G, Meng L, Yang K, Liu H. New perspectives on microbiome-dependent gut-brain pathways for the treatment of depression with gastrointestinal symptoms: from bench to bedside. J Zhejiang Univ Sci B 2025; 26:1-25. [PMID: 39428337 PMCID: PMC11735910 DOI: 10.1631/jzus.b2300343] [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: 07/09/2023] [Accepted: 08/29/2023] [Indexed: 10/22/2024]
Abstract
Patients with depression are more likely to have chronic gastrointestinal (GI) symptoms than the general population, but such symptoms are considered only somatic symptoms of depression and lack special attention. There is a chronic lack of appropriate diagnosis and effective treatment for patients with depression accompanied by GI symptoms, and studying the association between depression and GI disorders (GIDs) is extremely important for clinical management. There is growing evidence that depression is closely related to the microbiota present in the GI tract, and the microbiota-gut-brain axis (MGBA) is creating a new perspective on the association between depression and GIDs. Identifying and treating GIDs would provide a key opportunity to prevent episodes of depression and may also improve the outcome of refractory depression. Current studies on depression and the microbially related gut-brain axis (GBA) lack a focus on GI function. In this review, we combine preclinical and clinical evidence to summarize the roles of the microbially regulated GBA in emotions and GI function, and summarize potential therapeutic strategies to provide a reference for the study of the pathomechanism and treatment of depression in combination with GI symptoms.
Collapse
Affiliation(s)
- Menglin Liu
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Brain Disease Regional Diagnosis and Treatment Center, Zhengzhou 450000, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
| | - Genhao Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
- The First Affiliated Hospital of Zhengzhou University, Department of Geriatrics, Zhengzhou 450052, China
| | - Lingkai Meng
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Kuo Yang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Huayi Liu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China.
| |
Collapse
|
5
|
Yu QX, Wang DD, Dong PJ, Zheng LH. Probiotics Combined With Trimebutine for the Treatment of Irritable Bowel Syndrome Patients: A Systematic Review and Meta-Analysis. J Gastroenterol Hepatol 2025. [PMID: 39780334 DOI: 10.1111/jgh.16858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE This study aimed to assess the efficacy and safety of probiotics combined with trimebutine in the treatment of irritable bowel syndrome (IBS), addressing an important gap in current treatment strategies. METHODS Randomized controlled trials (RCTs) of trimebutine combined with probiotics for the treatment of IBS were collected from various databases. All retrieved articles were screened and assessed for quality. The methodological quality of the included studies was assessed following the guidelines recommended by the Cochrane Collaboration. The meta-analysis of the included studies was conducted using RevMan 5.3 software. RESULTS A total of 37 RCTs involving 4360 participants were included in this study. Among them, the treatment group consisted of 2177 participants, and the control group consisted of 2183 participants. The results showed that the overall efficacy of trimebutine combined with probiotics in the treatment of IBS was significantly higher than that of trimebutine alone (odds ratio [OR] = 5.09, 95 % confidence interval [CI] [4.19, 6.20], p < 0.00001). The effective rate in the combination therapy group was 93.5 % compared with 73.8 % in the trimebutine alone group. The safety profile was favorable, with adverse event rates of 1.75 % and 1.69 % in the combination and monotherapy groups, respectively. The most common adverse events were mild and included dry mouth, nausea and dizziness. No serious adverse events were reported in either group. Subgroup analysis based on the type of probiotic intervention showed that combination use was better than trimebutine alone, and the differences between each subgroup were statistically significant. Combination use of compound Lactobacillus capsules had the best effect (OR = 16.03, 95 % CI [4.57, 56.21], p < 0.0001]. These results highlight the potential role of strain-specific benefits in IBS treatment and suggest that probiotic strain selection may significantly influence treatment outcomes. CONCLUSIONS The combination of trimebutine and probiotics is more effective in the treatment of IBS compared with trimebutine alone. CLINICAL TRIAL REGISTRATION https://www.crd.york.ac.uk/prospero/, identifier: CRD42024516044.
Collapse
Affiliation(s)
- Qiu-Xiang Yu
- Department of Proctology, China-Japan Friendship Hospital, Beijing, China
| | - Dong-Dong Wang
- Department of Surgical, Tumote Right Banner Hospital, Baotou, Inner Mongolia, China
| | - Peng-Ju Dong
- Department of Surgical, Tumote Right Banner Hospital, Baotou, Inner Mongolia, China
| | - Li-Hua Zheng
- Department of Proctology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
6
|
Radoš L, Golčić M, Mikolašević I. The Relationship Between the Modulation of Intestinal Microbiota and the Response to Immunotherapy in Patients with Cancer. Biomedicines 2025; 13:96. [PMID: 39857680 PMCID: PMC11761299 DOI: 10.3390/biomedicines13010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The intestinal microbiota is an important part of the human body, and its composition can affect the effectiveness of immunotherapy. In the last few years, the modulation of intestinal microbiota in order to improve the effectiveness of immunotherapy has become a current topic in the scientific community, but there is a lack of research in this area. In this review, the goal was to analyze the current relevant literature related to the modulation of intestinal microbiota and the effectiveness of immunotherapy in the treatment of cancer. The effects of antibiotics, probiotics, diet, and fecal microbial transplantation were analyzed separately. It was concluded that the use of antibiotics, especially broad-spectrum types or larger quantities, causes dysbiosis of the intestinal microbiota, which can reduce the effectiveness of immunotherapy. While dysbiosis could be repaired by probiotics and thus improve the effectiveness of immunotherapy, the use of commercial probiotics without evidence of intestinal dysbiosis has not yet been sufficiently tested to confirm its safety for cancer for immunotherapy-treated cancer patients. A diet consisting of sufficient amounts of fiber, as well as a diet with higher salt content positively correlates with the success of immunotherapy. Fecal transplantation is a safe and realistic adjuvant option for the treatment of cancer patients with immunotherapy, but more clinical trials are necessary. Modulating the microbiota composition indeed changes the effectiveness of immunotherapy, but in the future, more human studies should be organized to precisely determine the types and procedures of microbiota modulation.
Collapse
Affiliation(s)
- Laura Radoš
- Department for Emergency Medicine of Primorsko-Goranska County, 51000 Rijeka, Croatia;
| | - Marin Golčić
- Clinic for Tumors, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia;
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Mikolašević
- Clinic for Tumors, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia;
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
7
|
Quigley EMM, Shanahan F. Probiotics in Health Care: A Critical Appraisal. Annu Rev Med 2025; 76:129-141. [PMID: 39527719 DOI: 10.1146/annurev-med-042423-042315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Consumption of probiotic products continues to increase, perhaps driven by an interest in gut health. However, the field is filled with controversy, inconsistencies, misuse of terminology, and poor communication. While the probiotic concept is biologically plausible and in some cases mechanistically well established, extrapolation of preclinical results to humans has seldom been proven in well-conducted clinical trials. With noteworthy exceptions, clinical guidance has often been derived not from large, adequately powered clinical trials but rather from comparisons of disparate, small studies with insufficient power to identify the optimal strain. The separation of probiotics from live biotherapeutic products has brought some clarity from a regulatory perspective, but in both cases, consumers should expect scientific rigor and strong supporting evidence for health claims.
Collapse
Affiliation(s)
- Eamonn M M Quigley
- Lynda K. and David M. Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, Texas, USA
| | - Fergus Shanahan
- Department of Medicine and Alimentary Pharmabiotic Centre, Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland;
| |
Collapse
|
8
|
Duman H, Karav S. Fiber and the gut microbiome and its impact on inflammation. NUTRITION IN THE CONTROL OF INFLAMMATION 2025:51-76. [DOI: 10.1016/b978-0-443-18979-1.00004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Zheng S, Lou Y, Zhang J, Wang Y, Lv L. Association of dietary live microbe intake with all-cause and cardiovascular mortality in an older population: Evidence from NHANES 2003-2018. Arch Gerontol Geriatr 2024; 131:105741. [PMID: 39756187 DOI: 10.1016/j.archger.2024.105741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/12/2024] [Accepted: 12/30/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND The relationship between dietary live microbe intake, non-dietary prebiotics/probiotics, and mortality in older adults remains unclear. METHODS Participants from the National Health and Nutrition Examination Survey 2003-2018 were included. Participants were categorized into three groups based on estimated live microbe intake: low, medium, and high. Additionally, they were divided by their consumption of foods with medium or high microbial content. Text mining was employed to identify the use of non-dietary prebiotics and probiotics by analyzing the names and ingredients of dietary supplements and drugs. Associations between live microbe intake, non-dietary prebiotics/probiotics, and all-cause or cardiovascular mortality were assessed using Kaplan-Meier survival curves and Cox regression models. Inverse Probability of Treatment Weighting was used for sensitivity analysis RESULT: A total of 7882 participants were included in the study. During a mean follow-up of 8.08 years, all-cause mortality was 18 % lower in older adults with high dietary live microbe intake in fully adjusted models (HR, 0.82; 95 %CI 0.70-0.96), and cardiovascular mortality was reduced by 23 % (HR, 0.77; 95 %CI 0.61-0.98). The risk of all-cause mortality was reduced by 21 % in the G3 group compared with the G1 group (HR, 0.79; 95 % CI 0.69-0.89) and a 29 % reduction in the risk of cardiovascular disease-specific death (HR, 0.71;95 %CI 0.59-0.86). In addition, we also observed that nondietary prebiotic and probiotic supplement intake was also associated with a reduced risk of mortality in an older US population. CONCLUSION In older U.S. adults, higher dietary live microorganisms and non-dietary probiotics/prebiotics intake was associated with a reduced risk of all-cause and cardiovascular mortality.
Collapse
Affiliation(s)
- Shuang Zheng
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China; Department of Pathology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yanqing Lou
- Department of Pathology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China; Department of Pathology, The Second People's Hospital of Fuyang, Hangzhou, Zhejiang Province, China
| | - Jiali Zhang
- Department of Pathology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu Wang
- Department of Pathology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China; Department of Pathology, Maternal and child health hospital of Changxing County, Huzhou, Zhejiang Province, China
| | - Lugang Lv
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
10
|
Hu D, Wu X, Song P, Hou M, Pan L, Yang X, Sun Q, Ni Y. Dietary Supplementation with Multi-strain Probiotic Formulation (Bifidobacterium B8101, Lactobacillus L8603, Saccharomyces bayanus S9308, and Enterococcus SF9301), Betaine or their Combination Promotes Growth Performance Via Improving Intestinal Development in Broilers. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10434-w. [PMID: 39715924 DOI: 10.1007/s12602-024-10434-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
This study aimed to investigate the effect of a multi-strain probiotic (Bifidobacterium B8101, Lactobacillus L8603, Saccharomyces bayanus S9308, Enterococcus SF9301), betaine, and their combination on intestinal epithelial development and growth performance in broilers. A total of 2800 one-day-old Ross 308 chickens were randomly divided into four groups: control (Ctrl) fed with a basal diet, multi-strain probiotic (Pb) group fed with basal diet + 100 mg/day/bird probiotic (1-14 d), betaine (Bet) fed with basal diet + 0.1% betaine (1-35 d), and a combination (Pb&Bet) fed with both probiotics and betaine. Each group was set with 10 replicates, with 70 chickens in each replicate. Result showed that betaine significantly increased the body weight (BW) of broilers at 14 d of age and decreased the feed conversion ratio (FCR) from 1 to 14 d of age. Multi-strain probiotic significantly increased BW at 21 and 35 d of age, and decreased FCR from 15 to 21 d of age. Pb&Bet group exhibited a higher BW but lower FCR than Ctrl throughout entire experiment (p < 0.05). Consistently, Pb&Bet group had a higher pectoralis muscle weight, fiber diameter and cross-sectional area compared to Ctrl group (p < 0.05). Pb&Bet group also increased villus height and the ratio of villus height to crypt depth (V/C) in duodenum at both 21 d and 35 d of age. Moreover, at 35 d of age, the mucin 2 (MUC2) expression in duodenum and jejunum was significantly increased in Pb&Bet group, and the interaction of betaine and probiotics was observed on claudin 1 (CLDN1), zonula occludens 1 (ZO1), and junctional adhesion molecule 2 (JAM2) expression in the ileum (p < 0.05). In conclusion, the combination of probiotics and betaine shows better potential for improving growth performance and promoting small intestinal development.
Collapse
Affiliation(s)
- Dan Hu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoting Wu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Pin Song
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Manman Hou
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Li'an Pan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaoran Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Qinwei Sun
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
11
|
Al-Beltagi M. Nutritional management and autism spectrum disorder: A systematic review. World J Clin Pediatr 2024; 13:99649. [PMID: 39654662 PMCID: PMC11572612 DOI: 10.5409/wjcp.v13.i4.99649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/21/2024] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) presents unique challenges related to feeding and nutritional management. Children with ASD often experience feeding difficulties, including food selectivity, refusal, and gastrointestinal issues. Various interventions have been explored to address these challenges, including dietary modifications, vitamin supplementation, feeding therapy, and behavioral interventions. AIM To provide a comprehensive overview of the current evidence on nutritional management in ASD. We examine the effectiveness of dietary interventions, vitamin supplements, feeding therapy, behavioral interventions, and mealtime practices in addressing the feeding challenges and nutritional needs of children with ASD. METHODS We systematically searched relevant literature up to June 2024, using databases such as PubMed, PsycINFO, and Scopus. Studies were included if they investigated dietary interventions, nutritional supplements, or behavioral strategies to improve feeding behaviors in children with ASD. We assessed the quality of the studies and synthesized findings on the impact of various interventions on feeding difficulties and nutritional outcomes. Data extraction focused on intervention types, study designs, participant characteristics, outcomes measured, and intervention effectiveness. RESULTS The review identified 316 studies that met the inclusion criteria. The evidence indicates that while dietary interventions and nutritional supplements may offer benefits in managing specific symptoms or deficiencies, the effectiveness of these approaches varies. Feeding therapy and behavioral interventions, including gradual exposure and positive reinforcement, promise to improve food acceptance and mealtime behaviors. The findings also highlight the importance of creating supportive mealtime environments tailored to the sensory and behavioral needs of children with ASD. CONCLUSION Nutritional management for children with ASD requires a multifaceted approach that includes dietary modifications, supplementation, feeding therapy, and behavioral strategies. The review underscores the need for personalized interventions and further research to refine treatment protocols and improve outcomes. Collaborative efforts among healthcare providers, educators, and families are essential to optimize this population's nutritional health and feeding practices. Enhancing our understanding of intervention sustainability and long-term outcomes is essential for optimizing care and improving the quality of life for children with ASD and their families.
Collapse
Affiliation(s)
- Mohammed Al-Beltagi
- Department of Pediatric, Faculty of Medicine, Tanta University, Tanta 31511, Alghrabia, Egypt
- Department of Pediatric, University Medical Center, King Abdulla Medical City, Arabian Gulf University, Manama 26671, Bahrain
| |
Collapse
|
12
|
Deehan EC, Al Antwan S, Witwer RS, Guerra P, John T, Monheit L. Revisiting the Concepts of Prebiotic and Prebiotic Effect in Light of Scientific and Regulatory Progress-A Consensus Paper From the Global Prebiotic Association. Adv Nutr 2024; 15:100329. [PMID: 39481540 PMCID: PMC11616045 DOI: 10.1016/j.advnut.2024.100329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/18/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
The term prebiotic has been used for almost 3 decades and has undergone numerous updates over the years. The scientific literature reveals that despite continuous efforts to establish a globally unified definition to guide jurisdictional regulations and product innovations, ambiguity continues to surround the terms prebiotic and prebiotic effect, leading to products that lack in full regulatory adherence being marketed worldwide. Thus, to reflect the current state of scientific research and knowledge and for the continuous advancement of the category, an update to the current prebiotic definition is warranted. This update includes removing the term selectivity, considering additional locations of action besides the gut, highlighting prebiotic performance benefits such as cognitive and athletic, and providing a clear standalone definition for prebiotic effect. The Global Prebiotic Association (GPA) is a leading information and industry hub committed to raising awareness about prebiotics, their emerging and well-established health benefits, and prebiotic product integrity and efficacy. In this position paper, GPA builds on previous prebiotic definitions to propose the following expanded definition for prebiotic: "a compound or ingredient that is utilized by the microbiota producing a health or performance benefit." In addition to prebiotic, GPA also defines prebiotic effect as "a health or performance benefit that arises from alteration of the composition and/or activity of the microbiota, as a direct or indirect result of the utilization of a specific and well-defined compound or ingredient by microorganisms." With these 2 definitions, GPA aims to paint a clearer picture for the term prebiotic, and by incorporating an industry point of view, these updated definitions may be used alongside current scientific and regulatory perspectives to move the category forward.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE, United States; Nebraska Food for Health Center, University of Nebraska, Lincoln, NE, United States; Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States.
| | | | - Rhonda S Witwer
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; ADM, Decatur, IL, United States
| | - Paula Guerra
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada.
| | - Tania John
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; SGS Nutrasource, Guelph, Ontario, Canada
| | - Len Monheit
- Scientific & Technical Committee, Global Prebiotic Association, Chicago, IL, United States; Global Prebiotic Association/Industry Transparency Center, Chicago, IL, United States
| |
Collapse
|
13
|
Waliaula PK, Kiarie EG, Diarra MS. Predisposition factors and control strategies of avian pathogenic Escherichia coli in laying hens. Front Vet Sci 2024; 11:1474549. [PMID: 39559543 PMCID: PMC11571327 DOI: 10.3389/fvets.2024.1474549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/23/2024] [Indexed: 11/20/2024] Open
Abstract
Shift in laying hens housing from conventional cage-based systems to alternatives has impacted their health and performance. Microorganisms colonize young chick in the early stages of their physiological and immune development. These colonizing microbes originate from parent and the environment. Escherichia coli is among the normal gut colonizing bacteria however, some E. coli strains known as avian pathogenic E. coli (APEC), cause local or systemic infections (colibacillosis) responsible of significant economic losses to the poultry industry. Potential APEC strains and other poultry gut microbiota are influenced by several factors such as housing system, and the use of feed additives (prebiotics, probiotics, symbiotic, among others). This review will discuss the status of pullets and layers immunity, gut health, and predisposing factors of colibacillosis. Dietary interventions and some colibacillosis mitigation strategies in pullets and laying hens are reviewed and discussed. With the development of sequencing technologies and the use of feed additives as alternatives to antibiotics, future studies need to understand some of the complex associations between the feed additives, the rearing environment, and their selective pressure on gut microbiota, including E. coli, and their impacts on immune development in pullets and hens.
Collapse
Affiliation(s)
- Paul K. Waliaula
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Elijah G. Kiarie
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| | - Moussa S. Diarra
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| |
Collapse
|
14
|
Yu J, Li L, Tao X, Chen Y, Dong D. Metabolic interactions of host-gut microbiota: New possibilities for the precise diagnosis and therapeutic discovery of gastrointestinal cancer in the future-A review. Crit Rev Oncol Hematol 2024; 203:104480. [PMID: 39154670 DOI: 10.1016/j.critrevonc.2024.104480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/20/2024] Open
Abstract
Gastrointestinal (GI) cancer continues to pose a significant global health challenge. Recent advances in our understanding of the complex relationship between the host and gut microbiota have shed light on the critical role of metabolic interactions in the pathogenesis and progression of GI cancer. In this study, we examined how microbiota interact with the host to influence signalling pathways that impact the formation of GI tumours. Additionally, we investigated the potential therapeutic approach of manipulating GI microbiota for use in clinical settings. Revealing the complex molecular exchanges between the host and gut microbiota facilitates a deeper understanding of the underlying mechanisms that drive cancer development. Metabolic interactions hold promise for the identification of microbial signatures or metabolic pathways associated with specific stages of cancer. Hence, this study provides potential strategies for the diagnosis, treatment and management of GI cancers to improve patient outcomes.
Collapse
Affiliation(s)
- Jianing Yu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, China
| | - Lu Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Yanwei Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
15
|
Iatcu OC, Hamamah S, Covasa M. Harnessing Prebiotics to Improve Type 2 Diabetes Outcomes. Nutrients 2024; 16:3447. [PMID: 39458444 PMCID: PMC11510484 DOI: 10.3390/nu16203447] [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: 09/18/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The gut microbiota, a complex ecosystem of microorganisms in the human gastrointestinal tract (GI), plays a crucial role in maintaining metabolic health and influencing disease susceptibility. Dysbiosis, or an imbalance in gut microbiota, has been linked to the development of type 2 diabetes mellitus (T2DM) through mechanisms such as reduced glucose tolerance and increased insulin resistance. A balanced gut microbiota, or eubiosis, is associated with improved glucose metabolism and insulin sensitivity, potentially reducing the risk of diabetes-related complications. Various strategies, including the use of prebiotics like inulin, fructooligosaccharides, galactooligosaccharides, resistant starch, pectic oligosaccharides, polyphenols, β-glucan, and Dendrobium officinale have been shown to improve gut microbial composition and support glycemic control in T2DM patients. These prebiotics can directly impact blood sugar levels while promoting the growth of beneficial bacteria, thus enhancing glycemic control. Studies have shown that T2DM patients often exhibit a decrease in beneficial butyrate-producing bacteria, like Roseburia and Faecalibacterium, and an increase in harmful bacteria, such as Escherichia and Prevotella. This review aims to explore the effects of different prebiotics on T2DM, their impact on gut microbiota composition, and the potential for personalized dietary interventions to optimize diabetes management and improve overall health outcomes.
Collapse
Affiliation(s)
- Oana C. Iatcu
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
| | - Sevag Hamamah
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
- Department of Internal Medicine, Scripps Mercy Hospital, San Diego, CA 92103, USA
| | - Mihai Covasa
- Department of Biomedical Sciences, College of Medicine and Biological Science, University of Suceava, 720229 Suceava, Romania;
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA;
| |
Collapse
|
16
|
Que M, Li S, Xia Q, Li X, Luo X, Zhan G, Luo A. Microbiota-gut-brain axis in perioperative neurocognitive and depressive disorders: Pathogenesis to treatment. Neurobiol Dis 2024; 200:106627. [PMID: 39111702 DOI: 10.1016/j.nbd.2024.106627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024] Open
Abstract
An increasing number of people undergo anesthesia and surgery. Perioperative neurocognitive and depressive disorders are common central nervous system complications with similar pathogeneses. These conditions pose a deleterious threat to human health and a significant societal burden. In recent years, numerous studies have focused on the role of the gut microbiota and its metabolites in the central nervous system via the gut-brain axis. Its involvement in perioperative neurocognitive and depressive disorders has attracted considerable attention. This review aimed to elucidate the role of the gut microbiota and its metabolites in the pathogenesis of perioperative neurocognitive and depressive disorders, as well as the value of targeted interventions and treatments.
Collapse
Affiliation(s)
- Mengxin Que
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Xia
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaofeng Zhan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ailin Luo
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health; Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Chen Y, Ni H, Zhang H. Exploring the relationship between live microbe intake and obesity prevalence in adults. Sci Rep 2024; 14:21724. [PMID: 39289456 PMCID: PMC11408724 DOI: 10.1038/s41598-024-72961-4] [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: 04/19/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Obesity has become a global health problem. In recent years, the influence of dietary microbes in the obese population has attracted the attention of scholars. Our study aimed to investigate the link between live microbe intake and obesity in adults. Participants (aged over 20 years) for this study were from the 1999-2018 National Health and Nutrition Examination Survey (NHANES). Participants were categorised into low, medium and high dietary live microbe intake groups. Linear regression was used to analyse the link between live microbe intake and body mass index (BMI) and waist circumference (WC). Logistic regression was used to analyse the link between live microbe intake and obesity and abdominal obesity prevalence. Restricted cubic spline curves (RCS) were used to check whether there was a non-linear relationship between live microbe intake and obesity. A total of 42,749 participants were included in this study and the number of obese reached 15,463. We found that live microbe intake was negatively linked to BMI and WC. In models adjusted for all confounders, the high live microbe intake group had lower obesity (OR = 0.812, 95%CI: 0.754-0.873) and abdominal obesity prevalence (OR = 0.851, 95%CI: 0.785-0.923) than the lowest intake group. Upon further quantification of live microbe intake, we found similar results. RCS analyses showed that live microbe intake was nonlinearly negatively correlated with BMI, WC, obesity, and abdominal obesity prevalence (P for non-linearity < 0.05). This study preliminarily reveals a negative link between live microbe intake and obesity in adults.
Collapse
Affiliation(s)
- Yuting Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54, Youdian Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| | - Haixiang Ni
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54, Youdian Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| | - Hong Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Zhejiang Chinese Medical University, No.54, Youdian Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
18
|
Zhu J, Liu X, Liu N, Zhao R, Wang S. Lactobacillus plantarum alleviates high-fat diet-induced obesity by altering the structure of mice intestinal microbial communities and serum metabolic profiles. Front Microbiol 2024; 15:1425764. [PMID: 39282560 PMCID: PMC11392860 DOI: 10.3389/fmicb.2024.1425764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Obesity, which is always accompanied by disorders of lipid metabolism and dysbiosis of the gut microbiota, has become a global epidemic recognised by the World Health Organisation, necessitating innovative strategies and a globally accepted agreement on treating obesity and its related complications. Probiotics, as major active ingredients in many foods, offer potential as biological treatments for obesity prevention and management. Lactobacillus plantarum (L. plantarum) possesses a wide range of biological activities and is widely used to alleviate and ameliorate various diseases. This research demonstrated that Lactobacillus plantarum reduces the weight increase and fat build-up caused by a high-fat diet (HFD) in mice, while also improving glucose tolerance and insulin sensitivity in obese mice. Results indicated that L. plantarum effectively controlled the intestinal microbial community's structure, counteracted disruptions in gut flora caused by HFD, normalized the Firmicutes to Bacteroidota ratio (F/B), and decreased the prevalence of detrimental bacteria Desulfovibrio and Clostridia. Serum metabolomics findings indicate notable alterations in serum metabolites across various groups, notably the increased levels of Isoprothiolane and Inosine, key regulators of lipid metabolism disorders and enhancers of fat burning. These differential metabolites were mainly enriched in unsaturated fatty acid biosynthesis, sulfur metabolism, fatty acid biosynthesis, and purine metabolism. Consequently, we propose that L. plantarum has the potential to alter the gut microbial community's composition, positioning it as a promising option for obesity therapy.
Collapse
Affiliation(s)
- Junwen Zhu
- Department of Cardiology, The First People's Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling, China
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Xueying Liu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Naiyuan Liu
- Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, China
| | - Ruochi Zhao
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| | - Shuangshuang Wang
- Department of Cardiology, The First People's Hospital of Wenling, Wenling Hospital of Wenzhou Medical University, Wenling, China
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Affiliated First Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
19
|
Ihara E, Manabe N, Ohkubo H, Ogasawara N, Ogino H, Kakimoto K, Kanazawa M, Kawahara H, Kusano C, Kuribayashi S, Sawada A, Takagi T, Takano S, Tomita T, Noake T, Hojo M, Hokari R, Masaoka T, Machida T, Misawa N, Mishima Y, Yajima H, Yamamoto S, Yamawaki H, Abe T, Araki Y, Kasugai K, Kamiya T, Torii A, Nakajima A, Nakada K, Fukudo S, Fujiwara Y, Miwa H, Kataoka H, Nagahara A, Higuchi K. Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023. Digestion 2024; 105:480-497. [PMID: 39197422 PMCID: PMC11633876 DOI: 10.1159/000541121] [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: 06/21/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
The Japan Gastroenterological Association (JGA) published the first version of clinical guidelines for chronic diarrhea 2023. These guidelines describe the definition, classification, diagnostic criteria, diagnostic testing methods, epidemiology, pathophysiology, and treatment of chronic diarrhea, and provide flowcharts for the diagnosis and treatment of chronic diarrhea based on the latest evidence. Treatment for chronic diarrhea begins by distinguishing secondary chronic constipation with a clear etiology, such as drug-induced diarrhea, food-induced diarrhea, systemic disease-associated diarrhea, infection-associated diarrhea, organic disease-associated diarrhea, and bile acid diarrhea. The first line of treatment for chronic diarrhea in the narrow sense, defined in these guidelines as functional diarrhea in routine medical care, is lifestyle modification and dietary therapy. The first medicines to be considered for oral treatment are probiotics for regulating the gut microbiome and anti-diarrheals. Other medications, such as 5HT3 receptor antagonists, anticholinergics, Kampo medicine, psychotherapy, antibiotics, bulking agents, adrenergic agonists, and somatostatin analogs, lack sufficient evidence for their use, highlighting a challenge for future research. This Clinical Guidelines for Chronic Diarrhea 2023, which provides the best clinical strategies for treating chronic diarrhea in Japan, will also be useful for medical treatment worldwide.
Collapse
Affiliation(s)
- Eikichi Ihara
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriaki Manabe
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hidenori Ohkubo
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Naotaka Ogasawara
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Haruei Ogino
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Kazuki Kakimoto
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Motoyori Kanazawa
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hidejiro Kawahara
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Chika Kusano
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Shiko Kuribayashi
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Akinari Sawada
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Tomohisa Takagi
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Shota Takano
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Toshihiko Tomita
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Toshihiro Noake
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Mariko Hojo
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Ryota Hokari
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Tatsuhiro Masaoka
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Tomohiko Machida
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Noboru Misawa
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Yoshiyuki Mishima
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hiroshi Yajima
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Sayuri Yamamoto
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hiroshi Yamawaki
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Tatsuya Abe
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Yasumi Araki
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Kunio Kasugai
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Takeshi Kamiya
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Akira Torii
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Atsushi Nakajima
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Koji Nakada
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Shin Fukudo
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Yasuhiro Fujiwara
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hiroto Miwa
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Hiromi Kataoka
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Akihito Nagahara
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| | - Kazuhide Higuchi
- Guidelines Committee for Creating and Evaluating the “Evidence-Based Clinical Guidelines for Chronic Diarrhea 2023”, The Japanese Gastroenterological Association, Bunkyo-ku, Japan
| |
Collapse
|
20
|
Wang J, Jiang M, Li X, Ye Y, Xie Y, Wu T, Chen Y, Yu H, Wu H, Yang Z, Zhou E. Inulin Supplementation Alleviates Ochratoxin A-Induced Kidney Injury through Modulating Intestinal Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18682-18696. [PMID: 39135376 DOI: 10.1021/acs.jafc.4c04382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Ochratoxin A (OTA) is a prevalent mycotoxin found in feed that causes significant kidney injury in animals. Further investigation was needed to devise strategies for treating OTA-induced kidney damage through the gut-kidney axis. Evidence indicates the crucial role of intestinal microbiota in kidney damage development. Inulin, a dietary fiber, protects kidneys by modulating intestinal microbiota and promoting short-chain fatty acid (SCFA) production. However, its precise mechanism in OTA-induced kidney damage remained unclear. In this study, chickens were orally administered OTA and inulin for 2 weeks to investigate inulin's effects on OTA-induced kidney damage and underlying mechanisms. The alteration of intestinal microbiota, SCFAs contents, and SCFA receptors was further analyzed. Results demonstrated that inulin supplementation influenced intestinal microbiota, increased SCFAs production, and mitigated OTA-induced kidney damage in chickens. The importance of microbiota in mediating inulin's renal protection was further confirmed by antibiotic and fecal microbiota transplantation experiments. Additionally, inulin exhibited antioxidant and anti-inflammatory properties, alleviating NLRP3 inflammasome activation and pyroptosis. In summary, inulin protected chickens from OTA-induced kidney damage, which might provide a potential strategy to mitigate the harmful effects of mycotoxins through prebiotics and safeguard renal health.
Collapse
Affiliation(s)
- Jingjing Wang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Mingzhen Jiang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Xuhai Li
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Yingrong Ye
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Yueqing Xie
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Ting Wu
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Yichun Chen
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Hongsen Yu
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Hanpeng Wu
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Zhengtao Yang
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| | - Ershun Zhou
- College of Life Science and Engineering, Foshan University, Foshan, Guangdong 528231, China
| |
Collapse
|
21
|
Rahayu ES, Yoga WK, Komalasari H, Mariyatun M, Yuda WA, Manurung NEP, Hasan PN, Suharman S, Pamungkaningtyas FH, Nurfiana DA, Pramesi PC, Gatya M, Therdtatha P, Nakayama J, Juffrie M, Djaafar TF, Marwati T, Utami T. Probiotic Chocolate Containing Lactobacillus plantarum Dad-13 Alters the Gut Microbiota Composition of Undernourished Children in Lombok: A Randomized Double-Blind Trial. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2024; 2024:9493797. [PMID: 39132547 PMCID: PMC11316911 DOI: 10.1155/2024/9493797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 08/13/2024]
Abstract
The present study investigated the ingestion effect of chocolate probiotic containing Lactobacillus plantarum Dad-13 in undernourished children. A 100-day observation was conducted on undernourished children in Lombok, who were divided into probiotic (n = 28) and placebo (n = 28) groups. Fecal sampling was performed on the 10th and 100th days and further analyzed for gut microbiota composition, short-chain fatty acid (SCFA), and fecal pH. A significant difference was found in the diversity index, fecal pH, and several microbiotas at the phylum and genus levels. At the phylum level, Bacteroidetes was significantly higher in the probiotic group, and a higher relative abundance (RA) of Firmicutes was found in the placebo group. At the genus level, significant differences were observed in some bacteria, such as Bifidobacterium and Prevotella. Therefore, it can be concluded that the probiotic intervention in this study resulted in changes of gut microbiota diversity and fecal pH. Trial Registration: Thai Clinical Trials Registry identifier: TCTR20220425001.
Collapse
Affiliation(s)
- Endang S. Rahayu
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Wahyu K. Yoga
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Husnita Komalasari
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Mariyatun Mariyatun
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Nancy E. P. Manurung
- Food Technology Study ProgramDepartment of Chemical EngineeringPoliteknik Negeri Sriwijaya, Palembang 30128, Indonesia
| | - Pratama N. Hasan
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Suharman Suharman
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Dina A. Nurfiana
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Putrika C. Pramesi
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Mifta Gatya
- Center for Food and Nutrition StudiesUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | | | - Jiro Nakayama
- Department of Bioscience and BiotechnologyFaculty of AgricultureKyushu University, Fukuoka 819-0395, Japan
| | - Mohammad Juffrie
- Department of Public HealthUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| | - Titiek F. Djaafar
- Research Center for Food Technology and ProcessNational Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Tri Marwati
- Research Center for Food Technology and ProcessNational Research and Innovation Agency, Yogyakarta 55861, Indonesia
| | - Tyas Utami
- Faculty of Agricultural TechnologyUniversitas Gadjah Mada, Yogyakarta 55281, Indonesia
| |
Collapse
|
22
|
Hu Y, Aljumaah MR, Azcarate-Peril MA. Galacto-Oligosaccharides and the Elderly Gut: Implications for Immune Restoration and Health. Adv Nutr 2024; 15:100263. [PMID: 38897384 PMCID: PMC11292246 DOI: 10.1016/j.advnut.2024.100263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/23/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024] Open
Abstract
The increasing prevalence of noncommunicable diseases in the aging population has been correlated with a decline in innate and adaptive immune responses; hence, it is imperative to identify approaches to improve immune function, prevent related disorders, and reduce or treat age-associated health complications. Prebiotic supplementation is a promising approach to modulate the gut microbiome and immune system, offering a potential strategy to maintain the integrity of immune function in older individuals. This review summarizes the current research on prebiotic galacto-oligosaccharide (GOS) immunomodulatory mechanisms mediated by bacterial-derived metabolites, including short-chain fatty acids and secondary bile acids, to maintain immune homeostasis. The potential applications of GOS as immunotherapy for age-related disease prevention in older individuals are also highlighted. This aligns with the global shift toward proactive healthcare and emphasizes the significance of early intervention in directing an individual's health trajectory.
Collapse
Affiliation(s)
- Yunan Hu
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States; UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Mashael R Aljumaah
- UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States; Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, United States; Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maria Andrea Azcarate-Peril
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, United States; UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
23
|
Wei X, Qian S, Yang Y, Mo J. Microbiome-based therapies for periodontitis and peri-implantitis. Oral Dis 2024; 30:2838-2857. [PMID: 37890080 DOI: 10.1111/odi.14782] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/16/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023]
Abstract
OBJECTIVES Periodontitis and peri-implantitis are oral infectious-inflammatory diseases associated with oral microbial dysbiosis. Microbiome-based therapies, characterized by manipulation of the microbiota, are emerging as promising therapeutic approaches to resolve the microbial dysbiosis and associated dysregulation of immune system. This review aims at summarizing recent progress on microbiome-based therapies in periodontitis and peri-implantitis, promoting a further understanding of the related therapeutic mechanisms. SUBJECTS AND METHODS Pertinent literatures focused on microbiome-based therapies for periodontitis and peri-implantitis are obtained from PubMed and Web of Science. RESULTS In this article, we review the roles and therapeutic mechanisms of four microbiome-based therapies, including probiotics, postbiotics, predatory bacteria and phages, and microbiota transplantation, in the management of periodontitis and peri-implantitis. Challenges facing this field are also discussed, highlighting the areas that require more attention and investigation. CONCLUSIONS Microbiome-based therapies may serve as effective treatment for periodontitis and peri-implantitis. This review presents a new viewpoint to this field.
Collapse
Affiliation(s)
- Xindi Wei
- Department of Oral and Maxillo-facial Implantology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Shujiao Qian
- Department of Oral and Maxillo-facial Implantology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yijie Yang
- Department of Oral and Maxillo-facial Implantology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Jiaji Mo
- Department of Oral and Maxillo-facial Implantology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
24
|
Safarpour D, Stover N, Shprecher DR, Hamedani AG, Pfeiffer RF, Parkman HP, Quigley EM, Cloud LJ. Consensus practice recommendations for management of gastrointestinal dysfunction in Parkinson disease. Parkinsonism Relat Disord 2024; 124:106982. [PMID: 38729797 DOI: 10.1016/j.parkreldis.2024.106982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Gastrointestinal (GI) dysfunction is a common non-motor feature of Parkinson disease (PD). GI symptoms may start years before the onset of motor symptoms and impair quality of life. Robust clinical trial data is lacking to guide screening, diagnosis and treatment of GI dysfunction in PD. OBJECTIVE To develop consensus statements on screening, diagnosis, and treatment of GI dysfunction in PD. METHODS The application of a modified Delphi panel allowed for the synthesis of expert opinions into clinical statements. Consensus was predefined as a level of agreement of 100 % for each item. Five virtual Delphi rounds were held. Two movement disorders neurologists reviewed the literature on GI dysfunction in PD and developed draft statements based on the literature review. Draft statements were distributed among the panel that included five movement disorder neurologists and two gastroenterologists, both experts in GI dysmotility and its impact on PD symptoms. All members reviewed the statements and references in advance of the virtual meetings. In the virtual meetings, each statement was discussed, edited, and a vote was conducted. If there was not 100 % consensus, further discussions and modifications ensued until there was consensus. RESULTS Statements were developed for screening, diagnosis, and treatment of common GI symptoms in PD and were organized by anatomic segments: oral cavity and esophagus, stomach, small intestine, and colon and anorectum. CONCLUSIONS These consensus recommendations offer a practical framework for the diagnosis and treatment of GI dysfunction in PD.
Collapse
Affiliation(s)
- Delaram Safarpour
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR, USA.
| | - Natividad Stover
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Ali G Hamedani
- Departments of Neurology, Ophthalmology, and Epidemiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ronald F Pfeiffer
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Henry P Parkman
- Section of Gastroenterology, Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | - Eamonn Mm Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - Leslie J Cloud
- Department of Neurology, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
25
|
Coqueiro JM, Costa LD, Silva LCE, Dos Santos Conceição L, da Silva Cardoso P, Ferreira Ribeiro CD, Otero DM. Trends in research on cacti: the food of the future. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:4939-4949. [PMID: 38314878 DOI: 10.1002/jsfa.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/26/2023] [Accepted: 01/10/2024] [Indexed: 02/07/2024]
Abstract
Cacti are a distinguished group of plants that stand out for their great nutritional values, diverse uses, and unique morphology, allowing them to grow and thrive under different conditions such as dry, xeric, and even low-temperature environments. The world is going through significant climate changes that are affecting the agriculture system. Therefore, sustainable and multifunctional crops, as many species of the Cactaceae family are, might be a good alternative in the near future. In this work, the uses of cacti in human food were analyzed through a scientific prospection from the point of view of their temporal and spatial distribution and potential uses. Brazil is the country with more publications related to the scope of this work, followed by Mexico. The presence of cacti in these countries can influence their interest in these species, which might reflect the results encountered in this study. The uses and ethnobotanical applications of cacti vary in different countries worldwide. Cactus is consumed fresh (in salads), in preparations (jams and sweets), and juices, being also present in traditional dishes in countries like Mexico. This study emphasizes cacti's importance in people's diets and ongoing world changes. Their ability to thrive even in hot environments with low water resources will lead to a greater focus on these species in the upcoming years. Furthermore, these plants have great flavor and contain several beneficial chemical compounds with desirable nutritional and health properties. Therefore, knowledge dissemination combined with technological innovations will allow greater use of these multifunctional species for human consumption. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
| | | | | | | | - Patrick da Silva Cardoso
- Graduate Program in Food, Nutrition, and Health, Nutrition School, Federal University of Bahia, Salvador, Brazil
| | - Camila Duarte Ferreira Ribeiro
- Graduate Program in Food, Nutrition, and Health, Nutrition School, Federal University of Bahia, Salvador, Brazil
- Graduate Program in Food Science, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| | - Deborah Murowaniecki Otero
- Graduate Program in Food, Nutrition, and Health, Nutrition School, Federal University of Bahia, Salvador, Brazil
- Graduate Program in Food Science, Faculty of Pharmacy, Federal University of Bahia, Salvador, Brazil
| |
Collapse
|
26
|
Zhang X, Yang Y, Ma X, Cao H, Sun Y. Probiotics relieve perioperative postoperative cognitive dysfunction induced by cardiopulmonary bypass through the kynurenine metabolic pathway. Sci Rep 2024; 14:12822. [PMID: 38834581 DOI: 10.1038/s41598-024-59275-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/09/2024] [Indexed: 06/06/2024] Open
Abstract
Postoperative cognitive dysfunction (POCD) has become the popular critical post-operative consequences, especially cardiopulmonary bypass surgery, leading to an increased risk of mortality. However, no therapeutic effect about POCD. Probiotics are beneficial bacteria living in the gut and help to reduce the risk of POCD. However, the detailed mechanism is still not entirely known. Therefore, our research aims to uncover the effect and mechanism of probiotics in relieving POCD and to figure out the possible relationship between kynurenine metabolic pathway. 36 rats were grouped into three groups: sham operated group (S group, n = 12), Cardiopulmonary bypass group (CPB group, n = 12), and probiotics+CPB (P group, n = 12). After CPB model preparation, water maze test and Garcia score scale was performed to identify the neurological function. Immunofluorescence and Hematoxylin and eosin staining has been used for hippocampal neurons detection. Brain injury related proteins, oxidative stress factors, and inflammatory factors were detected using enzyme-linked immunosorbent assays (ELISA). Neuronal apoptosis was detected by TdT-mediated dUTP nick end-labeling (TUNEL) staining and western blot. High-performance liquid chromatography/mass spectrometry (HPLC/MS) was performed to detect the key factors of the kynurenine metabolic pathway. Our results demonstrated that probiotics improved neurological function of post-CPB rats. The administration of probiotics ameliorated memory and learning in spatial terms CPB rats (P < 0.05). Hematoxylin and eosin (H&E) staining data, S-100β and neuron-specific enolase (NSE) data convinced that probiotics agonists reduced brain damage in CPB rats (P < 0.05). Moreover, probiotics regulated inflammatory factors, meanwhile attenuated hippocampal neuronal apoptosis. Probiotics alleviated POCD in rats with CPB through regulation of kynurenine metabolic signaling pathway.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, Hubei, China
- Postgraduate Training Base, The General Hospital of Northern Theater Command, Jinzhou Medical University, Jinzhou, 121013, Liaoning, China
| | - Yanzhang Yang
- Department of Anesthesiology, Chifeng Municipal Hospital, Chifeng, 024000, Inner Mongolia, China
| | - Xinyi Ma
- Postgraduate Training Base, The General Hospital of Northern Theater Command, Dalian Medical University, Dalian, 116051, Liaoning, China
| | - Huijuan Cao
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Yingjie Sun
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
27
|
Zhang Z, Hu Y, Zhang N, Li J, Lu J, Wei H. Dietary supplementation with non-digestible isomaltooligosaccharide and Lactiplantibacillus plantarum ZDY2013 ameliorates DSS-induced colitis via modulating intestinal barrier integrity and the gut microbiota. Food Funct 2024; 15:5908-5920. [PMID: 38738338 DOI: 10.1039/d4fo00421c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Non-digestible oligosaccharides have attracted attention due to their critical role in maintaining the balance of a host's gut microbiota. Lactiplantibacillus plantarum ZDY2013 was isolated from traditional fermented acid beans, which could metabolize many complex carbohydrates and had intestinal immunomodulatory effects. In our study, the ameliorative effect of a combination of non-digestible isomaltooligosaccharide (IMO) and L. plantarum ZDY2013 was investigated in dextran sulfate sodium (DSS)-induced colitis mice. The results showed that IMO could specifically promote L. plantarum ZDY2013 intestinal colonization after five days of gavage and ameliorate the symptoms of colitis (survival rate, DAI score, colon length, etc.) as well as colon tissue integrity. IMO combined with L. plantarum ZDY2013 increased the levels of intestinal tight junction proteins (ZO-1 and claudin) and mucin (MUC-2), followed by alleviation of inflammatory responses (decreased the expression of IL-1β, TNF-α, and IL-6 and increased the expression of IL-10 and IL-22) and the level of oxidative stress (decreased the level of COX-2 and iNOS and increased the expression of T-AOC and SOD). Furthermore, the combination increased the diversity of the gut microbiota and modulated the microbial structural component (decreased the abundance of Escherichia and Helicobacter and increased the abundance of Lactobacillus and SCFA-producing related species). Taken together, our results suggested that the consumption of IMO and L. plantarum ZDY2013 could improve the symptoms of colitis in mice by improving the intestinal barrier along with regulating the composition and metabolites of the gut microbiota.
Collapse
Affiliation(s)
- Zhihong Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
- Chongqing Research Institute, Nanchang University, Chongqing 402660, China
| | - Yingsheng Hu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Na Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jinmei Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jinlin Lu
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Hua Wei
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
- Sino-German Joint Research Institute, Nanchang University, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
28
|
Abstract
Increasing research has been conducted on the role of probiotics in disease treatment. Kefir, a safe, low-cost probiotic fermented milk drink, has been investigated in many in vitro and animal studies, although parameters for human therapeutic dose or treatment time have not yet been determined. Here we perform a scoping review of clinical studies that have used kefir as a therapeutic agent, compiling the results for perspectives to support and direct further research. This review was based on Joanna Briggs Institute guidelines, including studies on the effects of kefir-fermented milk in humans. Using the term KEFIR, the main international databases were searched for studies published in English, Spanish or Portuguese until 9 March 2022. A total of 5835 articles were identified in the four databases, with forty-four eligible for analysis. The research areas were classified as metabolic syndrome and type 2 diabetes, gastrointestinal health/disorders, maternal/child health and paediatrics, dentistry, oncology, women's and geriatric health, and dermatology. The many study limitations hampered generalisation of the results. The small sample sizes, methodological variation and differences in kefir types, dosage and treatment duration prevented clear conclusions about its benefits for specific diseases. We suggest using a standard therapeutic dose of traditionally prepared kefir in millilitres according to body weight, making routine consumption more feasible. The studies showed that kefir is safe for people without serious illnesses.
Collapse
Affiliation(s)
- Milena Klippel Bessa
- Postgraduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
| | | | - Renan Rangel Bonamigo
- Postgraduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, 90050-170, Porto Alegre, RS, Brazil
| |
Collapse
|
29
|
Nasreen S, Ali S, Andleeb S, Summer M, Hussain T, Imdad K, Ara C, Tahir HM. Mechanisms of medicinal, pharmaceutical, and immunomodulatory action of probiotics bacteria and their secondary metabolites against disease management: an overview. Folia Microbiol (Praha) 2024; 69:549-565. [PMID: 38532057 DOI: 10.1007/s12223-024-01155-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 03/05/2024] [Indexed: 03/28/2024]
Abstract
Probiotics or bacteriotherapy is today's hot issue for public entities (Food and Agriculture Organization, and World Health Organization) as well as health and food industries since Metchnikoff and his colleagues hypothesized the correlation between probiotic consumption and human's health. They contribute to the newest and highly efficient arena of promising biotherapeutics. These are usually attractive in biomedical applications such as gut-related diseases like irritable bowel disease, diarrhea, gastrointestinal disorders, fungal infections, various allergies, parasitic and bacterial infections, viral diseases, and intestinal inflammation, and are also worth immunomodulation. The useful impact of probiotics is not limited to gut-related diseases alone. Still, these have proven benefits in various acute and chronic infectious diseases, like cancer, human immunodeficiency virus (HIV) diseases, and high serum cholesterol. Recently, different researchers have paid special attention to investigating biomedical applications of probiotics, but consolidated data regarding bacteriotherapy with a detailed mechanistically applied approach is scarce and controversial. The present article reviews the bio-interface of probiotic strains, mainly (i) why the demand for probiotics?, (ii) the current status of probiotics, (iii) an alternative to antibiotics, (iv) the potential applications towards disease management, (v) probiotics and industrialization, and (vi) futuristic approach.
Collapse
Affiliation(s)
- Sundas Nasreen
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University, Lahore, 54000, Pakistan.
| | - Saiqa Andleeb
- Department of Zoology, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Muhammad Summer
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Tauqeer Hussain
- Department of Zoology, Government College University, Lahore, 54000, Pakistan
| | - Kaleem Imdad
- Department of Bioscience, COMSATS Institute of Information Technology (CIIT), Islamabad, 45550, Pakistan
| | - Chaman Ara
- Institute of Zoology, University of the Punjab, Lahore, Pakistan
| | | |
Collapse
|
30
|
Eveleens Maarse BC, Eggink HM, Warnke I, Bijlsma S, van den Broek TJ, Oosterman JE, Caspers MPM, Sybesma W, Gal P, van Kraaij SJW, Schuren FHJ, Moerland M, Hoevenaars FPM. Impact of fibre supplementation on microbiome and resilience in healthy participants: A randomized, placebo-controlled clinical trial. Nutr Metab Cardiovasc Dis 2024; 34:1416-1426. [PMID: 38499450 DOI: 10.1016/j.numecd.2024.01.028] [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: 09/06/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND AND AIMS The gut microbiome exerts important roles in health, e.g., functions in metabolism and immunology. These functions are often exerted via short-chain fatty acid (SCFA) production by gut bacteria. Studies demonstrating causal relationships between interventions targeting the microbiome and clinical outcomes are limited. This study aimed to show a causal relationship between microbiome modulation through fibre intervention and health. METHODS AND RESULTS This randomized, double-blind, cross-over study included 65 healthy subjects, aged 45-70 years, with increased metabolic risk (i.e., body mass index [BMI] 25-30 kg/m2, low to moderate daily dietary fibre intake, <30g/day). Subjects took daily a fibre mixture of Acacia gum and carrot powder or placebo for 12 weeks, with an 8-week wash-out period. Faecal samples for measurement of SCFAs and microbiome analysis were collected every 4 weeks. Before and after each intervention period subjects underwent the mixed-meal PhenFlex challenge Test (PFT). Health effects were expressed as resilience to the stressors of the PFT and as fasting metabolic and inflammatory state. The fibre mixture exerted microbiome modulation, with an increase in β-diversity (p < 0.001). α-diversity was lower during fibre mixture intake compared to placebo after 4, 8 and 12 weeks (p = 0.002; p = 0.012; p = 0.031). There was no effect observed on faecal SCFA concentrations, nor on any of the primary clinical outcomes (Inflammatory resilience: p = 0.605, Metabolic resilience: p = 0.485). CONCLUSION Although the intervention exerted effects on gut microbiome composition, no effects on SCFA production, on resilience or fasting metabolic and inflammatory state were observed in this cohort. REGISTRATION NUMBER CLINICALTRIALS.GOV: NCT04829396.
Collapse
Affiliation(s)
- Boukje C Eveleens Maarse
- Centre for Human Drug Research, Leiden, the Netherlands; Leiden University Medical Center, Leiden, the Netherlands
| | - Hannah M Eggink
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Ines Warnke
- dsm-firmenich, CH-4303, Kaiseraugst, Switzerland
| | - Sabina Bijlsma
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Tim J van den Broek
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Johanneke E Oosterman
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Martien P M Caspers
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | | | - Pim Gal
- Centre for Human Drug Research, Leiden, the Netherlands; Leiden University Medical Center, Leiden, the Netherlands
| | - Sebastiaan J W van Kraaij
- Centre for Human Drug Research, Leiden, the Netherlands; Leiden University Medical Center, Leiden, the Netherlands
| | - Frank H J Schuren
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands
| | - Matthijs Moerland
- Centre for Human Drug Research, Leiden, the Netherlands; Leiden University Medical Center, Leiden, the Netherlands
| | - Femke P M Hoevenaars
- TNO, Netherlands Organisation for Applied Scientific Research, Leiden, the Netherlands.
| |
Collapse
|
31
|
Yang K, Chen Y, Wang M, Zhang Y, Yuan Y, Hou H, Mao YH. The Improvement and Related Mechanism of Microecologics on the Sports Performance and Post-Exercise Recovery of Athletes: A Narrative Review. Nutrients 2024; 16:1602. [PMID: 38892536 PMCID: PMC11174581 DOI: 10.3390/nu16111602] [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: 04/24/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The diversity and functionality of gut microbiota may play a crucial role in the function of human motor-related systems. In addition to traditional nutritional supplements, there is growing interest in microecologics due to their potential to enhance sports performance and facilitate post-exercise recovery by modulating the gut microecological environment. However, there is a lack of relevant reviews on this topic. This review provides a comprehensive overview of studies investigating the effects of various types of microecologics, such as probiotics, prebiotics, synbiotics, and postbiotics, on enhancing sports performance and facilitating post-exercise recovery by regulating energy metabolism, mitigating oxidative-stress-induced damage, modulating immune responses, and attenuating bone loss. Although further investigations are warranted to elucidate the underlying mechanisms through which microecologics exert their effects. In summary, this study aims to provide scientific evidence for the future development of microecologics in athletics.
Collapse
Affiliation(s)
- Keer Yang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Yonglin Chen
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Minghan Wang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Yishuo Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Haoyang Hou
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
| | - Yu-Heng Mao
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China; (K.Y.); (Y.C.); (M.W.); (Y.Z.); (Y.Y.); (H.H.)
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| |
Collapse
|
32
|
Chen X, He L, Zhang C, Zheng G, Lin S, Zou Y, Lu Y, Feng Y, Zheng D. Exploring new avenues of health protection: plant-derived nanovesicles reshape microbial communities. J Nanobiotechnology 2024; 22:269. [PMID: 38764018 PMCID: PMC11103870 DOI: 10.1186/s12951-024-02500-w] [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: 03/10/2024] [Accepted: 04/27/2024] [Indexed: 05/21/2024] Open
Abstract
Symbiotic microbial communities are crucial for human health, and dysbiosis is associated with various diseases. Plant-derived nanovesicles (PDNVs) have a lipid bilayer structure and contain lipids, metabolites, proteins, and RNA. They offer unique advantages in regulating microbial community homeostasis and treating diseases related to dysbiosis compared to traditional drugs. On the one hand, lipids on PDNVs serve as the primary substances that mediate specific recognition and uptake by bacteria. On the other hand, due to the multifactorial nature of PDNVs, they have the potential to enhance growth and survival of beneficial bacterial while simultaneously reducing the pathogenicity of harmful bacteria. In addition, PDNVs have the capacity to modulate bacterial metabolism, thus facilitating the establishment of a harmonious microbial equilibrium and promoting stability within the microbiota. These remarkable attributes make PDNVs a promising therapeutic approach for various conditions, including periodontitis, inflammatory bowel disease, and skin infection diseases. However, challenges such as consistency, isolation methods, and storage need to be addressed before clinical application. This review aims to explore the value of PDNVs in regulating microbial community homeostasis and provide recommendations for their use as novel therapeutic agents for health protection.
Collapse
Affiliation(s)
- Xiaohang Chen
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Lianghang He
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Chaochao Zhang
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Genggeng Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuoqi Lin
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuchun Zou
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yan Feng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
33
|
Velderrain-Armenta F, González-Ochoa G, Tamez-Guerra P, Romero-Arguelles R, Romo-Sáenz CI, Gomez-Flores R, Flores-Mendoza L, Icedo-García R, Soñanez-Organis JG. Bifidobacterium longum and Chlorella sorokiniana Combination Modulates IFN-γ, IL-10, and SOCS3 in Rotavirus-Infected Cells. Int J Mol Sci 2024; 25:5514. [PMID: 38791551 PMCID: PMC11122607 DOI: 10.3390/ijms25105514] [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: 04/01/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
Rotavirus is the main cause of acute diarrhea in children up to five years of age. In this regard, probiotics are commonly used to treat or prevent gastroenteritis including viral infections. The anti-rotavirus effect of Bifidobacterium longum and Chlorella sorokiniana, by reducing viral infectivity and improving IFN-type I response, has been previously reported. The present study aimed to study the effect of B. longum and/or C. sorokiniana on modulating the antiviral cellular immune response mediated by IFN-γ, IL-10, SOCS3, STAT1, and STAT2 genes in rotavirus-infected cells. To determine the mRNA relative expression of these genes, HT-29 cells were treated with B. longum and C. sorokiniana alone or in combination, followed by rotavirus infection. In addition, infected cells were treated with B. longum and/or C. sorokiniana. Cellular RNA was purified, used for cDNA synthesis, and amplified by qPCR. Our results demonstrated that the combination of B. longum and C. sorokiniana stimulates the antiviral cellular immune response by upregulating IFN-γ and may block pro-inflammatory cytokines by upregulating IL-10 and SOCS3. The results of our study indicated that B. longum, C. sorokiniana, or their combination improve antiviral cellular immune response and might modulate pro-inflammatory responses.
Collapse
Affiliation(s)
- Felizardo Velderrain-Armenta
- Department of Chemistry-Biology and Agriculture, Interdisciplinary Faculty of Biology Sciences and Health, University of Sonora, Navojoa C.P. 85880, Mexico; (F.V.-A.); (L.F.-M.); (R.I.-G.); (J.G.S.-O.)
| | - Guadalupe González-Ochoa
- Department of Chemistry-Biology and Agriculture, Interdisciplinary Faculty of Biology Sciences and Health, University of Sonora, Navojoa C.P. 85880, Mexico; (F.V.-A.); (L.F.-M.); (R.I.-G.); (J.G.S.-O.)
| | - Patricia Tamez-Guerra
- Laboratory of Immunology and Virology, Falculty of Biological Sciences, Autonomous University of Nuevo Leon, San Nicolás de los Garza C.P. 66455, Mexico; (R.R.-A.); (C.I.R.-S.); (R.G.-F.)
| | - Ricardo Romero-Arguelles
- Laboratory of Immunology and Virology, Falculty of Biological Sciences, Autonomous University of Nuevo Leon, San Nicolás de los Garza C.P. 66455, Mexico; (R.R.-A.); (C.I.R.-S.); (R.G.-F.)
| | - César I. Romo-Sáenz
- Laboratory of Immunology and Virology, Falculty of Biological Sciences, Autonomous University of Nuevo Leon, San Nicolás de los Garza C.P. 66455, Mexico; (R.R.-A.); (C.I.R.-S.); (R.G.-F.)
| | - Ricardo Gomez-Flores
- Laboratory of Immunology and Virology, Falculty of Biological Sciences, Autonomous University of Nuevo Leon, San Nicolás de los Garza C.P. 66455, Mexico; (R.R.-A.); (C.I.R.-S.); (R.G.-F.)
| | - Lilian Flores-Mendoza
- Department of Chemistry-Biology and Agriculture, Interdisciplinary Faculty of Biology Sciences and Health, University of Sonora, Navojoa C.P. 85880, Mexico; (F.V.-A.); (L.F.-M.); (R.I.-G.); (J.G.S.-O.)
| | - Ramona Icedo-García
- Department of Chemistry-Biology and Agriculture, Interdisciplinary Faculty of Biology Sciences and Health, University of Sonora, Navojoa C.P. 85880, Mexico; (F.V.-A.); (L.F.-M.); (R.I.-G.); (J.G.S.-O.)
| | - José G. Soñanez-Organis
- Department of Chemistry-Biology and Agriculture, Interdisciplinary Faculty of Biology Sciences and Health, University of Sonora, Navojoa C.P. 85880, Mexico; (F.V.-A.); (L.F.-M.); (R.I.-G.); (J.G.S.-O.)
| |
Collapse
|
34
|
Wójcik R, Małaczewska J, Tobolski D, Miciński J, Kaczorek-Łukowska E, Zwierzchowski G. The Effect of Orally Administered Multi-Strain Probiotic Formulation ( Lactobacillus, Bifidobacterium) on the Phagocytic Activity and Oxidative Metabolism of Peripheral Blood Granulocytes and Monocytes in Lambs. Int J Mol Sci 2024; 25:5068. [PMID: 38791112 PMCID: PMC11120738 DOI: 10.3390/ijms25105068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Probiotic feed additives have attracted considerable research interest in recent years because the effectiveness of probiotics can differ across microbial strains and the supplemented macroorganisms. The present study was conducted on 16 lambs divided equally into two groups (C-control and E-experimental). The examined lambs were aged 11 days at the beginning of the experiment and 40 days at the end of the experiment. The diet of group E lambs was supplemented with a multi-strain probiotic formulation (Lactobacillus plantarum AMT14, Lactobacillus plantarum AMT4, Lactobacillus rhamnosus AMT15, and Bifidobacterium animalis AMT30), whereas group C lambs did not receive the probiotic additive. At the beginning of the experiment (day 0) and on experimental days 15 and 30, blood was sampled from the jugular vein to determine and compare: phagocytic activity (Phagotest) and oxidative metabolism (Phagoburst) of peripheral blood granulocytes and monocytes by flow cytometry. An analysis of the phagocytic activity of granulocytes and monocytes revealed significantly higher levels of phagocytic activity (expressed as the percentage of phagocytic cells and mean fluorescence intensity) in lambs that were administered the multi-strain probiotic formulation compared with lambs in the control group. The probiotic feed additive also exerted a positive effect on the oxidative metabolism of both granulocytes and monocytes (expressed as the percentage of oxidative metabolism and mean fluorescence intensity) after stimulation with Escherichia coli bacteria and with PMA (4-phorbol-12-β-myristate-13-acetate). These findings suggest that the tested probiotic formulation may have a positive effect on the immune status of lambs.
Collapse
Affiliation(s)
- Roman Wójcik
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-718 Olsztyn, Poland; (R.W.); (J.M.); (E.K.-Ł.)
| | - Joanna Małaczewska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-718 Olsztyn, Poland; (R.W.); (J.M.); (E.K.-Ł.)
| | - Dawid Tobolski
- Department of Internal Diseases with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 14, 10-957 Olsztyn, Poland;
| | - Jan Miciński
- Department of Sheep and Goat Breeding, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-917 Olsztyn, Poland;
| | - Edyta Kaczorek-Łukowska
- Department of Microbiology and Clinical Immunology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-718 Olsztyn, Poland; (R.W.); (J.M.); (E.K.-Ł.)
| | - Grzegorz Zwierzchowski
- Department of Biochemistry, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Oczapowskiego 1A, 10-719 Olsztyn, Poland
| |
Collapse
|
35
|
Zhu X, Zhao L, Lei L, Zhu Y, Xu J, Liu L. Fecal microbiota transplantation ameliorates abdominal obesity through inhibiting microbiota-mediated intestinal barrier damage and inflammation in mice. Microbiol Res 2024; 282:127654. [PMID: 38417203 DOI: 10.1016/j.micres.2024.127654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/01/2024]
Abstract
Abdominal obesity (AO), characterized by the excessive abdominal fat accumulation, has emerged as a significant public health concern due to its metabolic complications and escalating prevalence worldwide, posing a more pronounced threat to human health than general obesity. While certain studies have indicated that intestinal flora contributed to diet-induced general obesity, the precise involvement of gut microbiota in the development of AO, specifically the accumulation of abdominal fat, remains inadequately explored. In this study, the 16 S rDNA sequencing was employed to analyze gut flora alterations, and the intestinal microbiota dysbiosis characterized by a vanishing decline of Akkermansia was found in the AO group. Along with notable gut microbiota changes, the intestinal mucosal barrier damage and metabolic inflammation were detected, which collectively promoted metabolic dysregulation in AO. Furthermore, the metabolic inflammation and AO were ameliorated after the intestinal microbiota depletion with antibiotics (ABX) drinking, underscoring a significant involvement of gut microbiota dysbiosis in the progression of AO. More importantly, our findings demonstrated that the transplantation of healthy intestinal flora successfully reversed the gut microbiota dysbiosis, particularly the decline of Akkermansia in the AO group. The gut flora reshaping has led to the repair of gut barrier damage and mitigation of metabolic inflammation, which ultimately ameliorated abdominal fat deposition. Our study established the role of interactions between gut flora, mucus barrier, and metabolic inflammation in the development of AO, thereby offering a theoretical foundation for the clinical application of fecal microbiota transplantation (FMT) as a treatment for AO.
Collapse
Affiliation(s)
- Xiaoqiang Zhu
- Central Laboratory, Wuhan Fourth Hospital, Wuhan, China; Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China; National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| | - Lijun Zhao
- Hubei Jiangxia Laboratory, Wuhan, China; National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Lei
- Central Laboratory, Wuhan Fourth Hospital, Wuhan, China; Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China
| | - Yanhong Zhu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Xu
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China
| | - Li Liu
- Department of Pharmacy, Wuhan Fourth Hospital, Wuhan, China.
| |
Collapse
|
36
|
Choi SI, Kim N, Choi Y, Nam RH, Jang JY, Cho SY. The Effect of Clostridium butyricum on Gut Microbial Changes and Functional Profiles of Metabolism in High-fat Diet-fed Rats Depending on Age and Sex. J Neurogastroenterol Motil 2024; 30:236-250. [PMID: 38576373 PMCID: PMC10999835 DOI: 10.5056/jnm23096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/24/2023] [Accepted: 11/16/2023] [Indexed: 04/06/2024] Open
Abstract
Background/Aims A high-fat diet (HFD) causes dysbiosis and promotes inflammatory responses in the colon. This study aims to evaluate the effects of Clostridium butyricum on HFD-induced gut microbial changes in rats. Methods Six-week-old Fischer-344 rats with both sexes were given a control or HFD during 8 weeks, and 1-to-100-fold diluted Clostridium butyricum were administered by gavage. Fecal microbiota analyses were conducted using 16S ribosomal RNA metagenomic sequencing and predictive functional profiling of microbial communities in metabolism. Results A significant increase in Ruminococcaceae and Lachnospiraceae, which are butyric acid-producing bacterial families, was observed in the probiotics groups depending on sex. In contrast, Akkermansia muciniphila, which increased through a HFD regardless of sex, and decreased in the probiotics groups. A. muciniphila positively correlated with Claudin-1 expression in males (P < 0.001) and negatively correlated with the expression of Claudin-2 (P = 0.042), IL-1β (P = 0.037), and IL-6 (P = 0.044) in females. In terms of functional analyses, a HFD decreased the relative abundances of M00131 (carbohydrate metabolism module), M00579, and M00608 (energy metabolism), and increased those of M00307 (carbohydrate metabolism), regardless of sex. However, these changes recovered especially in male C. butyricum groups. Furthermore, M00131, M00579, and M00608 showed a positive correlation and M00307 showed a negative correlation with the relative abundance of A. muciniphila (P < 0.001). Conclusion The beneficial effects of C. butyricum on HFD-induced gut dysbiosis in young male rats originate from the functional profiles of carbohydrate and energy metabolism.
Collapse
Affiliation(s)
- Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Jae Young Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Yup Cho
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Choi SI, Kim N, Nam RH, Jang JY, Kim EH, Ha S, Kang K, Lee W, Shin CM, Lee DH. Sex Difference in the Effect of Bifidobacterium longum on Repeated Water Avoidance Stress-induced Gut Dysbiosis in Wistar Rats. J Cancer Prev 2024; 29:16-23. [PMID: 38567109 PMCID: PMC10982521 DOI: 10.15430/jcp.23.042] [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] [Received: 12/23/2023] [Revised: 12/31/2023] [Accepted: 01/02/2024] [Indexed: 04/04/2024] Open
Abstract
Dysbiosis in gut microbiota is known to contribute to development of irritable bowel syndrome. We tried to investigate the effect of Bifidobacterium longum on repeated water avoidance stress (WAS) in a Wistar rat model. The three groups (no-stress, WAS, and WAS with B. longum) of rats were allocated to sham or WAS for 1 hour daily for 10 days, and B. longum was administered through gavage for 10 days. Fecal pellet numbers were counted at the end of each 1-hour session of WAS. After 10 days of repeated WAS, the rats were eutanized, and the feces were collected. WAS increased fecal pellet output (FPO) significantly in both sexes (P < 0.001), while the female B. longum group showed significantly decreased FPO (P = 0.005). However, there was no consistent change of myeloperoxidase activity and mRNA expression of interleukin-1β and TNF-α. Mast cell infiltration at colonic submucosa increased in the female WAS group (P = 0.016). In terms of fecal microbiota, the repeated WAS groups in both sexes showed different beta-diversity compared to control and WAS with B. longum groups. WAS-induced mast cell infiltration was reduced by the administration of B. longum in female rats. Moreover, administration of B. longum relieved WAS-caused dysbiosis, especially in female rats. In conclusion, B. longum was beneficial for WAS-induced stress in rats, especially in females.
Collapse
Affiliation(s)
- Soo In Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jae Young Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Hye Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - SungChan Ha
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | | | | | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
38
|
Bekar C, Ozmen O, Ozkul C, Ayaz A. Inulin protects against the harmful effects of dietary emulsifiers on mice gut microbiome. PeerJ 2024; 12:e17110. [PMID: 38525281 PMCID: PMC10961058 DOI: 10.7717/peerj.17110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/23/2024] [Indexed: 03/26/2024] Open
Abstract
Background The prevalence of inflammatory bowel diseases is increasing, especially in developing countries, with adoption of Western-style diet. This study aimed to investigate the effects of two emulsifiers including lecithin and carboxymethyl cellulose (CMC) on the gut microbiota, intestinal inflammation and the potential of inulin as a means to protect against the harmful effects of emulsifiers. Methods In this study, male C57Bl/6 mice were divided into five groups (n:6/group) (control, CMC, lecithin, CMC+inulin, and lecithin+inulin). Lecithin and CMC were diluted in drinking water (1% w/v) and inulin was administered daily at 5 g/kg for 12 weeks. Histological examination of the ileum and colon, serum IL-10, IL-6, and fecal lipocalin-2 levels were analyzed. 16S rRNA gene V3-V4 region amplicon sequencing was performed on stool samples. Results In the CMC and lecithin groups, shortening of the villus and a decrease in goblet cells were observed in the ileum and colon, whereas inulin reversed this effect. The lipocalin level, which was 9.7 ± 3.29 ng in the CMC group, decreased to 4.1 ± 2.98 ng with the administration of inulin. Bifidobacteria and Akkermansia were lower in the CMC group than the control, while they were higher in the CMC+inulin group. In conclusion, emulsifiers affect intestinal health negatively by disrupting the epithelial integrity and altering the composition of the microbiota. Inulin is protective on their harmful effects. In addition, it was found that CMC was more detrimental to microbiota composition than lecithin.
Collapse
Affiliation(s)
- Cansu Bekar
- Department of Nutrition and Dietetics, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ceren Ozkul
- Department of Pharmaceutical Microbiology, Hacettepe University, Ankara, Turkey
| | - Aylin Ayaz
- Department of Nutrition and Dietetics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
39
|
Sun Y, Wang X, Li L, Zhong C, Zhang Y, Yang X, Li M, Yang C. The role of gut microbiota in intestinal disease: from an oxidative stress perspective. Front Microbiol 2024; 15:1328324. [PMID: 38419631 PMCID: PMC10899708 DOI: 10.3389/fmicb.2024.1328324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Recent studies have indicated that gut microbiota-mediated oxidative stress is significantly associated with intestinal diseases such as colorectal cancer, ulcerative colitis, and Crohn's disease. The level of reactive oxygen species (ROS) has been reported to increase when the gut microbiota is dysregulated, especially when several gut bacterial metabolites are present. Although healthy gut microbiota plays a vital role in defending against excessive oxidative stress, intestinal disease is significantly influenced by excessive ROS, and this process is controlled by gut microbiota-mediated immunological responses, DNA damage, and intestinal inflammation. In this review, we discuss the relationship between gut microbiota and intestinal disease from an oxidative stress perspective. In addition, we also provide a summary of the most recent therapeutic approaches for preventing or treating intestinal diseases by modifying gut microbiota.
Collapse
Affiliation(s)
- Yiqi Sun
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xurui Wang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Li
- Department of Anorectal Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Zhong
- Traditional Chinese Medicine Department of Orthopaedic and Traumatic, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Zhang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Xiangdong Yang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Mingyue Li
- Special Needs Outpatient Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chao Yang
- Surgery of Traditional Chinese Medicine Department, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
40
|
Gkitsaki I, Potsaki P, Dimou I, Laskari Z, Koutelidakis A, Giaouris E. Development of a functional Greek sheep yogurt incorporating a probiotic Lacticaseibacillus rhamnosus wild-type strain as adjunct starter culture. Heliyon 2024; 10:e24446. [PMID: 38312657 PMCID: PMC10835171 DOI: 10.1016/j.heliyon.2024.e24446] [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] [Received: 09/28/2023] [Revised: 12/23/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024] Open
Abstract
Greek yogurt is a fermented dairy product of high nutritional value that can be used as a matrix for the delivery of probiotics. The aim of this study was to develop a new probiotic Greek sheep yogurt with upgraded quality and functional characteristics. To do this, yogurt was manufactured by fermenting pasteurized milk with the commercial starter culture (Streptococcus thermophilus (ST), Lactobacillus bulgaricus (LB)) together with a probiotic Lacticaseibacillus rhamnosus (LR) wild-type strain (probiotic yogurt; PY). As a control, yogurt manufactured with only the starter culture (ST, LB) was used (conventional yogurt; CY) The survival of all three lactic acid bacteria (LAB) species (ST, LB, and LR) was monitored throughout the products' shelf life (storage at 4 °C for 25 days), and also following exposure to a static in vitro digestion model (SIVDM). The population dynamics of total aerobic plate count (APC), Enterobacteriaceae, yeasts and molds grown in both yogurts were also determined. The total antioxidant activity (AA) of yogurts was comparatively determined using in parallel two different assays, whereas the Folin-Ciocalteu assay was used to determine their total phenolic content (TPC). At each sampling day, yogurts were also evaluated for their pH, titratable acidity (TA) and main sensory characteristics. The population of probiotic LR remained stable during the shelf life (and above 108 CFU/g). Yogurt starters (ST, LB) were not detected following SIVDM, whereas LR (in PY) presented a reduction of about only one log. The AA and TPC of PY were found significantly higher than that of CY (P < 0.05). At the end of storage (25th day), neither pH nor TA differed significantly between the two yogurt types, while no fungal growth was observed in the PY. Consumer sensory analysis did not reveal important differences between the two yogurt types during their shelf life. To sum up, the novel yogurt was able to deliver to consumers a high number of probiotic cells (>108 CFU/g), presented increased antioxidant power, had an expanded shelf life, and maintained its good sensory attributes.
Collapse
Affiliation(s)
- Ioanna Gkitsaki
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Panagiota Potsaki
- Laboratory of Nutrition and Public Health, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Ioanna Dimou
- Laboratory of Nutrition and Public Health, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Zoi Laskari
- Mystakelli Traditional Dairy Products, 81103 Mantamados, Lesvos, Greece
| | - Antonios Koutelidakis
- Laboratory of Nutrition and Public Health, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| | - Efstathios Giaouris
- Laboratory of Food Microbiology and Hygiene, Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Myrina, Lemnos, Greece
| |
Collapse
|
41
|
Siu PLK, Choy CT, Chan HHY, Leung RKK, Chan UK, Zhou J, Wong CH, Lee YW, Chan HW, Lo CJY, Tsui JCC, Loo SKF, Tsui SKW. A Novel Multi-Strain E3 Probiotic Formula Improved the Gastrointestinal Symptoms and Quality of Life in Chinese Psoriasis Patients. Microorganisms 2024; 12:208. [PMID: 38276193 PMCID: PMC10820679 DOI: 10.3390/microorganisms12010208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/12/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Psoriasis is a chronic immune-mediated inflammatory disease affecting the skin and other systems. Gastrointestinal disease was found to be correlated with psoriasis in previous studies and it can significantly affect the quality of life of psoriasis patients. Despite the importance of the gut microbiome in gut and skin health having already been demonstrated in many research studies, the potential effect of probiotics on GI comorbidities in psoriasis patients is unclear. To investigate the effects of probiotics on functional GI comorbidities including irritable bowel syndrome, functional constipation, and functional diarrhea in psoriasis patients, we conducted a targeted 16S rRNA sequencing and comprehensive bioinformatic analysis among southern Chinese patients to compare the gut microbiome profiles of 45 psoriasis patients over an 8-week course of novel oral probiotics. All the participants were stratified into responders and non-responders according to their improvement in GI comorbidities, which were based on their Bristol Stool Form Scale (BSFS) scores after intervention. The Dermatological Life Quality Index (DLQI) score revealed a significant improvement in quality of life within the responder group (DLQI: mean 10.4 at week 0 vs. mean 15.9 at week 8, p = 0.0366). The proportion of psoriasis patients without GI comorbidity manifestation at week 8 was significantly higher than that at week 0 (week 0: Normal 53.33%, Constipation/Diarrhea 46.67%; week 8: Normal 75.56%, Constipation/Diarrhea 24.44%, p = 0.0467). In addition, a significant difference in the gut microbiome composition between the responders and non-responders was observed according to alpha and beta diversities. Differential abundance analysis revealed that the psoriasis patients exhibited (1) an elevated relative abundance of Lactobacillus acidophilus, Parabacteroides distasonis, and Ruminococcus bromii and (2) a reduced relative abundance of Oscillibacter, Bacteroides vulgatus, Escherichia sp., and Biophila wadsworthia after the 8-week intervention. The responders also exhibited a higher relative abundance of Fusicatenibacter saccharivorans when compared to the non-responders. In summary, our study discovers the potential clinical improvement effects of the novel probiotic formula in improving GI comorbidities and quality of life in psoriasis patients. We also revealed the different gut microbiome composition as well as the gut microbial signatures in the patients who responded to probiotics. These findings could provide insight into the use of probiotics in the management of psoriasis symptoms.
Collapse
Affiliation(s)
- Pui Ling Kella Siu
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Chi Tung Choy
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Helen Hoi Yin Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Ross Ka Kit Leung
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Un Kei Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Junwei Zhou
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Chi Ho Wong
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Yuk Wai Lee
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Ho Wang Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Claudia Jun Yi Lo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Joseph Chi Ching Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
| | - Steven King Fan Loo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Dermatology Centre, CUHK Medical Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Stephen Kwok Wing Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong, China; (P.L.K.S.)
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
42
|
Maleki S, Razavi SH, Yadav H, Letizia Manca M. New horizon to the world of gut microbiome: seeds germination. Crit Rev Food Sci Nutr 2024:1-19. [PMID: 38227048 DOI: 10.1080/10408398.2023.2300703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The second brain of humans has been known as the microbiome. The microbiome is a dynamic network composed of commensal bacteria, archaea, viruses, and fungi colonized in the human gastrointestinal tract. They play a vital role in human health by metabolizing components, maturation of the immune system, and taking part in the treatment of various diseases. Two important factors that can affect the gut microbiome's composition and/or function are the food matrix and methods of food processing. Based on scientific research, the consumption of whole grains can make positive changes in the gut microbiota. Seeds contain different microbiota-accessible substrates that can resist digestion in the upper gastrointestinal tract. Seed germination is one of the simplest and newest food processing approaches to improve seeds' bioavailability and overall nutritional value. During germination, the dormant hydrolytic seed's enzymes have been activated and then metabolize the macromolecules. The quality and quantity of bioactive compounds like prebiotics, fiber, phenolic compounds (PC), total free amino acids, and γ-aminobutyric acid (GABA) can increase even up to 4-10 folds in some cases. These components stimulate the survival and growth of healthful bacteria like probiotics and boost their activity. This effect depends on several parameters, e.g., germination environmental conditions. This review aims to provide up-to-date and latest research about promoting bioactive components during seed germination and investigating their impacts on gut microbiota to understand the possible direct and indirect effects of seed germination on the microbiome and human health.
Collapse
Affiliation(s)
- Sima Maleki
- Bioprocess Engineering Laboratory (BPEL), Department of Food Science, Engineering and Technology, Faculty of Agriculture Engineering, University of Tehran, Karaj, Iran
| | - Seyed Hadi Razavi
- Bioprocess Engineering Laboratory (BPEL), Department of Food Science, Engineering and Technology, Faculty of Agriculture Engineering, University of Tehran, Karaj, Iran
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, and Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Maria Letizia Manca
- Department of Scienze della Vita e dell'Ambiente, University of Cagliari, Cagliari, Italy
| |
Collapse
|
43
|
Momo Kadia B, Otiti MI, Ramsteijn AS, Sow D, Faye B, Heffernan C, Hall LJ, Webster JP, Walker AW, Allen S. Modulating the early-life gut microbiota using pro-, pre-, and synbiotics to improve gut health, child development, and growth. Nutr Rev 2024; 82:244-247. [PMID: 37167530 PMCID: PMC10777666 DOI: 10.1093/nutrit/nuad050] [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] [Indexed: 05/13/2023] Open
Abstract
In children exposed to poor hygiene and sanitation, invasion of the gut by pathogenic microbes can result in a subclinical enteropathy termed "environmental enteric dysfunction" (EED) that contributes to undernutrition, growth faltering, and impaired organ development. EED may already be present by age 6-12 weeks; therefore, interventions that can be started early in life, and used alongside breastfeeding, are needed to prevent or ameliorate EED. A healthy gut microbiota is critical for intestinal development and repair, nutrient digestion and absorption, and resisting colonization or overgrowth by pathogens. However, its development can be impaired by several environmental factors. Dietary supplementation with pro-, pre-, or synbiotics may be a pragmatic and safe means of building the resilience of the developing gut microbiota against adverse environmental factors, thereby preventing EED.
Collapse
Affiliation(s)
- Benjamin Momo Kadia
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Mary Iwaret Otiti
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | | | - Doudou Sow
- Service de Parasitologie-Mycologie, UFR Sciences de la Santé, Université Gaston Berger, Saint Louis, Senegal
| | - Babacar Faye
- Service de Parasitologie-Mycologie, Faculté de Médecine, Université Cheikh Anta Diop, Dakar, Senegal
| | | | - Lindsay J Hall
- Intestinal Health, School of Life Sciences, ZIEL—Institute for Food & Health, Technical University of Munich, Freising, Germany
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Joanne P Webster
- Centre for Emerging, Endemic and Exotic Diseases (CEEED), Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - Alan W Walker
- Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| | - Stephen Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
44
|
Li F, Yu C, Zhao Q, Wang Z, Wang Z, Chang Y, Xu Z, Han X, Li H, Liu Y, Hu S, Chang S, Tang T, Li Y. Exploring the intestinal ecosystem: from gut microbiota to associations with subtypes of inflammatory bowel disease. Front Cell Infect Microbiol 2024; 13:1304858. [PMID: 38239508 PMCID: PMC10794348 DOI: 10.3389/fcimb.2023.1304858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/06/2023] [Indexed: 01/22/2024] Open
Abstract
Objective Significant differences have been discovered between subtypes of Crohn's disease (CD) and ulcerative colitis (UC). The role of gut microbiota in promoting the onset of UC and CD is established, but conclusions regarding subtype-specific analyses remain limited. Methods This study aims to explore the influence of gut microbiota on subtypes of UC and CD, offering novel insights into the pathogenesis and treatment of UC and CD.Two-sample Mendelian randomization (MR) analysis was employed to examine the causal relationship between subtypes of UC and CD and gut microbiota composition. Gut microbiota data were sourced from the International Consortium MiBioGen, while UC and CD data were obtained from FINNGEN. Eligible single nucleotide polymorphisms (SNPs) were selected as instrumental variables. Multiple analytical approaches such as inverse variance-weighted (IVW), MR-Egger regression, weighted median, weighted mode, and MR-RAPS were utilized. Sensitivity analyses including MR-Egger intercept test, Cochran's Q test, and leave-one-out analysis were conducted for quality control. Subsequently, we employed multivariable IVW, MR-Egger, weighted median, and LASSO regression methods to identify independently significant genera or families and conducted sensitivity analyses. Results We have determined that Hungatella, Acidaminococcaceae, and 15 other microbial taxa act as protective factors for various CD and UC subtypes, while Terrisporobacter, Anaerostipes, and 23 other microbial taxa are associated with increased risk for different CD and UC subtypes. Furthermore, through multivariable MR analysis, we have identified significant genera or families with independent effects. Conclusion Our study confirms a causal relationship between dysbiosis of gut microbiota and the occurrence of CD and UC subtypes. Furthermore, it validates etiological distinctions among different subtypes of CD and UC. A novel approach to adjunctive therapy involving distinct UC or CD subtypes may involve the use of probiotics and represents a potential avenue for future treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Tongyu Tang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| | - Yuqin Li
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
45
|
Liu Y, Xing Y, Kong X. Exploratory analysis on the relationship between dietary live microbe intake and arthritis: a national population based cross-sectional study. Front Nutr 2024; 10:1328238. [PMID: 38239838 PMCID: PMC10794527 DOI: 10.3389/fnut.2023.1328238] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Objective The pathogenesis of arthritis such as rheumatoid arthritis (RA) and osteoarthritis (OA) remains unclear. This study aims to investigate whether the intake of live dietary microbes can be used as an auxiliary means for the treatment of arthritis. Methods Data used in the present research were originated from the US National Health and Nutrition Examination Survey (NHANES) from 2003-2018. Participants involved in the present study were categorized into three groups based on the dietary live microbe classification system, namely low, medium, and high dietary live microbe groups. The analyses utilized weighted univariate and multivariate logistic regression. The restricted cubic spline plot was used to explore the relationship between the high dietary live microbe group and the odds of arthritis. Results 12,844 participants were included in the present study. The intake of high live dietary microbes in RA group was lower than that in healthy control group and OA group. The proportion of RA patients in the high live dietary microbe group was lower than that in the low and medium live dietary microbe group. Following the comprehensive adjustment for covariates, it was observed that participants in both the high and medium dietary live microbe groups exhibited lower odds of RA compared to those in the low dietary live microbe group (High OR: 0.71, 95% CI: 0.53-0.96; Medium OR: 0.77, 95% CI: 0.59-1.00, p = 0.02). A restricted cubic spline plot indicates a negative correlation between the quantity of dietary live microbes and the occurrence of RA within the high dietary live microbe group. Conclusion The results of our study revealed a significant difference in dietary live microbe intake between healthy and RA patients. Higher dietary intake was correlated with a decreased odds of RA. However, no significant association was found between the occurrence of OA and the quantity of dietary live microbes.
Collapse
Affiliation(s)
| | | | - Xiaodan Kong
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
46
|
Kan HX, Cao Y, Ma Y, Zhang YL, Wang J, Li J, Li JN. Efficacy and safety of probiotics, prebiotics, and synbiotics for the prevention of colorectal cancer and precancerous lesion in high-risk populations: A systematic review and meta-analysis of randomized controlled trials. J Dig Dis 2024; 25:14-26. [PMID: 38126945 DOI: 10.1111/1751-2980.13247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES Colorectal cancer (CRC) is highly prevalent worldwide and is a leading cause of cancer-related death. Probiotics, prebiotics, and synbiotics have recently attracted attention as preventive measures against colorectal neoplasms. We aimed to analyze the findings of randomized controlled trials (RCTs) on the effects of probiotics, prebiotics, and synbiotics in patients at a high risk of CRC, outlining the challenges and future prospects of using probiotics to prevent colorectal tumors and providing evidence for clinical physicians in particular. METHODS PubMed, EMBASE, and the Cochrane Library databases were searched for relevant studies published up to January 7, 2022. RCTs conducted on populations with a high risk of CRC who received probiotics, prebiotics or synbiotics in comparison with placebo, candidate agent or no treatment were included. The primary outcome was the incidence or recurrence of any colorectal neoplasms. Additional outcomes included their effects on the diversity of gut microbiota and relevant inflammatory biomarkers. Safety outcomes were also analyzed. Two authors independently screened and selected studies based on pre-specified eligible criteria, performed data extraction and risk-of-bias assessment independently. RESULTS Nine RCTs were included in the systematic review and meta-analysis. Probiotic supplementation significantly reduced adenoma incidence, but no significant benefit was observed in CRC incidence. Additionally, probiotics modulated gut microbiota and inflammatory biomarkers. CONCLUSION Probiotics may have beneficial effects in the prevention of CRC. More RCTs with larger sample sizes are warranted to further confirm these findings.
Collapse
Affiliation(s)
- Hao Xuan Kan
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Cao
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Ma
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Lun Zhang
- Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Ji Li
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Jing Nan Li
- Department of Gastroenterology, Key Laboratory of Gut Microbiota Translational Medicine Research, Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
47
|
Van Syoc EP, Damani J, DiMattia Z, Ganda E, Rogers CJ. The Effects of Bifidobacterium Probiotic Supplementation on Blood Glucose: A Systematic Review and Meta-Analysis of Animal Models and Clinical Evidence. Adv Nutr 2024; 15:100137. [PMID: 37923223 PMCID: PMC10831893 DOI: 10.1016/j.advnut.2023.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/21/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Probiotic supplementation is a potential therapeutic for metabolic diseases, including obesity, metabolic syndrome (MetS), and type 2 diabetes (T2D), but most studies deliver multiple species of bacteria in addition to prebiotics or oral pharmaceuticals. This may contribute to conflicting evidence in existing meta-analyses of probiotics in these populations and warrants a systematic review of the literature to assess the contribution of a single probiotic genus to better understand the contribution of individual probiotics to modulate blood glucose. We conducted a systematic review and meta-analysis of animal studies and human randomized controlled trials (RCTs) to assess the effects of Bifidobacterium (BF) probiotic supplementation on markers of glycemia. In a meta-analysis of 6 RCTs, BF supplementation had no effect on fasting blood glucose {FBG; mean difference [MD] = -1.99 mg/dL [95% confidence interval (CI): -4.84, 0.86], P = 0.13}, and there were no subgroup differences between subjects with elevated FBG concentrations and normoglycemia. However, BF supplementation reduced FBG concentrations in a meta-analysis comprised of studies utilizing animal models of obesity, MetS, or T2D [n = 16; MD = -36.11 mg/dL (CI: -49.04, -23.18), P < 0.0001]. Translational gaps from animal to human trials include paucity of research in female animals, BF supplementation in subjects that were normoglycemic, and lack of methodologic reporting regarding probiotic viability and stability. More research is necessary to assess the effects of BF supplementation in human subjects with elevated FBG concentrations. Overall, there was consistent evidence of the efficacy of BF probiotics to reduce elevated FBG concentrations in animal models but not clinical trials, suggesting that BF alone may have minimal effects on glycemic control, may be more effective when combined with multiple probiotic species, or may be more effective in conditions of hyperglycemia rather than elevated FBG concentrations.
Collapse
Affiliation(s)
- Emily P Van Syoc
- Dual-Title Ph.D Program in Integrative & Biomedical Physiology and Clinical & Translational Science, The Pennsylvania State University, University Park, PA, United States; Department of Animal Science, The Pennsylvania State University, University Park, PA, United States; The One Health Microbiome Center, The Pennsylvania State University, University Park, PA, United States
| | - Janhavi Damani
- The Intercollege Graduate Degree Program in Integrative and Biomedical Physiology, Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Zachary DiMattia
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, United States
| | - Erika Ganda
- Department of Animal Science, The Pennsylvania State University, University Park, PA, United States; The One Health Microbiome Center, The Pennsylvania State University, University Park, PA, United States
| | - Connie J Rogers
- Department of Nutritional Sciences, University of Georgia, Athens, GA, United States.
| |
Collapse
|
48
|
Xu X, Xu T, Wei J, Chen T. Gut microbiota: an ideal biomarker and intervention strategy for aging. MICROBIOME RESEARCH REPORTS 2024; 3:13. [PMID: 38841415 PMCID: PMC11149087 DOI: 10.20517/mrr.2023.68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/15/2023] [Indexed: 06/07/2024]
Abstract
Population aging is a substantial challenge for the global sanitation framework. Unhealthy aging tends to be accompanied by chronic diseases such as cardiovascular disease, diabetes, and cancer, which undermine the welfare of the elderly. Based on the fact that aging is inevitable but retarding aging is attainable, flexible aging characterization and efficient anti-aging become imperative for healthy aging. The gut microbiome, as the most dynamic component interacting with the organism, can affect the aging process through its own structure and metabolites, thus holding the potential to become both an ideal aging-related biomarker and an intervention strategy. This review summarizes the value of applying gut microbiota as aging-related microbial biomarkers in diagnosing aging state and monitoring the effect of anti-aging interventions, ultimately pointing to the future prospects of microbial intervention strategies in maintaining healthy aging.
Collapse
Affiliation(s)
- Xuan Xu
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Huankui Academy, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Tangchang Xu
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
- Authors contributed equally
| | - Jing Wei
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Tingtao Chen
- Institution of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, Jiangxi, China
| |
Collapse
|
49
|
Jean Wilson E, Sirpu Natesh N, Ghadermazi P, Pothuraju R, Prajapati DR, Pandey S, Kaifi JT, Dodam JR, Bryan JN, Lorson CL, Watrelot AA, Foster JM, Mansell TJ, Joshua Chan SH, Batra SK, Subbiah J, Rachagani S. Red Cabbage Juice-Mediated Gut Microbiota Modulation Improves Intestinal Epithelial Homeostasis and Ameliorates Colitis. Int J Mol Sci 2023; 25:539. [PMID: 38203712 PMCID: PMC10778654 DOI: 10.3390/ijms25010539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Gut microbiota plays a crucial role in inflammatory bowel diseases (IBD) and can potentially prevent IBD through microbial-derived metabolites, making it a promising therapeutic avenue. Recent evidence suggests that despite an unclear underlying mechanism, red cabbage juice (RCJ) alleviates Dextran Sodium Sulfate (DSS)-induced colitis in mice. Thus, the study aims to unravel the molecular mechanism by which RCJ modulates the gut microbiota to alleviate DSS-induced colitis in mice. Using C57BL/6J mice, we evaluated RCJ's protective role in DSS-induced colitis through two cycles of 3% DSS. Mice were daily gavaged with PBS or RCJ until the endpoint, and gut microbiota composition was analyzed via shotgun metagenomics. RCJ treatment significantly improved body weight (p ≤ 0.001), survival in mice (p < 0.001) and reduced disease activity index (DAI) scores. Further, RCJ improved colonic barrier integrity by enhancing the expression of protective colonic mucins (p < 0.001) and tight junction proteins (p ≤ 0.01) in RCJ + DSS-treated mice compared to the DSS group. Shotgun metagenomic analysis revealed an enrichment of short-chain fatty acids (SCFAs)-producing bacteria (p < 0.05), leading to increased Peroxisome Proliferator-Activated Receptor Gamma (PPAR-γ) activation (p ≤ 0.001). This, in turn, resulted in repression of the nuclear factor κB (NFκB) signaling pathway, causing decreased production of inflammatory cytokines and chemokines. Our study demonstrates colitis remission in a DSS-induced mouse model, showcasing RCJ as a potential modulator for gut microbiota and metabolites, with promising implications for IBD prevention and treatment.
Collapse
Affiliation(s)
- Emily Jean Wilson
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - Nagabhishek Sirpu Natesh
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65201, USA; (N.S.N.); (J.R.D.); (J.N.B.)
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| | - Parsa Ghadermazi
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA; (P.G.)
| | - Ramesh Pothuraju
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dipakkumar R. Prajapati
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Sanjit Pandey
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Jussuf T. Kaifi
- Department of Surgery, School of Medicine, University of Missouri, Columbia, MO 65211, USA;
| | - John R. Dodam
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65201, USA; (N.S.N.); (J.R.D.); (J.N.B.)
| | - Jeffrey N. Bryan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65201, USA; (N.S.N.); (J.R.D.); (J.N.B.)
| | - Christian L. Lorson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO 65211, USA;
| | - Aude A. Watrelot
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA 50011, USA;
| | - Jason M. Foster
- Department of Surgery, Division of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Thomas J. Mansell
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA;
| | - Siu Hung Joshua Chan
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA; (P.G.)
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jeyamkondan Subbiah
- Department of Food Science, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO 65201, USA; (N.S.N.); (J.R.D.); (J.N.B.)
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
50
|
Zhang YS, Juhasz AL, Xi JF, Ma LQ, Zhou D, Li HB. Dietary Galactooligosaccharides Supplementation as a Gut Microbiota-Regulating Approach to Lower Early Life Arsenic Exposure. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:19463-19472. [PMID: 37943691 DOI: 10.1021/acs.est.3c07168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Prebiotics may stimulate beneficial gut microorganisms. However, it remains unclear whether they can lower the oral bioavailability of early life arsenic (As) exposure via regulating gut microbiota and altering As biotransformation along the gastrointestinal (GI) tract. In this study, weanling mice were exposed to arsenate (iAsV) via diet (7.5 μg As g-1) amended with fructooligosaccharides (FOS), galactooligosaccharides (GOS), and inulin individually at 1% and 5% (w/w). Compared to As exposure control mice, As concentrations in mouse blood, liver, and kidneys and As urinary excretion factor (UEF) were reduced by 43.7%-74.1% when treated with 5% GOS. The decrease corresponded to a significant proliferation of Akkermansia and Psychrobacter, reduced percentage of inorganic arsenite (iAsIII) and iAsV by 47.4% and 65.4%, and increased proportion of DMAV in intestinal contents by 101% in the guts of mice treated with 5% GOS compared to the As control group. In contrast, FOS and inulin either at l% or 5% did not reduce As concentration in mouse blood, liver, and kidneys or As UEF. These results suggest that GOS supplementation may be a gut microbiota-regulating approach to lower early life As exposure via stimulating the growth of Akkermansia and Psychrobacter and enhancing As methylation in the GI tract.
Collapse
Affiliation(s)
- Yao-Sheng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Albert L Juhasz
- Future Industries Institute, University of South Australia, Mawson Lakes, South Australia 5095, Australia
| | - Jin-Feng Xi
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Lena Q Ma
- Institute of Soil and Water Resources and Environmental Science, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongmei Zhou
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hong-Bo Li
- State Key Laboratory of Pollution Control and Resource Reuse, Jiangsu Key Laboratory of Vehicle Emissions Control, School of the Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|