1
|
Wang L, Wang R, He Q, Hu Q, Yang J, Tang X. Assessment of the Effectiveness of Probiotics-assisted Physical Interventions in the Management of Chronic Periodontitis: A Randomized Controlled Clinical Trial. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10369-2. [PMID: 39304607 DOI: 10.1007/s12602-024-10369-2] [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: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Live micro-ecological agents, such as probiotics, have demonstrated a significant role in the preservation of human health, encompassing oral health maintenance and regulation of oral microbiota. Here, a total of 20 patients diagnosed with chronic periodontitis were recruited and randomly assigned into two cohorts based on completion of physiotherapy: a placebo group (n = 10) and a probiotic group (n = 10). The actual efficacy was assessed by administering chewable tablets (5 × 109 CFU/tablet) containing the probiotics Lactobacillus salivarius LS97, Lactobacillus paracasei LC86, and Lactobacillus acidophilus LA85 to patients with chronic periodontitis. For the placebo group, chewable tablets without probiotics were administered, while maintaining consistency with the rest of the ingredients used in the probiotic group. Saliva and plaque samples were collected at different time points (0, 1, and 3 months) and subjected to 16S amplicon sequencing for microbial structure analysis. Salivary IgA content was determined using enzyme immunoassay, whereas clinical chronic periodontal pocket depth (PD) and bleeding on probe index (BOP +) were employed to evaluate the actual efficacy of probiotic-assisted physiological intervention in chronic periodontitis treatment. Compared to the placebo group, the probiotic intervention resulted in a significant increase in salivary IgA levels among patients, accompanied by a notable decrease in PD and BOP + levels. Furthermore, the probiotic intervention led to a substantial reduction in Fusobacterium and Porphyromonas counts, while significantly increasing Lactobacillus abundance within the dental plaque microbiota of patients. Importantly, no significant alterations were observed in the overall structure of both salivary and dental plaque microbiota following the probiotic intervention. The administration of this live probiotic agent consistently and significantly enhances the oral immune response in patients with chronic periodontitis, thereby augmenting the effectiveness of physical interventions for this condition. Moreover, it effectively reduces the abundance of pathogenic microbes associated with chronic periodontitis without causing substantial alterations to the salivary and dental plaque microbiota composition. Trial registration: Chinese Clinical Trial Registry (ChiCTR) ( https://www.chictr.org.cn ) under the registration number ChiCTR2300074108.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Ruixue Wang
- Department of Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Qinghui He
- Department of Endodontics, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China.
| | - Jingpeng Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China.
| | - Xuna Tang
- Department of Specialist Clinic, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of Stomatology, Nanjing University, Nanjing, China.
| |
Collapse
|
2
|
Zhao J, Guo Y, Jiang Q, Lan H, Hung WL, Lynch B. Bifidobacterium longum subsp. infantis YLGB-1496-Toxicological evaluation. J Appl Toxicol 2024. [PMID: 39252460 DOI: 10.1002/jat.4688] [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: 03/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/11/2024]
Abstract
Bifidobacterium infantis YLGB-1496, originally isolated from breast milk from a Taiwanese mother, is under study for use as a probiotic. As part of safety assessment, an Ames, in vivo mouse micronucleus, and in vivo mouse spermatocyte chromosome aberration assay were conducted along with a 13-week oral rat toxicity study. B. infantis YLGB-1496 had no activity in any of the genotoxicity assays. Administration of the bacteria to Sprague-Dawley rats at doses ranging from 0 to 1.5 g/kg bw/day had no treatment-related effects on any of the endpoints measured. There appear to be no concerns for translocation or pathogenicity of B. infantis YLGB-1496 based on extensive experience with the species in general. The results of the current investigations support potential use of B. infantis YLGB-1496 as a probiotic in infant formula.
Collapse
Affiliation(s)
- Jian Zhao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Yueyi Guo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, China
| | - Qiuyue Jiang
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Hohhot, China
| | - Barry Lynch
- Intertek Health Sciences Inc., Mississauga, ON, Canada
| |
Collapse
|
3
|
Matera M, Guandalini S. How the Microbiota May Affect Celiac Disease and What We Can Do. Nutrients 2024; 16:1882. [PMID: 38931237 PMCID: PMC11206804 DOI: 10.3390/nu16121882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Celiac disease (CeD) is an autoimmune disease with a strong association with human leukocyte antigen (HLA), characterized by the production of specific autoantibodies and immune-mediated enterocyte killing. CeD is a unique autoimmune condition, as it is the only one in which the environmental trigger is known: gluten, a storage protein present in wheat, barley, and rye. How and when the loss of tolerance of the intestinal mucosa to gluten occurs is still unknown. This event, through the activation of adaptive immune responses, enhances epithelial cell death, increases the permeability of the epithelial barrier, and induces secretion of pro-inflammatory cytokines, resulting in the transition from genetic predisposition to the actual onset of the disease. While the role of gastrointestinal infections as a possible trigger has been considered on the basis of a possible mechanism of antigen mimicry, a more likely alternative mechanism appears to involve a complex disruption of the gastrointestinal microbiota ecosystem triggered by infections, rather than the specific effect of a single pathogen on intestinal mucosal homeostasis. Several lines of evidence show the existence of intestinal dysbiosis that precedes the onset of CeD in genetically at-risk subjects, characterized by the loss of protective bacterial elements that both epigenetically and functionally can influence the response of the intestinal epithelium leading to the loss of gluten tolerance. We have conducted a literature review in order to summarize the current knowledge about the complex and in part still unraveled dysbiosis that precedes and accompanies CeD and present some exciting new data on how this dysbiosis might be prevented and/or counteracted. The literature search was conducted on PubMed.gov in the time frame 2010 to March 2024 utilizing the terms "celiac disease and microbiota", "celiac disease and microbiome", and "celiac disease and probiotics" and restricting the search to the following article types: Clinical Trials, Meta-Analysis, Review, and Systematic Review. A total of 364 papers were identified and reviewed. The main conclusions of this review can be outlined as follows: (1) quantitative and qualitative changes in gut microbiota have been clearly documented in CeD patients; (2) intestinal microbiota's extensive and variable interactions with enterocytes, viral and bacterial pathogens and even gluten combine to impact the inflammatory immune response to gluten and the loss of gluten tolerance, ultimately affecting the pathogenesis, progression, and clinical expression of CeD; (3) gluten-free diet fails to restore the eubiosis of the digestive tract in CeD patients, and also negatively affects microbial homeostasis; (4) new tools allowing targeted microbiota therapy, such as the use of probiotics (a good example being precision probiotics like the novel strain of B. vulgatus (20220303-A2) begin to show exciting potential applications.
Collapse
Affiliation(s)
- Mariarosaria Matera
- Pediatric Clinical Microbiomics Service, Misericordia Hospital, Via Senese 161, 58100 Grosseto, Italy;
| | - Stefano Guandalini
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Celiac Disease Center, University of Chicago Medicine, 5841 S. Maryland Ave. MC 4065, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Junaid M, Lu H, Li Y, Liu Y, Din AU, Qi Z, Xiong Y, Yan J. Novel Synergistic Probiotic Intervention: Transcriptomic and Metabolomic Analysis Reveals Ameliorative Effects on Immunity, Gut Barrier, and Metabolism of Mice during Salmonella typhimurium Infection. Genes (Basel) 2024; 15:435. [PMID: 38674370 PMCID: PMC11050207 DOI: 10.3390/genes15040435] [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: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Salmonella typhimurium (S. typhimurium), a prevalent cause of foodborne infection, induces significant changes in the host transcriptome and metabolome. The lack of therapeutics with minimal or no side effects prompts the scientific community to explore alternative therapies. This study investigates the therapeutic potential of a probiotic mixture comprising Lactobacillus acidophilus (L. acidophilus 1.3251) and Lactobacillus plantarum (L. plantarum 9513) against S. typhimurium, utilizing transcriptome and metabolomic analyses, a novel approach that has not been previously documented. Twenty-four SPF-BALB/c mice were divided into four groups: control negative group (CNG); positive control group (CPG); probiotic-supplemented non-challenged group (LAPG); and probiotic-supplemented Salmonella-challenged group (LAPST). An RNA-sequencing analysis of small intestinal (ileum) tissue revealed 2907 upregulated and 394 downregulated DEGs in the LAPST vs. CPG group. A functional analysis of DEGs highlighted their significantly altered gene ontology (GO) terms related to metabolism, gut integrity, cellular development, and immunity (p ≤ 0.05). The KEGG analysis showed that differentially expressed genes (DEGs) in the LAPST group were primarily involved in pathways related to gut integrity, immunity, and metabolism, such as MAPK, PI3K-Akt, AMPK, the tryptophan metabolism, the glycine, serine, and threonine metabolism, ECM-receptor interaction, and others. Additionally, the fecal metabolic analysis identified 1215 upregulated and 305 downregulated metabolites in the LAPST vs. CPG group, implying their involvement in KEGG pathways including bile secretion, propanoate metabolism, arginine and proline metabolism, amino acid biosynthesis, and protein digestion and absorption, which are vital for maintaining barrier integrity, immunity, and metabolism. In conclusion, these findings suggest that the administration of a probiotic mixture improves immunity, maintains gut homeostasis and barrier integrity, and enhances metabolism in Salmonella infection.
Collapse
Affiliation(s)
- Muhammad Junaid
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Hongyu Lu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yu Liu
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Ahmad Ud Din
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| | - Yi Xiong
- Guangxi Center for Animals Disease Control and Prevention, Nanning 530004, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning 530004, China; (M.J.); (H.L.); (Y.L.); (Y.L.); (Z.Q.)
| |
Collapse
|
5
|
Majeed M, Nagabhushanam K, Arumugam S, Chadalavada N, Seepana J, Annamalai T, Murali A, Prakasan P, Mundkur L. Probiotic Weizmannia coagulans MTCC 5856 as adjunct therapy in children's acute diarrhea-a randomized, double-blind, placebo-controlled study. Front Pediatr 2024; 11:1338126. [PMID: 38269290 PMCID: PMC10806110 DOI: 10.3389/fped.2023.1338126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024] Open
Abstract
Objectives Acute diarrhea in children is generally managed by replacing the lost fluid with oral rehydration solution (ORS). Probiotic supplementation has been reported to reduce the severity of diarrhea. In the present study, we investigated the effect of Weizmannia coagulans (Bacillus coagulans) MTCC 5856, along with ORS on acute diarrhea of all causes in non-hospitalized children. Methods A total of 110 children of ages between 1 and 10 were enrolled in a double-blind placebo-controlled study and were randomly allocated to receive W. coagulans MTCC 5856 (4 × 108 spores, N = 54) + ORS and zinc (Zn) or a placebo (N = 56) + ORS and (Zn) for 5 days. The consistency of the stool, mean duration of diarrhea in hours, mean diarrhea frequency per day, and the dehydration status were collected as efficacy endpoints. Safety was evaluated by the occurrence of adverse events. Results The mean age of the children was 5.55 ± 2.57 years (61 boys and 49 girls). The mean duration of diarrhea was 51.31 ± 20.99 h in the W. coagulans MTCC 5856 group and 62.74 ± 24.51 h in the placebo (p = 0.011) group. The frequency of diarrhea was lower in children supplemented with the probiotic, but the difference was not statistically significant. The perceived efficacy score and dehydration status improved significantly in the W. coagulans MTCC 5856 group compared with the placebo group. No adverse events were recorded. Conclusion The results of the study suggest that W. coagulans MTCC 5856 could be supplemented along with ORS and zinc to reduce the duration of diarrhea in non-hospitalized children. Clinical Trial Registration ClinicalTrials.gov, identifier CTRI/2022/06/043239.
Collapse
Affiliation(s)
- Muhammed Majeed
- Research and Development, Sami-Sabinsa Group Limited, Peenya Industrial Area, Bangalore, India
- Research and Development, Sabinsa Corporation, East Windsor, NJ, United States
| | | | - Sivakumar Arumugam
- Research and Development, Sami-Sabinsa Group Limited, Peenya Industrial Area, Bangalore, India
| | | | - Jyotsna Seepana
- Department of Pediatrics, Government Medical College and Government General Hospital (old RIMSGGH), Srikakulam, India
| | - Thumjaa Annamalai
- Department of Pediatrics, Aarupadai Veedu Medical College & Hospital (AVMCH), Pondicherry, India
| | - Avinash Murali
- Research and Development, Sami-Sabinsa Group Limited, Peenya Industrial Area, Bangalore, India
| | - Priji Prakasan
- Research and Development, Sami-Sabinsa Group Limited, Peenya Industrial Area, Bangalore, India
| | - Lakshmi Mundkur
- Research and Development, Sami-Sabinsa Group Limited, Peenya Industrial Area, Bangalore, India
| |
Collapse
|
6
|
Happel AU, Rametse L, Perumaul B, Diener C, Gibbons SM, Nyangahu DD, Donald KA, Gray C, Jaspan HB. Bifidobacterium infantis supplementation versus placebo in early life to improve immunity in infants exposed to HIV: a protocol for a randomized trial. BMC Complement Med Ther 2023; 23:367. [PMID: 37853370 PMCID: PMC10583347 DOI: 10.1186/s12906-023-04208-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/20/2023] Open
Abstract
INTRODUCTION Infants who are born from mothers with HIV (infants who are HIV exposed but uninfected; iHEU) are at higher risk of morbidity and display multiple immune alterations compared to infants who are HIV-unexposed (iHU). Easily implementable strategies to improve immunity of iHEU, and possibly subsequent clinical health outcomes, are needed. iHEU have altered gut microbiome composition and bifidobacterial depletion, and relative abundance of Bifidobacterium infantis has been associated with immune ontogeny, including humoral and cellular vaccine responses. Therefore, we will assess microbiological and immunological phenotypes and clinical outcomes in a randomized, double-blinded trial of B. infantis Rosell®-33 versus placebo given during the first month of life in South African iHEU. METHODS This is a parallel, randomised, controlled trial. Two-hundred breastfed iHEU will be enrolled from the Khayelitsha Site B Midwife Obstetric Unit in Cape Town, South Africa and 1:1 randomised to receive 8 × 109 CFU B. infantis Rosell®-33 daily or placebo for the first 4 weeks of life, starting on day 1-3 of life. Infants will be followed over 36 weeks with extensive collection of meta-data and samples. Primary outcomes include gut microbiome composition and diversity, intestinal inflammation and microbial translocation and cellular vaccine responses. Additional outcomes include biological (e.g. gut metabolome and T cell phenotypes) and clinical (e.g. growth and morbidity) outcome measures. DISCUSSION The results of this trial will provide evidence whether B. infantis supplementation during early life could improve health outcomes for iHEU. ETHICS AND DISSEMINATION Approval for this study has been obtained from the ethics committees at the University of Cape Town (HREC Ref 697/2022) and Seattle Children's Research Institute (STUDY00003679). TRIAL REGISTRATION Pan African Clinical Trials Registry Identifier: PACTR202301748714019. CLINICAL TRIALS gov: NCT05923333. PROTOCOL VERSION Version 1.8, dated 18 July 2023.
Collapse
Affiliation(s)
- Anna-Ursula Happel
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa.
| | - Lerato Rametse
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Brandon Perumaul
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | | | - Sean M Gibbons
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
- eScience Institute, University of Washington, Seattle, WA, 98195, USA
| | - Donald D Nyangahu
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
| | - Kirsten A Donald
- Division of Developmental Paediatrics, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Klipfontein Road Rondebosch, Cape Town, 7700, South Africa
- The Neuroscience Institute, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
| | - Clive Gray
- Division of Molecular Biology and Human Genetics, Stellenbosch University, Francie Van Zijl Drive, Tygerberg, 7505, South Africa
| | - Heather B Jaspan
- Department of Pathology, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Anzio Road, Observatory, Cape Town, 7925, South Africa
- Seattle Children's Research Institute, 307 Westlake Ave. N, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA
- Department of Global Health, University of Washington, 1510 San Juan Road NE, Seattle, WA, 98195, USA
| |
Collapse
|
7
|
Saviano A, Petruzziello C, Brigida M, Morabito Loprete MR, Savioli G, Migneco A, Ojetti V. Gut Microbiota Alteration and Its Modulation with Probiotics in Celiac Disease. Biomedicines 2023; 11:2638. [PMID: 37893012 PMCID: PMC10603901 DOI: 10.3390/biomedicines11102638] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/03/2023] [Accepted: 08/14/2023] [Indexed: 10/29/2023] Open
Abstract
Celiac disease (CD) is a chronic inflammation of the small intestine triggered by gluten ingestion in genetically predisposed people. Recent literature studies highlight the possible role of the gut microbiota in the pathogenesis of this disease. The gut microbiota is a complex community of microorganisms that can interact with the innate and adaptative immune systems. A condition of dysbiosis, which refers to an alteration in the composition and function of the human gut microbiota, can lead to a dysregulated immune response. This condition may contribute to triggering gluten intolerance, favoring the development and/or progression of CD in genetically susceptible patients. Interestingly, studies on children and adults with CD showed a different microbiome profile in fecal samples, with a different degree of "activity" for the disease. From this point of view, our review aimed to collect and discuss modern evidence about the alteration of the gut microbiota and its modulation with probiotics, with possible future indications in the management of patients affected by CD.
Collapse
Affiliation(s)
- Angela Saviano
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (A.S.); (A.M.)
| | - Carmine Petruzziello
- Department of Emergency Medicine, Ospedale San Carlo di Nancy, GVM Care and Research, 00165 Rome, Italy
| | - Mattia Brigida
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy
| | - Maria Rita Morabito Loprete
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (A.S.); (A.M.)
| | - Gabriele Savioli
- Department of Emergency Medicine, Fondazione Policlinico San Matteo University Hospital, 27100 Pavia, Italy;
| | - Alessio Migneco
- Department of Emergency Medicine, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy; (A.S.); (A.M.)
| | - Veronica Ojetti
- Department of Emergency Medicine-Fondazione Policlinico Universitario A. Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
8
|
Sankova MV, Nikolenko VN, Sankov SV, Sinelnikov MY. SARS-CoV-2 and microbiome. AUTOIMMUNITY, COVID-19, POST-COVID19 SYNDROME AND COVID-19 VACCINATION 2023:279-337. [DOI: 10.1016/b978-0-443-18566-3.00023-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Gut Microbiota and Enteral Nutrition Tolerance in Non-Abdominal Infection Septic ICU Patients: An Observational Study. Nutrients 2022; 14:nu14245342. [PMID: 36558501 PMCID: PMC9783285 DOI: 10.3390/nu14245342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
Background: The effect of gut microbiota on enteral nutrition tolerance in critically ill patients is unclear. Methods: Non-abdominal sepsis patients in an ICU, sorted by whether they reached 20 Kcal/kg/day on the 3rd day of EN, were divided into tolerance and intolerance groups. Their feces on day 1 and day 3 of EN initiation were collected for 16s rDNA and short-chain fatty acid (SCFA) testing. Results: There were 14 patients included in the tolerance group and 10 in the intolerance group. On EN day 1, the OTUs and microbiota diversity were higher in the tolerance group than in the intolerance group. The ratio of Firmicutes to Bacteroidetes was higher in the intolerance group on EN day 1. The genus Parabacteroides were the most significantly elevated in the tolerance group. On EN day 3, the genus Escherichia-Shigella was the most significantly elevated in the tolerance group. On EN day 3, the levels of SCFA decreased more significantly in the intolerance group. Conclusion: Enteral nutrition tolerance is associated with microbiota features and short-chain fatty acid levels. A higher ratio of Firmicutes to Bacteroidetes and microbiota diversity on EN day 1 may help in the early prediction of EN tolerance.
Collapse
|
10
|
The Relationship between Gastrointestinal Health, Micronutrient Concentrations, and Autoimmunity: A Focus on the Thyroid. Nutrients 2022; 14:nu14173572. [PMID: 36079838 PMCID: PMC9460308 DOI: 10.3390/nu14173572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Currently, there is a lack of understanding of why many patients with thyroid dysfunction remain symptomatic despite being biochemically euthyroid. Gastrointestinal (GI) health is imperative for absorption of thyroid-specific nutrients as well as thyroid function directly. This comprehensive narrative review describes the impact of what the authors have conceptualized as the “nutrient–GI–thyroid axis”. Compelling evidence reveals how gastrointestinal health could be seen as the epicenter of thyroid-related care given that: (1) GI conditions can lower thyroid-specific nutrients; (2) GI care can improve status of thyroid-specific nutrients; (3) GI conditions are at least 45 times more common than hypothyroidism; (4) GI care can resolve symptoms thought to be from thyroid dysfunction; and (5) GI health can affect thyroid autoimmunity. A new appreciation for GI health could be the missing link to better nutrient status, thyroid status, and clinical care for those with thyroid dysfunction.
Collapse
|
11
|
Shen W, Qiu W, Lin Q, Zeng C, Liu Y, Huang W, Zhou H. The Gut Microbiome of Preterm Infants Treated With Aminophylline Is Closely Related to the Occurrence of Feeding Intolerance and the Weight Gain. Front Nutr 2022; 9:905839. [PMID: 35719163 PMCID: PMC9198222 DOI: 10.3389/fnut.2022.905839] [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: 03/28/2022] [Accepted: 04/29/2022] [Indexed: 11/21/2022] Open
Abstract
Background Aminophylline is widely used in the treatment of preterm infants, but it can cause feeding intolerance events, in which gut microbial dysbiosis may have a role. This study aims to investigate the relationship between the gut microbiome of preterm infants treated with aminophylline and the occurrence of feeding intolerance and weight gain rate. Methods This study included a cohort of 118 preterm infants. Survival analysis and multivariate Cox regression were used to evaluate the relationship between aminophylline treatment and the occurrence of feeding intolerance. 16S rRNA V4 region gene sequencing was used to characterize the microbiome of fecal samples from the cohort. Linear discriminant analysis effect size was used to analyze the differential abundance of bacteria related to aminophylline treatment. Wilcoxon test, Kruskal–Wallis test, Spearman correlation coefficients and generalized linear mixed models were used to analyze the correlation between the differential bacteria and feeding intolerance events as well as the weight gain. Results The results showed that the use of aminophylline could significantly increase the occurrence of feeding intolerance. The relative abundances of Streptococcus and Rothia in the gut microbiome of preterm infants were positively correlated with both the occurrence of feeding intolerance and the use of aminophylline, while the relative abundance of Staphylococcus was negatively correlated. In particular, preterm infants with a lower relative abundance of Rothia were more likely to develop feeding intolerance associated with aminophylline, and this difference existed before the onset of feeding intolerance. Moreover, it took longer for individuals with a lower relative abundance of Streptococcus to reach 2 kg weight. The contribution of Streptococcus to weight gain was greater than that of Bifidobacterium or Lactobacillus. Conclusion The gut microbiome in preterm infants treated with aminophylline was characterized by a decrease in Streptococcus and Rothia and an increase in Staphylococcus. These microbes, especially Rothia, were positively correlated with the occurrence of feeding intolerance. Streptococcus but not Bifidobacter likely participated in the weight gain of preterm infants in early life.
Collapse
Affiliation(s)
- Wei Shen
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen Qiu
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Lin
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chao Zeng
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Liu
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weimin Huang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Effects of probiotics on immunity and iron homeostasis: A mini-review. Clin Nutr ESPEN 2022; 49:24-27. [PMID: 35623819 DOI: 10.1016/j.clnesp.2022.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/23/2022] [Accepted: 03/19/2022] [Indexed: 02/07/2023]
Abstract
Iron deficiency remains a major problem in both developed and developing countries. Iron supplementation has been used as a standard intervention for the prevention and treatment of iron deficiency anemia (IDA). There are many factors affecting the efficacy, including stunting, infections or inflammations, and genetics. Recently, some studies have been conducted to further investigate the effects of probiotics on immunity and iron homeostasis. This mini review discusses about some important factors that can improve the management of IDA.
Collapse
|
13
|
Nogacka AM, Arboleya S, Nikpoor N, Auger J, Salazar N, Cuesta I, Alvarez-Buylla JR, Mantecón L, Solís G, Gueimonde M, Tompkins TA, de los Reyes-Gavilán CG. In Vitro Probiotic Modulation of the Intestinal Microbiota and 2′Fucosyllactose Consumption in Fecal Cultures from Infants at Two Months of Age. Microorganisms 2022; 10:microorganisms10020318. [PMID: 35208773 PMCID: PMC8876326 DOI: 10.3390/microorganisms10020318] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/26/2022] [Accepted: 01/26/2022] [Indexed: 01/17/2023] Open
Abstract
2′-fucosyllactose (2′FL) is one of the most abundant oligosaccharides in human milk, with benefits on neonatal health. Previous results point to the inability of the fecal microbiota from some infants to ferment 2′FL. We evaluated a probiotic formulation, including the strains Lactobacillus helveticus Rosell®-52 (R0052), Bifidobacterium longum subsp. infantis Rosell®-33 (R0033), and Bifidobacterium bifidum Rosell®-71 (R0071), individually or in an 80:10:10 combination on the microbiota and 2′FL degradation. Independent batch fermentations were performed with feces from six full-term infant donors of two months of age (three breastfed and three formula-fed) with added probiotic formulation or the constituent strains in the presence of 2′FL. Microbiota composition was analyzed by 16S rRNA gene sequencing. Gas accumulation, pH decrease and 2′FL consumption, and levels of different metabolites were determined by chromatography. B. bifidum R0071 was the sole microorganism promoting a partial increase of 2′FL degradation during fermentation in fecal cultures of 2′FL slow-degrading donors. However, major changes in microbiota composition and metabolic activity occurred with L. helveticus R0052 or the probiotic formulation in cultures of slow degraders. Further studies are needed to decipher the role of the host intestinal microbiota in the efficacy of these strains.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Naghmeh Nikpoor
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Jeremie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Jorge R. Alvarez-Buylla
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
| | - Laura Mantecón
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Gonzalo Solís
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
| | - Thomas A. Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, QC H4P 2R2, Canada; (N.N.); (J.A.); (T.A.T.)
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (S.A.); (N.S.); (I.C.); (J.R.A.-B.); (M.G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain; (L.M.); (G.S.)
- Correspondence: (A.M.N.); (C.G.d.l.R.-G.); Tel.: +34-985-89-21-31 (A.M.N.)
| |
Collapse
|
14
|
Lee H, Li Z, Christensen B, Peng Y, Li X, Hernell O, Lönnerdal B, Slupsky CM. Metabolic Phenotype and Microbiome of Infants Fed Formula Containing Lactobacillus paracasei Strain F-19. Front Pediatr 2022; 10:856951. [PMID: 35558362 PMCID: PMC9087039 DOI: 10.3389/fped.2022.856951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Early childhood nutrition drives the development of the gut microbiota. In contrast to breastfeeding, feeding infant formula has been shown to impact both the gut microbiota and the serum metabolome toward a more unfavorable state. It is thought that probiotics may alter the gut microbiota and hence create a more favorable metabolic outcome. To investigate the impact of supplementation with Lactobacillus paracasei spp. paracasei strain F-19 on the intestinal microbiota and the serum metabolome, infants were fed a formula containing L. paracasei F19 (F19) and compared to a cohort of infants fed the same standard formula without the probiotic (SF) and a breast-fed reference group (BF). The microbiome, as well as serum metabolome, were compared amongst groups. Consumption of L. paracasei F19 resulted in lower community diversity of the gut microbiome relative to the SF group that made it more similar to the BF group at the end of the intervention (4 months). It also significantly increased lactobacilli and tended to increase bifidobacteria, also making it more similar to the BF group. The dominant genus in the microbiome of all infants was Bifidobacterium throughout the intervention, which was maintained at 12 months. Although the serum metabolome of the F19 group was more similar to the group receiving the SF than the BF group, increases in serum TCA cycle intermediates and decreases in several amino acids in the metabolome of the F19 group were observed, which resulted in a metabolome that trended toward the BF group. Overall, L. paracasei F19 supplementation did not override the impact of formula-feeding but did impact the microbiome and the serum metabolome in a way that may mitigate some unfavorable metabolic impacts of formula-feeding.
Collapse
Affiliation(s)
- Hanna Lee
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Zailing Li
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | | | - Yongmei Peng
- Department of Child Health Care, Children's Hospital, Fudan University, Shanghai, China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Carolyn M Slupsky
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
15
|
Sornsenee P, Chatatikun M, Mitsuwan W, Kongpol K, Kooltheat N, Sohbenalee S, Pruksaphanrat S, Mudpan A, Romyasamit C. Lyophilized cell-free supernatants of Lactobacillus isolates exhibited antibiofilm, antioxidant, and reduces nitric oxide activity in lipopolysaccharide-stimulated RAW 264.7 cells. PeerJ 2021; 9:e12586. [PMID: 34909285 PMCID: PMC8641486 DOI: 10.7717/peerj.12586] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/11/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Probiotics can release bioactive substances known as postbiotics, which can inhibit pathogenic microorganisms, improve immunomodulation, reduce antioxidant production, and modulate the gut microbiota. METHODS In this study, we evaluated the in vitro antimicrobial effects, antioxidant activity, and anti-inflammatory potential of 10 lyophilized cell-free supernatants (LCFS) of Lactobacillus isolates. LCFS was obtained via centrifugation and subsequent lyophilization of the supernatant collected from the culture medium ofeach isolate. The antibacterial and antibiofilm activities of the LCFS were determined using broth microdilution. The antioxidant potential was evaluated by measuring the total phenolic and flavonoid contents and 2,2-Diphennyl-1-picrylhydrazyl (DPPH) and 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) radical cation (ABTS+) radical scavenging activities. RESULTS All the isolates were able to inhibit the four tested pathogens. The isolates exhibited strong antibiofilm activity and eradicated the biofilms formed by Acinetobacter buamannii and Escherichia coli. All the prepared Lactobacillus LCFS contained phenols and flavonoids and exhibited antioxidant activities in the DPPH and ABTS+ radical scavenging assays. The MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay revealed that LCFS was not cytotoxic to RAW 264.7 cells. In addition, the ten Lactobacillus LCFS decreased the production of nitric oxide. CONCLUSIONS All the isolates have beneficial properties. This research sheds light on the role of postbiotics in functional fermented foods and pharmaceutical products. Further research to elucidate the precise molecular mechanisms of action of probiotics is warranted.
Collapse
Affiliation(s)
- Phoomjai Sornsenee
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Moragot Chatatikun
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Center of Excellence Research for Meliodosis (CERM), Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Research Excellence Center for Innovation and Health Product, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Watcharapong Mitsuwan
- Research Center of Excellence in Innovation of Essential Oil, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- One Health Research Center, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Akkhraratchakumari Veterinary College, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Kantapich Kongpol
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Research Excellence Center for Innovation and Health Product, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Nateelak Kooltheat
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Research Excellence Center for Innovation and Health Product, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Sasirat Sohbenalee
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Supawita Pruksaphanrat
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Amron Mudpan
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| | - Chonticha Romyasamit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Center of Excellence Research for Meliodosis (CERM), Walailak University, Thasala, Nakhon Si Thammarat, Thailand
- Research Center of Excellence in Innovation of Essential Oil, Walailak University, Thasala, Nakhon Si Thammarat, Thailand
| |
Collapse
|
16
|
Li Z, Zhu G, Li C, Lai H, Liu X, Zhang L. Which Probiotic Is the Most Effective for Treating Acute Diarrhea in Children? A Bayesian Network Meta-Analysis of Randomized Controlled Trials. Nutrients 2021; 13:nu13124319. [PMID: 34959871 PMCID: PMC8706888 DOI: 10.3390/nu13124319] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 12/13/2022] Open
Abstract
Acute diarrhea is a major cause of morbidity and mortality in children under five. Probiotics are beneficial for treating acute diarrhea in children, but unclear which specific probiotic is the most effective. We performed a Bayesian network meta-analysis to examine the comparative effectiveness of probiotics. By searching EMBASE, PubMed, and the Cochrane Library up to 31 March 2021, randomized clinical trials (RCTs) on probiotics for treating acute diarrhea in children were included. Primary outcomes included the duration of diarrhea and diarrhea lasting ≥2 days, and secondary outcomes included the mean stool frequency on day 2 and duration of hospitalization, fever, and vomiting. We assessed the certainty of the evidence of outcomes according to Grading of Recommendations Assessment, Development, and Evaluation (GRADE) guideline. Eighty-four studies with twenty-one different interventions in 13,443 children were included. For the primary outcomes, moderate evidence indicated that, Lactobacillus reuteri [mean difference (MD) = -0.84 day; 95% confidence interval (CI), -1.39, -0.29], Bifidobacterium lactis (MD = -0.98 day; 95%CI, -1.82, -0.14), Saccharomyces boulardii (MD = -1.25 day; 95%CI, -1.59, -0.91), Lactobacillus species (spp.) plus Bifidobacterium spp. plus Saccharomyces spp. (MD = -1.19 day; 95%CI, -1.81, -0.58), and Bacillus spp. plus Enterococcus spp. plus Clostridium spp. (MD = -1.1 day; 95%CI, -1.84, -0.35) significantly reduced the duration of diarrhea when compared with placebo. Saccharomyces boulardii [Odds ratio (OR) = 0.22; 95%CI, 0.11, 0.41] and Lactobacillus reuteri (OR = 0.23; 95%CI, 0.090, 0.60) significantly reduced the risk of diarrhea lasting ≥2 days when compared with placebo or no treatment, with moderate evidence. Among all probiotics, Saccharomyces boulardii may be the most effective in reducing both duration of diarrhea (compared with placebo) and risk of diarrhea lasting ≥2 days (compared with placebo or no treatment), with moderate evidence. To be conclusive, Saccharomyces boulardii may be the most effective probiotic for treating acute diarrhea in children, followed by several other single-strain and multi-strain probiotics.
Collapse
Affiliation(s)
- Zengbin Li
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Guixian Zhu
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Chao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (C.L.); (X.L.)
| | - Hao Lai
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
| | - Xin Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Global Health Institute, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (C.L.); (X.L.)
| | - Lei Zhang
- China-Australia Joint Research Center for Infectious Diseases, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (Z.L.); (G.Z.); (H.L.)
- Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC 3053, Australia
- Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: ; Tel.: +86-29-82655135
| |
Collapse
|