1
|
EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA), Turck D, Bohn T, Cámara M, Castenmiller J, De Henauw S, Jos Á, Maciuk A, Mangelsdorf I, McNulty B, Naska A, Pentieva K, Siani A, Thies F, Aguilera‐Gómez M, Cubadda F, Frenzel T, Heinonen M, Knutsen HK, Neuhäuser‐Berthold M, Poulsen M, Prieto Maradona M, Schlatter JR, Siskos A, van Loveren H, Ackerl R, Kagkli DM, Hirsch‐Ernst KI. Safety of Clostridium butyricum TO-A as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J 2025; 23:e9371. [PMID: 40321162 PMCID: PMC12046288 DOI: 10.2903/j.efsa.2025.9371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver an opinion on Clostridium butyricum TO-A as a novel food (NF) pursuant to Regulation (EU) 2015/2283. The NF, which is proposed by the applicant to be used as a food supplement, is sufficiently characterised. The information provided on the production process, composition, stability and specifications of the NF is sufficient and does not raise safety concerns. Based on the findings of a repeated dose 90-day oral toxicity study in rats, and considering an uncertainty factor of 200, the Panel estimated a safe dose in humans of 4.5 × 106 colony forming unit (CFU)/kg body weight (bw) per day. However, considering that appropriate initial bacteria colonisation of the gastrointestinal tract in humans, in particular during the first 3 years of life, profoundly affects health during infancy and childhood, that disruptions to the microbiota early in life can have lasting health effects into adulthood, and taking into account that the 90-day oral toxicity study was conducted in adult rats, the Panel considers that the target population for the NF should be restricted to children above 3 years of age, adolescents and adults, excluding pregnant and lactating women. The Panel concludes that the NF, C. butyricum TO-A, is safe at 1.0 × 108 CFU/day for other children (3 to < 10 years), 2.0 × 108 CFU/day for adolescents from 10 to < 14 years, 2.8 × 108 CFU/day for adolescents from 14 to < 18 years and 3.2 × 108 CFU/day for adults, excluding pregnant and lactating women.
Collapse
|
2
|
Honda S, Eguchi H, Okino Y, Wang DS. The Probiotic Strain Clostridium butyricum TO-A Produces Butyrate by Utilizing Lactate and Acetate. Int J Mol Sci 2025; 26:2951. [PMID: 40243571 PMCID: PMC11988312 DOI: 10.3390/ijms26072951] [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: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Lactate-utilizing bacteria (LUB) are intestinal bacteria that produce butyrate from lactate and acetate, key metabolites in the gut. As LUB help maintain lactate and butyrate concentrations in the intestinal tract, they are promising probiotic candidates. Clostridium butyricum TO-A (CBTOA) has reportedly been effective in treating various gastrointestinal issues in humans and animals. Although CBTOA is known to increase intestinal butyrate levels, it is unclear how it utilizes lactate and acetate, similar to LUB, to produce butyrate. We investigated lactate utilization-related genes in CBTOA and examined the relationship between lactate and acetate utilization and butyrate production using peptone-yeast medium supplemented with D-lactate, L-lactate, and/or acetate. This study demonstrates for the first time that the probiotic strain CBTOA harbors lactate utilization-related genes and efficiently produces butyrate only in the presence of exogenous lactate and acetate instead of sugars. Furthermore, CBTOA expresses a lactate racemase that enables the bacterium to utilize both lactate enantiomers while regulating the ratio of D-lactate to L-lactate in the intestinal microenvironment via racemization. In conclusion, CBTOA efficiently produces butyrate utilizing lactate and acetate, similar to LUB; therefore, CBTOA could be an efficient butyrate supplier as a probiotic strain in the intestinal tract.
Collapse
Affiliation(s)
- Shotaro Honda
- Research Division, TOA Biopharma Co., Ltd., 606 Kondoh-cho, Tatebayashi 374-0042, Japan (Y.O.); (D.-S.W.)
| | | | | | | |
Collapse
|
3
|
Kim MJ, Lee YJ, Hussain Z, Park H. Effect of Probiotics on Improving Intestinal Mucosal Permeability and Inflammation after Surgery. Gut Liver 2025; 19:207-218. [PMID: 39327843 PMCID: PMC11907258 DOI: 10.5009/gnl240170] [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: 04/18/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 09/28/2024] Open
Abstract
Background/Aims We explored the mechanisms underlying the improvement of postoperative ileus (POI) following probiotic pretreatment. We assessed intestinal permeability, inflammation, tight junction (TJ) protein expression in the gut epithelium, and plasma interleukin (IL)-17 levels in a guinea pig model of POI. Methods Guinea pigs were divided into control, POI, and probiotic groups. The POI and probiotic groups underwent surgery, but the probiotic group received probiotics before the procedure. The ileum and proximal colon were harvested. Intestinal permeability was measured via horseradish peroxidase permeability. Inflammation was evaluated via leukocyte count in the intestinal wall muscle layer, and calprotectin expression in each intestinal wall layer was analyzed immunohistochemically. TJ proteins were analyzed using immunohistochemical staining, and plasma IL-17 levels were measured using an enzyme-linked immunosorbent assay. Results The POI group exhibited increased intestinal permeability and inflammation, whereas probiotic pretreatment reduced the extent of these POI-induced changes. Probiotics restored the expression of TJ proteins occludin and zonula occludens-1 in the proximal colon, which were increased in the POI group. Calprotectin expression significantly increased in the muscle layer of the POI group and was downregulated in the probiotic group; however, no distinct differences were observed between the mucosal and submucosal layers. Plasma IL-17 levels did not significantly differ among the groups. Conclusions Probiotic pretreatment may relieve POI by reducing intestinal permeability and inflammation and TJ protein expression in the gut epithelium. These findings suggest a potential therapeutic approach for POI management.
Collapse
Affiliation(s)
- Min-Jae Kim
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Zahid Hussain
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Division of Gastroenterology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
4
|
Nakatsukasa H, Takahashi M, Shibano M, Ishigami Y, Kawaguchi T, Nakamura Y, Kaneda H. Clinical impact of concomitant BIO-three use in advanced or recurrent non-small cell lung cancer treated with immune-checkpoint inhibitor. Int J Clin Oncol 2024; 29:1840-1849. [PMID: 39278980 DOI: 10.1007/s10147-024-02622-z] [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: 04/30/2024] [Accepted: 09/01/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been approved as first-line therapy for advanced non-small cell lung cancer (NSCLC). The probiotic MIYAIRI 588 can potentially improve the outcomes of patients with advanced NSCLC treated with ICI. However, the impact of other probiotics on ICI-treatment efficacy remains unclear. Thus, we aimed to clarify the association between BIO-three use and treatment outcomes in patients with advanced NSCLC treated with ICI. METHODS This retrospective study included patients aged ≥ 18 years with advanced or recurrent NSCLC who had received ICI monotherapy or ICI plus chemotherapy. Concomitant therapy with probiotic bacteria was defined as receiving it within 180 days before ICI therapy. RESULTS Here, 289 patients were enrolled, including 23 (8.0%) receiving BIO-three. In the multivariable analysis, the progression-free survival (PFS) and overall survival (OS) of patients receiving BIO-three tended to be longer than those of patients not receiving probiotic therapy (PFS, hazard ratio [HR] 0.75; 95% confidence interval [CI] 0.43-1.30; p = 0.33; OS, HR 0.69; 95% CI 0.37-1.28; p = 0.24). After propensity score matching with weighted adjustment, patients receiving BIO-three tended to have prolonged PFS (median PFS [range] 7.6 months [2.6-17.4] vs 3.2 months [1.6-7.0]; HR 0.53; 95% CI 0.25-1.12; p = 0.09) and OS (median OS [range] 25.6 months [10.8-not reached] vs 10.9 months [7.3-not reached]; HR 0.57; 95% CI 0.24-1.36; p = 0.20) than those not receiving probiotic therapy. CONCLUSION This study suggests the prognostic impact of concomitant BIO-three use in patients with advanced NSCLC on ICI treatment.
Collapse
Affiliation(s)
- Hitomi Nakatsukasa
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masaya Takahashi
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Quality and Safety Management, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masahito Shibano
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Yusuke Ishigami
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasutaka Nakamura
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| |
Collapse
|
5
|
Bourqqia-Ramzi M, Mansilla-Guardiola J, Muñoz-Rodriguez D, Quarta E, Lombardo-Hernandez J, Murciano-Cespedosa A, Conejero-Meca FJ, Mateos González Á, Geuna S, Garcia-Esteban MT, Herrera-Rincon C. From the Microbiome to the Electrome: Implications for the Microbiota-Gut-Brain Axis. Int J Mol Sci 2024; 25:6233. [PMID: 38892419 PMCID: PMC11172653 DOI: 10.3390/ijms25116233] [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: 04/09/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The gut microbiome plays a fundamental role in metabolism, as well as the immune and nervous systems. Microbial imbalance (dysbiosis) can contribute to subsequent physical and mental pathologies. As such, interest has been growing in the microbiota-gut-brain brain axis and the bioelectrical communication that could exist between bacterial and nervous cells. The aim of this study was to investigate the bioelectrical profile (electrome) of two bacterial species characteristic of the gut microbiome: a Proteobacteria Gram-negative bacillus Escherichia coli (E. coli), and a Firmicutes Gram-positive coccus Enterococcus faecalis (E. faecalis). We analyzed both bacterial strains to (i) validate the fluorescent probe bis-(1,3-dibutylbarbituric acid) trimethine oxonol, DiBAC4(3), as a reliable reporter of the changes in membrane potential (Vmem) for both bacteria; (ii) assess the evolution of the bioelectric profile throughout the growth of both strains; (iii) investigate the effects of two neural-type stimuli on Vmem changes: the excitatory neurotransmitter glutamate (Glu) and the inhibitory neurotransmitter γ-aminobutyric acid (GABA); (iv) examine the impact of the bioelectrical changes induced by neurotransmitters on bacterial growth, viability, and cultivability using absorbance, live/dead fluorescent probes, and viable counts, respectively. Our findings reveal distinct bioelectrical profiles characteristic of each bacterial species and growth phase. Importantly, neural-type stimuli induce Vmem changes without affecting bacterial growth, viability, or cultivability, suggesting a specific bioelectrical response in bacterial cells to neurotransmitter cues. These results contribute to understanding the bacterial response to external stimuli, with potential implications for modulating bacterial bioelectricity as a novel therapeutic target.
Collapse
Affiliation(s)
- Marwane Bourqqia-Ramzi
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, 10126 Turin, Italy
| | - Jesús Mansilla-Guardiola
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
- Unit of Microbiology, Department of Genetic, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - David Muñoz-Rodriguez
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
| | - Elisa Quarta
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Torino, 10126 Turin, Italy
| | - Juan Lombardo-Hernandez
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
| | - Antonio Murciano-Cespedosa
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
- Neuro-Computing and Neuro-Robotics Research Group, Neural Plasticity Research Group Instituto Investigación Sanitaria Hospital Clínico San Carlos (IdISSC), Complutense University of Madrid, 28040 Madrid, Spain
| | - Francisco José Conejero-Meca
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
| | - Álvaro Mateos González
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
- University of Michigan-Shanghai Jiao Tong University Joint Institute, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Stefano Geuna
- Department of Clinical and Biological Sciences, Cavalieri Ottolenghi Neuroscience Institute, University of Turin, Ospedale San Luigi, 10043 Turin, Italy
| | - María Teresa Garcia-Esteban
- Unit of Microbiology, Department of Genetic, Physiology and Microbiology, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - Celia Herrera-Rincon
- Modeling, Data Analysis &Computational Tools for Biology Research Group, Biomathematics Unit, Department of Biodiversity, Ecology & Evolution, Faculty of Biological Sciences, Complutense University of Madrid, 28040 Madrid, Spain; (M.B.-R.); (J.M.-G.)
| |
Collapse
|
6
|
Boeder AM, Spiller F, Carlstrom M, Izídio GS. Enterococcus faecalis: implications for host health. World J Microbiol Biotechnol 2024; 40:190. [PMID: 38702495 DOI: 10.1007/s11274-024-04007-w] [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: 03/09/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
The microbiota represents a crucial area of research in maintaining human health due to its potential for uncovering novel biomarkers, therapies, and molecular mechanisms relevant to population identification and experimental model characterization. Among these microorganisms, Enterococcus faecalis, a Gram-positive bacterium found in the gastrointestinal tract of humans and animals, holds particular significance. Strains of this bacterial species have sparked considerable debate in the literature due to their dual nature; they can either be utilized as probiotics in the food industry or demonstrate resistance to antibiotics, potentially leading to severe illness, disability, and death. Given the diverse characteristics of Enterococcus faecalis strains, this review aims to provide a comprehensive understanding of their impact on various systems within the host, including the immunological, cardiovascular, metabolic, and nervous systems. Furthermore, we summarize the bacterium-host interaction characteristics and molecular effects to highlight their targets, features, and overall impact on microbial communities and host health.
Collapse
Affiliation(s)
- Ariela Maína Boeder
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Fernando Spiller
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Mattias Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Geison Souza Izídio
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil.
- Department of Cell Biology, Embryology and Genetics, Federal University of Santa Catarina, Florianópolis, Brazil.
- Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, Barcelona, Spain.
- Laboratório de Genética do Comportamento, Universidade Federal de Santa Catarina, Centro de Ciências Biológicas, Departamento de Biologia Celular, Embriologia e Genética, Florianopolis, SC, Brazil.
| |
Collapse
|
7
|
Higuchi T, Furuichi M, Maeda N, Tsugawa T, Ito K. Effects of probiotics in children with acute gastroenteritis: A systematic review and meta-analysis focusing on probiotics utilized in Japan. J Infect Chemother 2024; 30:337-342. [PMID: 37956795 DOI: 10.1016/j.jiac.2023.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Many randomized controlled trials and systematic reviews have evaluated the use of probiotics to treat acute infectious gastroenteritis. However, most probiotic species evaluated in previous large randomized controlled trials are unavailable in Japan. Our objective was to investigate the efficacy of probiotics utilized in Japan for acute gastroenteritis. METHODS The inclusion criterion was a randomized controlled study that compared probiotics with a placebo to treat children younger than 18 years with acute infectious gastroenteritis. We excluded studies that did not contain the following species available in Japan: Bifidobacterium spp., Lactobacillus acidophilus, Enterococcus faecium, Clostridium butyricum, and Bacillus subtilis and studies in low- or lower-middle-income countries. We searched PubMed, CENTRAL, and Igaku Chuo Zasshi from their inception to November 27, 2022. After the risk of bias assessment, data on diarrhea duration, number of hospitalizations, length of hospital stay, and adverse effects were extracted. RESULTS Fourteen studies were included in this meta-analysis. Diarrhea lasting longer than 48 h (7 articles, n = 878) was significantly lower in the probiotic group (risk ratio (RR) 0.70, 95 % confidence interval (CI) 0.59-0.83). The duration of diarrhea (14 articles; n = 1761) was 23.45 h (95 % CI 18.22-26.69) shorter in the probiotic group. Duration of hospitalization (6 articles; n = 971) was 17.73 h (95 % CI 6.9-28.56) shorter in the probiotic group. CONCLUSIONS Although the certainty of evidence is very low, the use of probiotics for acute gastroenteritis in children may improve diarrhea approximately one day earlier. This study was registered with PROSPERO (CRD 42023405559).
Collapse
Affiliation(s)
- Toru Higuchi
- Department of General Pediatrics, Aichi Children's Health and Medical Center, Aichi, Japan
| | - Munehiro Furuichi
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan.
| | - Naonori Maeda
- Department of Pediatrics, National Hospital Organization, Tokyo Medical Center, Tokyo, Japan
| | - Takeshi Tsugawa
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenta Ito
- Department of General Pediatrics, Aichi Children's Health and Medical Center, Aichi, Japan
| |
Collapse
|
8
|
Dang HT, Tran DM, Phung TTB, Bui ATP, Vu YH, Luong MT, Nguyen HM, Trinh HT, Nguyen TT, Nguyen AH, Van Nguyen AT. Promising clinical and immunological efficacy of Bacillus clausii spore probiotics for supportive treatment of persistent diarrhea in children. Sci Rep 2024; 14:6422. [PMID: 38494525 PMCID: PMC10944834 DOI: 10.1038/s41598-024-56627-9] [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: 08/15/2023] [Accepted: 03/08/2024] [Indexed: 03/19/2024] Open
Abstract
Persistent diarrhea is a severe gastroenteric disease with relatively high risk of pediatric mortality in developing countries. We conducted a randomized, double-blind, controlled clinical trial to evaluate the efficacy of liquid-form Bacillus clausii spore probiotics (LiveSpo CLAUSY; 2 billion CFU/5 mL ampoule) at high dosages of 4-6 ampoules a day in supporting treatment of children with persistent diarrhea. Our findings showed that B. clausii spores significantly improved treatment outcomes, resulting in a 2-day shorter recovery period (p < 0.05) and a 1.5-1.6 folds greater efficacy in reducing diarrhea symptoms, such as high frequency of bowel movement of ≥ 3 stools a day, presence of fecal mucus, and diapered infant stool scale types 4-5B. LiveSpo CLAUSY supportive treatment achieved 3 days (p < 0.0001) faster recovery from diarrhea disease, with 1.6-fold improved treatment efficacy. At day 5 of treatment, a significant decrease in blood levels of pro-inflammatory cytokines TNF-α, IL-17, and IL-23 by 3.24% (p = 0.0409), 29.76% (p = 0.0001), and 10.87% (p = 0.0036), respectively, was observed in the Clausy group. Simultaneously, there was a significant 37.97% decrease (p = 0.0326) in the excreted IgA in stool at day 5 in the Clausy group. Overall, the clinical study demonstrates the efficacy of B. clausii spores (LiveSpo CLAUSY) as an effective symptomatic treatment and immunomodulatory agent for persistent diarrhea in children.Trial registration: NCT05812820.
Collapse
Affiliation(s)
- Ha Thuy Dang
- Department of Gastroenterology, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Dien Minh Tran
- Department of Surgical Intensive Care Unit, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Thuy Thi Bich Phung
- Department of Molecular Biology for Infectious Diseases, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Anh Thi Phuong Bui
- Spobiotic Research Center, ANABIO R&D Ltd. Company, No. 22, Lot 7,8 Van Khe Urban, La Khe, Ha Dong, Hanoi, Vietnam
| | - Yen Hai Vu
- Department of Gastroenterology, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Minh Thi Luong
- Department of Gastroenterology, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Hang Minh Nguyen
- Department of Molecular Biology for Infectious Diseases, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Huong Thi Trinh
- Department of Molecular Biology for Infectious Diseases, Vietnam National Children's Hospital, No. 18/879 La Thanh, Dong Da, Hanoi, Vietnam
| | - Tham Thi Nguyen
- Spobiotic Research Center, ANABIO R&D Ltd. Company, No. 22, Lot 7,8 Van Khe Urban, La Khe, Ha Dong, Hanoi, Vietnam
| | - Anh Hoa Nguyen
- Spobiotic Research Center, ANABIO R&D Ltd. Company, No. 22, Lot 7,8 Van Khe Urban, La Khe, Ha Dong, Hanoi, Vietnam.
- LiveSpo Pharma Ltd. Company, N03T5, Ngoai Giao Doan Urban, Bac Tu Liem, Hanoi, Vietnam.
| | - Anh Thi Van Nguyen
- Spobiotic Research Center, ANABIO R&D Ltd. Company, No. 22, Lot 7,8 Van Khe Urban, La Khe, Ha Dong, Hanoi, Vietnam.
| |
Collapse
|
9
|
Yang T, Li L, Heng C, Sha P, Wang Y, Shen J, Jiang Z, Qian S, Wei C, Yang H, Zhu X, Wang T, Wu M, Wang J, Lu Q, Yin X. Sodium butyrate ameliorated diabetic nephropathy-associated tubulointerstitial inflammation by modulating the tight junctions of renal tubular epithelial cells. Food Funct 2024; 15:2628-2644. [PMID: 38358014 DOI: 10.1039/d2fo00940d] [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: 02/16/2024]
Abstract
As one of the most significant pathological changes of diabetic nephropathy (DN), tubulointerstitial fibrosis (TIF) had a close relationship with tubulointerstitial inflammation (TI), and the occurrence of TI could have resulted from the disrupted tight junctions (TJs) of renal tubular epithelial cells (RTECs). Studies have demonstrated that sodium butyrate (NaB), a typical short chain fatty acid (SCFA), played an important regulatory role in intestinal TJs and inflammation. In this study, our in vivo and in vitro results showed that accompanied by TI, renal tubular TJs were gradually disrupted in the process of DN-related TIF. In HG and LPS co-cultured HK-2 cells and db/db mice, NaB treatment regained the TJs of RTECs via the sphingosine 1-phosphate receptor-1 (S1PR1)/AMPK signaling pathway, relieving inflammation. Small interfering RNA of S1PR1, S1PR1 antagonist W146 and agonist SEW2871, and AMPK agonist AICAR were all used to further confirm the essential role of the S1PR1/AMPK signaling pathway in NaB's TJ protection in RTECs in vitro. Finally, NaB administration not only improved the renal function and TIF, but also relieved the TI of db/db mice. These findings suggested that the use of NaB might be a potential adjuvant treatment strategy for DN-associated TIF, and this protective effect was linked to the TJ modulation of RTECs via the S1PR1/AMPK signaling pathway, leading to the improvement of TI.
Collapse
Affiliation(s)
- Tingting Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Lin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Cai Heng
- Department of Pharmacy, JingJiang People's Hospital, Jingjiang 214500, China
| | - Pian Sha
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Yiying Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Jiaming Shen
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Zhenzhou Jiang
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Sitong Qian
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Chujing Wei
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Yang
- Department of Pharmacy, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou 213000, China
| | - Xia Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Tao Wang
- Department of Pharmacy, The affiliated hospital of Xuzhou Medical University, Xuzhou 221006, China
| | - Mengying Wu
- New drug screening center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Jianyun Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
10
|
Daca A, Jarzembowski T. From the Friend to the Foe- Enterococcus faecalis Diverse Impact on the Human Immune System. Int J Mol Sci 2024; 25:2422. [PMID: 38397099 PMCID: PMC10888668 DOI: 10.3390/ijms25042422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Enterococcus faecalis is a bacterium which accompanies us from the first days of our life. As a commensal it produces vitamins, metabolizes nutrients, and maintains intestinal pH. All of that happens in exchange for a niche to inhabit. It is not surprising then, that the bacterium was and is used as an element of many probiotics and its positive impact on the human immune system and the body in general is hard to ignore. This bacterium has also a dark side though. The plasticity and relative ease with which one acquires virulence traits, and the ability to hide from or even deceive and use the immune system to spread throughout the body make E. faecalis a more and more dangerous opponent. The statistics clearly show its increasing role, especially in the case of nosocomial infections. Here we present the summarization of current knowledge about E. faecalis, especially in the context of its relations with the human immune system.
Collapse
Affiliation(s)
- Agnieszka Daca
- Department of Physiopathology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Tomasz Jarzembowski
- Department of Microbiology, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
11
|
Iyer A, Mukherjee A, Gómez-Sala B, O'Connor EM, Kenny JG, Cotter PD. The impact of live dietary microbes on health: A scoping review. J Food Sci 2024; 89:773-792. [PMID: 38174642 DOI: 10.1111/1750-3841.16893] [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: 08/23/2023] [Revised: 11/16/2023] [Accepted: 12/02/2023] [Indexed: 01/05/2024]
Abstract
A systematic approach to collect, peruse, and summarize the available information relating to the potential benefits of consuming dietary microbes was pursued in this scoping review. This review focused on the research endpoints, experimental designs, and microbial exposure in experimental as well as observational research work. Using a structured- set of keywords, scientific databases were systematically searched to retrieve publications reporting outcomes pertaining to the use of dietary microbes in healthy, nonpatient populations. Searches were further tailored to focus on eight different health categories, namely, "antibiotic associated diarrhoea" (AAD), "gastrointestinal health" (GIH), "immunological health" (ImH), "cardiovascular health and metabolic syndrome" (CvHMS), "cancer prevention" (CanPr), "respiratory health" (ReH), "weight management" (WtMgt), and "urogenital health" (UrGH). Quality of evidence available in each publication was assessed using the Jadad scoring system. The search yielded 228 relevant publications describing 282 experimental cases comprising 62 research endpoints overall. A microbial dose of≥ 2 × 10 9 $\ge 2\times 10^9$ CFU.day-1 was associated with non-negative reported outcomes. Older population groups with a median age of 39 years were associated with positive outcomes. More high-quality research is required investigating the role of dietary microbes in maintaining general health, particularly in the health categories of UrGH, WtMgt, and CanPr.
Collapse
Affiliation(s)
- Ajay Iyer
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Arghya Mukherjee
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Beatriz Gómez-Sala
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Eibhlís M O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - John G Kenny
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk SFI Research Centre, Cork, Ireland
| | - Paul D Cotter
- Department of Food Biosciences, Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- VistaMilk SFI Research Centre, Cork, Ireland
| |
Collapse
|
12
|
Pal AD, Pal A. Probiotics: beneficial microbes for health and the food industry. MICROBIAL ESSENTIALISM 2024:47-86. [DOI: 10.1016/b978-0-443-13932-1.00026-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Mi J, He T, Hu X, Wang Z, Wang T, Qi X, Li K, Gao L, Liu C, Zhang Y, Wang S, Qiu Y, Liu Z, Song J, Wang X, Gao Y, Cui H. Enterococcus faecium C171: Modulating the Immune Response to Acute Lethal Viral Challenge. Int J Antimicrob Agents 2023; 62:106969. [PMID: 37758064 DOI: 10.1016/j.ijantimicag.2023.106969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/08/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Commensal bacteria modulate acute immune responses to infection in hosts. In this study, Enterococcus faecium C171 was screened and isolated. This strain has similar basic characteristics to the reference probiotic, including strong anti-inflammatory and anti-infective effects. E. faecium C171 inhibits the production of pro-Caspase-1 and significantly reduces the production of interleukin-1β (IL-1β) in vitro. These reactions were confirmed using the Transwell system. Live E. faecium C171 mainly exerted an inhibitory effect on acute inflammation, whereas the anti-infective and immune-activating effects were primarily mediated by the E. faecium C171-produced bacterial extracellular vesicles (Efm-C171-BEVs). Furthermore, in the specific pathogen-free (SPF) chicken model, oral administration of E. faecium C171 increased the relative abundance of beneficial microbiota (Enterococcus and Lactobacillus), particularly Enterococcus, the most important functional bacteria of the gut microbiota. E. faecium C171 significantly inhibited the acute inflammatory response induced by a highly virulent infectious disease, and reduced mortality in SPF chickens by 75%. In addition, E. faecium C171 induced high levels of CD3+, CD4-, and CD8- immunoregulatory cells and CD8+ killer T cells, and significantly improved the proliferative activity of T cells in peripheral blood mononuclear cells, and the secretion of interferon-γ. These findings indicate that E. faecium C171 and Efm-C171-BEVs are promising candidates for adjuvant treatment of acute inflammatory diseases and acute viral infections.
Collapse
Affiliation(s)
- Jielan Mi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Tana He
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xinyun Hu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Zhihao Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Tingting Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xiaole Qi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Kai Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Li Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Changjun Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yanping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Suyan Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yu Qiu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Zengqi Liu
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Jie Song
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Xiaomei Wang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Yulong Gao
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| | - Hongyu Cui
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
14
|
Li P, Chen X, Hou D, Chen B, Peng K, Huang W, Cao J, Zhao H. Positive effects of dietary Clostridium butyricum supplementation on growth performance, antioxidant capacity, immunity and viability against hypoxic stress in largemouth bass. Front Immunol 2023; 14:1190592. [PMID: 37711631 PMCID: PMC10498469 DOI: 10.3389/fimmu.2023.1190592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/16/2023] [Indexed: 09/16/2023] Open
Abstract
The effects of dietary supplementation of Clostridium butyricum (CB) on growth performance, serum biochemistry, antioxidant activity, mRNA levels of immune-related genes and resistance to hypoxia stress were studied in largemouth bass. Feed with CB0 (control, 0 CFU/kg), CB1 (4.3×108 CFU/kg), CB2 (7.5×108 CFU/kg), CB3 (1.5×109 CFU/kg) and CB4 (3.2×109 CFU/kg) CB for 56 days, and then a 3 h hypoxic stress experiment was performed. The results showed that dietary CB significantly increased the WGR (weight gain rate), SGR (specific growth rate), PDR (protein deposition rate) and ISI (Intestosomatic index) of largemouth bass (P<0.05). Hepatic GH (growth hormone)/IGF-1 (insulin-like growth factor-1) gene expression was significantly upregulated in the CB3 and CB4 groups compared with the CB0 group (P<0.05), while the FC (feed conversion) was significantly decreased (P<0.05). Serum TP (total protein) and GLU (glucose) levels were significantly higher in the CB4 group than in the CB0 group (P<0.05), while the contents of serum AST (aspartate transaminase), ALT (alanine transaminase), AKP (alkline phosphatase) and UN (urea nitrogen) in CB4 were significantly lower than those in CB0 (P<0.05). T-AOC (total antioxidant capacity), SOD (superoxide dismutase), CAT (catalase), POD (peroxidase) and GSH-Px (glutathione peroxidase) activities were significantly higher in CB3 and CB4 groups than in CB0 group (P<0. 05). The liver MDA (malondialdehyde) content of CB1, CB2, CB3 and CB4 groups was significantly higher than that of CB0 group (P<0. 05). The relative expressions of IL-1β (interleukin 1β), TNF-α (tumor necrosis factor α) and TLR22 (toll-like receptor-22) genes in CB2, CB3 and CB4 groups were significantly lower than those in CB0 group (P<0.05). The relative expression of IL-8 (malondialdehyde) and MyD88 (Myeloid differentiation factor 88) genes in the CB4 group was significantly lower than that in the CB0 group (P<0.05). The liver LZM (lysozyme) content of CB2, CB3 and CB4 groups was significantly higher than that of CB0 group (P<0. 05). The relative expression of IL-10 (interleukin 10) and TGF-β (transforming growth factor β) genes in the CB4 group was significantly higher than that in the CB0 group (P<0.05). Under hypoxic stress for 3 h, the CMR of CB0 group was significantly higher than that of CB1, CB2, CB3 and CB4 groups (P<0.05). Dietary CB can improve the growth performance and resistance to hypoxic stress of largemouth bass by regulating the expression of GH/IGF-1 gene and inflammatory factors and inhibiting TLR22/MyD88 signaling pathway.
Collapse
Affiliation(s)
- Peijia Li
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Xiaoying Chen
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Dongqiang Hou
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
- College of Fisheries, Guangdong Ocean University, Zhanjiang, China
| | - Bing Chen
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Kai Peng
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Wen Huang
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Junming Cao
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | - Hongxia Zhao
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Collaborative Innovation Center of Aquatic Sciences, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| |
Collapse
|
15
|
Im EJ, Lee HHY, Kim M, Kim MK. Evaluation of Enterococcal Probiotic Usage and Review of Potential Health Benefits, Safety, and Risk of Antibiotic-Resistant Strain Emergence. Antibiotics (Basel) 2023; 12:1327. [PMID: 37627747 PMCID: PMC10451534 DOI: 10.3390/antibiotics12081327] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/11/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Enterococci are often used in probiotics but can also cause nosocomial infections. As such, enterococcal consumption may have beneficial health effects, but a thorough evaluation of virulence absence and risk of antibiotic resistance spread is needed at the strain level. This article reviewed ten online health product shopping websites in the US. On these websites, 23 probiotic products using enterococci were found across 12 companies. In addition, this article reviewed studies that demonstrated the probiotic potential of enterococcal consumption (e.g., gastrointestinal and respiratory disease, hyperlipidemia alleviation, as well as infection prevention). To investigate the safety aspects of enterococci, the present work examined studies evaluating virulence factors and antibiotic resistance. Furthermore, this article assessed research that explored these virulent factors, specifically in probiotics containing enterococci, as well as the potential transfer mechanism of their antibiotic resistance. Based on reviewed data, enterococcal probiotic consumption has been proven beneficial for conditions or symptoms of multiple diseases without any apparent adverse effects. However, due to the plasmid- or transposon-mediated gene transfer ability of enterococci, surveillance monitoring and further studies regarding enterococcal consumption are warranted. Future studies that identify enterococcal strains safe to use in probiotics without virulence factors and antibiotic resistance are imperative for evidence-based decisions by health organizations and government agencies.
Collapse
Affiliation(s)
- Eric Jeeho Im
- College of Arts and Sciences, Washington University, St. Louis, MO 63130, USA;
| | - Harry Hyun-Yup Lee
- School of Osteopathic Medicine, Campbell University, Lillington, NC 27546, USA
| | - Minzae Kim
- College of Arts and Sciences, Boston University, Boston, MA 02215, USA
| | - Myo-Kyoung Kim
- Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA 95211, USA
| |
Collapse
|
16
|
Gou HZ, Zhang YL, Ren LF, Li ZJ, Zhang L. How do intestinal probiotics restore the intestinal barrier? Front Microbiol 2022; 13:929346. [PMID: 35910620 PMCID: PMC9330398 DOI: 10.3389/fmicb.2022.929346] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
The intestinal barrier is a structure that prevents harmful substances, such as bacteria and endotoxins, from penetrating the intestinal wall and entering human tissues, organs, and microcirculation. It can separate colonizing microbes from systemic tissues and prevent the invasion of pathogenic bacteria. Pathological conditions such as shock, trauma, stress, and inflammation damage the intestinal barrier to varying degrees, aggravating the primary disease. Intestinal probiotics are a type of active microorganisms beneficial to the health of the host and an essential element of human health. Reportedly, intestinal probiotics can affect the renewal of intestinal epithelial cells, and also make cell connections closer, increase the production of tight junction proteins and mucins, promote the development of the immune system, regulate the release of intestinal antimicrobial peptides, compete with pathogenic bacteria for nutrients and living space, and interact with the host and intestinal commensal flora to restore the intestinal barrier. In this review, we provide a comprehensive overview of how intestinal probiotics restore the intestinal barrier to provide new ideas for treating intestinal injury-related diseases.
Collapse
Affiliation(s)
- Hong-Zhong Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yu-Lin Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Long-Fei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhen-Jiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
17
|
Shridhar PB, Amachawadi RG, Tokach M, Patel I, Gangiredla J, Mammel M, Nagaraja TG. Whole genome sequence analyses-based assessment of virulence potential and antimicrobial susceptibilities and resistance of Enterococcus faecium strains isolated from commercial swine and cattle probiotic products. J Anim Sci 2022; 100:6527694. [PMID: 35150575 PMCID: PMC8908542 DOI: 10.1093/jas/skac030] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
Enterococcus faecium is one of the more commonly used bacterial species as a probiotic in animals. The organism, a common inhabitant of the gut of animals and humans, is a major nosocomial pathogen responsible for a variety infections in humans and sporadic infections in animals. In swine and cattle, E. faecium-based probiotic products are used for growth promotion and gut functional and health benefits. The objective of this study was to utilize whole genome sequence-based analysis to assess virulence potential, detect antimicrobial resistance genes, and analyze phylogenetic relationships of E. faecium strains from commercial swine and cattle probiotics. Genomic DNA extracted from E. faecium strains, isolated from commercial probiotic products of swine (n = 9) and cattle (n = 13), were sequenced in an Illumina MiSeq platform and analyzed. Seven of the nine swine strains and seven of the 13 cattle strains were identified as Enterococcus lactis, and not as E. faecium. None of the 22 probiotic strains carried major virulence genes required to initiate infections, but many carried genes involved in adhesion to host cells, which may benefit the probiotic strains to colonize and persist in the gut. Strains also carried genes encoding resistance to a few medically important antibiotics, which included aminoglycosides [aac(6')-Ii, aph(3')-III, ant(6)-Ia], macrolide, lincosamide and streptogramin B (msrC), tetracyclines [tet(L) and tet(M)], and phenicols [cat-(pc194)]. The comparison of the genotypic to phentypic AMR data showed presence of both related and unrelated genes in the probiotic strains. Swine and cattle probiotic E. faecium strains belonged to diverse sequence types. Phylogenetic analysis of the probiotic strains, and strains of human (n = 29), swine (n = 4), and cattle (n = 4) origin, downloaded from GenBank, indicated close clustering of strains belonging to the same species and source, but a few swine and cattle probiotic strains clustered closely with other cattle and human fecal strains. In conclusion, the absence of major virulence genes characteristic of the clinical E. faecium strains suggests that these probiotic strains are unlikely to initiate opportunistic infection. However, the carriage of AMR genes to medically important antibiotics and close clustering of the probiotic strains with other human and cattle fecal strains suggests that probiotic strains may pose risk to serve as a source of transmitting AMR genes to other gut bacteria.
Collapse
Affiliation(s)
- Pragathi B Shridhar
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506-5800, USA
| | - Raghavendra G Amachawadi
- Department of Clinical Sciences, Kansas State University, Manhattan, KS 66506-5800, USA,Corresponding author:
| | - Mike Tokach
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS 66506-5800, USA
| | - Isha Patel
- Division of Molecular Biology, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD 20708, USA
| | - Jayanthi Gangiredla
- Division of Molecular Biology, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD 20708, USA
| | - Mark Mammel
- Division of Molecular Biology, Center for Food Safety and Applied Nutrition, United States Food and Drug Administration, Laurel, MD 20708, USA
| | - T G Nagaraja
- Department of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, KS 66506-5800, USA
| |
Collapse
|
18
|
Zhou A, Yuan Y, Yang M, Huang Y, Li X, Li S, Yang S, Tang B. Crosstalk Between the Gut Microbiota and Epithelial Cells Under Physiological and Infectious Conditions. Front Cell Infect Microbiol 2022; 12:832672. [PMID: 35155283 PMCID: PMC8829037 DOI: 10.3389/fcimb.2022.832672] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/10/2022] [Indexed: 12/15/2022] Open
Abstract
The gastrointestinal tract (GIT) is considered the largest immunological organ, with a diverse gut microbiota, that contributes to combatting pathogens and maintaining human health. Under physiological conditions, the crosstalk between gut microbiota and intestinal epithelial cells (IECs) plays a crucial role in GIT homeostasis. Gut microbiota and derived metabolites can compromise gut barrier integrity by activating some signaling pathways in IECs. Conversely, IECs can separate the gut microbiota from the host immune cells to avoid an excessive immune response and regulate the composition of the gut microbiota by providing an alternative energy source and releasing some molecules, such as hormones and mucus. Infections by various pathogens, such as bacteria, viruses, and parasites, can disturb the diversity of the gut microbiota and influence the structure and metabolism of IECs. However, the interaction between gut microbiota and IECs during infection is still not clear. In this review, we will focus on the existing evidence to elucidate the crosstalk between gut microbiota and IECs during infection and discuss some potential therapeutic methods, including probiotics, fecal microbiota transplantation (FMT), and dietary fiber. Understanding the role of crosstalk during infection may help us to establish novel strategies for prevention and treatment in patients with infectious diseases, such as C. difficile infection, HIV, and COVID-19.
Collapse
Affiliation(s)
- An Zhou
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Yuan
- Institution of Basic Medicine, Third Military Medical University, Chongqing, China
| | - Min Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yujiao Huang
- The First Clinical College, ChongQing Medical University, Chongqing, China
| | - Xin Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shengpeng Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- *Correspondence: Shiming Yang, ; Bo Tang,
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- *Correspondence: Shiming Yang, ; Bo Tang,
| |
Collapse
|
19
|
YONESHIGE R, KUSUDA E, ANDO T. Effect of probiotics on blood fatty acid metabolism during the late middle stage of fattening period in Japanese Black cattle. J Vet Med Sci 2022; 84:319-324. [PMID: 35110457 PMCID: PMC8983281 DOI: 10.1292/jvms.21-0354] [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] [Indexed: 11/22/2022] Open
Abstract
This study was conducted to investigate the effects of probiotics administration on fatty acid metabolism in Japanese Black cattle as per changes in blood fatty acid concentrations and
blood biochemical tests. Eighteen clinically healthy Japanese Black female fattening cattle bred on the same fattening farm were randomly classified into the probiotics administration group
(n=9) or the control group (n=9). In the probiotics administration group, 50 g of probiotics were started per animal per day at the age of 18 months, and the administration period was 2
months from the start date of the study. Blood was collected twice before starting the probiotics administration and at 2 months after starting the probiotics administration. In the
probiotics administration group, palmitic, linoleic, arachidonic and α-linolenic acid tended to be higher at the end of the administration compared with those before probiotics
administration. Additionally, as a result of multiple comparison test, monounsaturated fatty acids at Post was significantly higher, and the ω6 / ω3 ratio was significantly lower than in the
control group. Vitamin A, E and albumin were significantly higher at the end of the administration than in the control group. In this study that administering probiotics to Japanese Black
cattle in the late middle stage of fattening period did not have a significant effect on fatty acid metabolism during feed digestion and absorption, but suggested that may alter some blood
fatty acids concentrations.
Collapse
Affiliation(s)
| | | | - Takaaki ANDO
- The United Graduate School of Veterinary Science, Yamaguchi University
| |
Collapse
|
20
|
The role of enteric dysbacteriosis and modulation of gut microbiota in the treatment of inflammatory bowel disease. Microb Pathog 2021; 165:105381. [PMID: 34974123 DOI: 10.1016/j.micpath.2021.105381] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022]
Abstract
The incidence of inflammatory bowel disease (IBD) is globally increasing. This disorder seriously affects the quality of life in patients. Interestingly, studies have detected that the intestinal flora imbalance is a critical factor in the progression of IBD. One potential treatment strategy for IBD involves regulating the composition and function of the intestinal flora. To date, a multitude of experiments have confirmed the relationship between intestinal flora, immune regulation, and anti-inflammation. The intestinal flora can reduce intestinal inflammation by regulating immunity and increasing the secretion of metabolic short-chain fatty acids. In this review, we discuss the composition and function of the intestinal flora, the relationship between the intestinal flora and the host, the role of intestinal flora disorders in IBD, and the progress in IBD treatment. Combining the regulation of the intestinal flora with probiotics treatment is considered a promising strategy for substantially improving the treatment of IBD.
Collapse
|
21
|
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; 13:4319. [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] [Grants] [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
| |
Collapse
|
22
|
Effectiveness of probiotics and synbiotics in reducing duration of acute infectious diarrhea in pediatric patients in developed countries: a systematic review and meta-analysis. Eur J Pediatr 2021; 180:2907-2920. [PMID: 33825068 DOI: 10.1007/s00431-021-04046-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 03/11/2021] [Accepted: 03/21/2021] [Indexed: 02/07/2023]
Abstract
Acute diarrhea is one of the most frequent causes of doctor visits and hospital admissions for children. Our objective was to evaluate the association between probiotics administration and reduction of acute infectious diarrhea duration in children dwelling in developed countries. Bibliographic databases, gray literature, and reference lists were searched up to September 29, 2019. Double-blind, randomized controlled trials that examined probiotics efficacy in children with acute infectious diarrhea residing in developed countries were included. Data were synthesized by generic inverse variance method using fixed- and random-effects model. Twenty trials met the eligibility criteria (n = 3469 patients) and were included in the qualitative synthesis, and 19 studies in meta-analysis. Twelve trials (n = 840) were assessed as high/unclear risk of bias and eight (n = 2629) as low risk of bias. Comparisons revealed a moderate effectiveness of probiotics in low risk of bias studies (MD = - 13.45 h; 95% CI - 24.26, - 2.62; p = 0.02, Bayesian meta-analysis pooled effect MD = - 0.38, 95% CrI - 2.3, 1.58) and a notable effect in studies with high/unclear risk for bias (MD = - 19.70 h; 95% CI - 28.09, - 11.31; p = 0.0004). In trials of optimal methodological quality (n = 1989), probiotics effect was absent (MD = - 3.32 h; 95% CI - 8.78, 2.13, p = 0.23).Conclusion: Outcomes suggest that probiotics do not demonstrate sufficient clinical impact in reducing diarrhea duration in children in the developed countries.Systematic Review Registration: This review is registered at PROSPERO (ID: CRD42020152966). What is Known: • Probiotics, due to the conflicting study results, are administered without adequate evidence as an adjuvant therapeutic agent for eliminating duration of acute infectious diarrhea in pediatric patients. What is New: • In developed countries, probiotics are demonstrated as ineffective in reducing the duration of acute infectious diarrhea in children.
Collapse
|
23
|
Li H, Liu X, Shang Z, Qiao J. Clostridium butyricum Helps to Alleviate Inflammation in Weaned Piglets Challenged With Enterotoxigenic Escherichia coli K88. Front Vet Sci 2021; 8:683863. [PMID: 34277756 PMCID: PMC8282889 DOI: 10.3389/fvets.2021.683863] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/11/2021] [Indexed: 12/05/2022] Open
Abstract
Background: Whether the probiotic Clostridium butyricum (CB) alleviates enterotoxigenic Escherichia coli (ETEC) K88-induced inflammation by regulating the activation of the toll-like receptor (TLR) signaling pathway is not clear, thus, we carried out this study. A total of 72 piglets (average body weight 7.09 ± 0.2 kg) were randomly divided into three groups of 24 piglets per group. Pigs were either fed a daily diet (NC, negative control), a diet tested every day by 1 × 109 CFU/mL ETEC K88 (PC, positive control), or a basal diet supplemented with 5 × 105 CFU/g CB and challenged with ETEC K88 (PC + CB group). Results: Our results showed that CB pretreatment attenuated the effect of ETEC K88 by decreasing C-reactive protein (CRP), which resulted in tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) production. Histological examination revealed that CB pretreatment alleviated intestinal villi injury caused by ETEC K88 challenge. Furthermore, CB pretreatment promoted mRNA expression of the negative regulators of TLR signaling, including myeloid differentiation factor (MyD88), toll-interacting protein (Tollip), and B cell CLL/lymphoma 3 (Bcl-3), in the intestines of ETEC K88-challenged piglets. ETEC K88-induced activation of nuclear factor kappa B (NF-κB) and nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor alpha (IκBα) was attenuated by CB pretreatment. Conclusion: These findings indicate that CB helps to maintain and strengthen the shape of intestinal villi and limits detrimental inflammatory responses, partly by inhibiting toll-like receptor 2 (TLR-2), toll-like receptor 4 (TLR-4), and toll-like receptor 5 (TLR-5) expression and inhibiting NF-κB p65, and promoting IκBα activation and synergism among its negative regulators.
Collapse
Affiliation(s)
- Haihua Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Xuejiao Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Zhiyuan Shang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Jiayun Qiao
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| |
Collapse
|
24
|
Chelliah R, Kim EJ, Daliri EBM, Antony U, Oh DH. In Vitro Probitotic Evaluation of Saccharomyces boulardii with Antimicrobial Spectrum in a Caenorhabditis elegans Model. Foods 2021; 10:foods10061428. [PMID: 34203095 PMCID: PMC8235530 DOI: 10.3390/foods10061428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 11/16/2022] Open
Abstract
In the present study, we screened for potential probiotic yeast that could survive under extreme frozen conditions. The antimicrobial and heat-stable properties of the isolated yeast strains Saccharomyces boulardii (S. boulardii) (KT000032, KT000033, KT000034, KT000035, KT000036, and KT000037) was analyzed and compared with commercial probiotic strains. The results revealed that the tested S. boulardii KT000032 strain showed higher resistance to gastric enzymes (bile salts, pepsin, and pancreatic enzyme) at low pH, with broad antibiotic resistance. In addition, the strain also showed efficient auto-aggregation and co-aggregation abilities and efficient hydrophobicity in the in-vitro and in-vivo C. elegens gut model. Further, the KT000032 strain showed higher antimicrobial efficiency against 13 different enteropathogens and exhibited commensal relationships with five commercial probiotic strains. Besides, the bioactive compounds produced in the cell-free supernatant of probiotic yeast showed thermo-tolerance (95 °C for two hours). Furthermore, the thermo-stable property of the strains will facilitate their incorporation into ready-to-eat food products under extreme food processing conditions.
Collapse
Affiliation(s)
- Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon 24341, Korea; (R.C.); (E.-J.K.); (E.B.-M.D.)
| | - Eun-Ji Kim
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon 24341, Korea; (R.C.); (E.-J.K.); (E.B.-M.D.)
| | - Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon 24341, Korea; (R.C.); (E.-J.K.); (E.B.-M.D.)
| | - Usha Antony
- Department of Biotechnology and Food Technology, Anna University, Chennai 600 025, India;
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Science, Kangwon National University, Chuncheon 24341, Korea; (R.C.); (E.-J.K.); (E.B.-M.D.)
- Correspondence: ; Tel.: +82-33-250-6457
| |
Collapse
|
25
|
Azad A, Ranjbaran A, Zareshahrabadi Z, Mehrabani D, Zahed Zahedani M, Talebanpour A, Zomorodian K. Protective Effects of the Probiotic Bacterium Streptococcus thermophilus on Candida albicans Morphogenesis and a Murine Model of Oral Candidiasis. IRANIAN JOURNAL OF MEDICAL SCIENCES 2021; 46:207-217. [PMID: 34083853 PMCID: PMC8163705 DOI: 10.30476/ijms.2020.82080.0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 09/30/2019] [Accepted: 10/20/2019] [Indexed: 11/19/2022]
Abstract
Background Oral candidiasis is a frequent form of candidiasis, caused by Candida species, in particular, Candida albicans (C. albicans). The transition of C. albicans from yeast to hyphae allows its attachment to epithelial cells, followed by biofilm formation, invasion, and tissue damage. Hence, we investigated the effect of Streptococcus salivarius subspecies thermophilus (S thermophilus) on the growth as well as biofilm and germ-tube formation of C. albicans both in vitro and in vivo in a murine model. Methods This experimental study was performed in the Department of Medical Mycology and Parasitology, School of Medicine, in collaboration with the Central Research Laboratory and the Comparative Biomedical Center, Shiraz University of Medical Sciences, Shiraz, Iran (2017 to 2018). The inhibitory activity of S. thermophilus against Candida species growth was evaluated using the broth microdilution method, and the inhibition of C. albicans biofilm formation was measured using the XTT assay. The inhibition of C. albicans germ-tube formation by S. thermophilus was evaluated using the plate assay and fluorescence microscopy. The experimental activity of the probiotic bacterium was assessed by culture and histopathological methods in six groups of five mice, comprising those treated with four concentrations of probiotics, fluconazole, and distilled water. The one-way analysis of variance, followed by a Tukey post hoc test, was used and a P value of less than 0.05 was considered significant. Results S. thermophilus inhibited Candida species growth at concentrations of 16 to 512 µg/mL. This probiotic inhibited the formation of C. albicans biofilms and germ tubes in a dose-dependent manner. S. thermophilus significantly reduced the colony-forming units in the mice receiving 30 mg/mL of this probiotic treatment compared with the control group (P=0.024). The histopathological analysis showed that Candida colonization was diminished in the mice following the administration of the probiotic. Conclusion Given the inhibitory activity of S. thermophilus against the growth, transition, and biofilm formation of C. albicans, it could be used in the management of oral candidiasis.
Collapse
Affiliation(s)
- Azita Azad
- Oral and Dental Disease Research Center, Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Ranjbaran
- Oral and Dental Disease Research Center, Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Zareshahrabadi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davood Mehrabani
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Zahed Zahedani
- Oral and Dental Disease Research Center, Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asana Talebanpour
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kamiar Zomorodian
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Basic Sciences in Infectious Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
Freedman SB, Horne R, Johnson-Henry K, Xie J, Williamson-Urquhart S, Chui L, Pang XL, Lee B, Schuh S, Finkelstein Y, Gouin S, Farion KJ, Poonai N, Hurley K, Schnadower D, Sherman PM, Pediatric Emergency Research Canada Probiotic Regimen for Outpatient Gastroenteritis Utility of Treatment (PROGUT) Trial Group. Probiotic stool secretory immunoglobulin A modulation in children with gastroenteritis: a randomized clinical trial. Am J Clin Nutr 2021; 113:905-914. [PMID: 34269370 PMCID: PMC8023833 DOI: 10.1093/ajcn/nqaa369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND We previously conducted the Probiotic Regimen for Outpatient Gastroenteritis Utility of Treatment (PROGUT) study, which identified no improvements in children with acute gastroenteritis (AGE) administered a probiotic. However, the aforementioned study did not evaluate immunomodulatory benefits. OBJECTIVES The object of this study was to determine if stool secretory immunoglobulin A (sIgA) concentrations in children with AGE increase more among participants administered a Lactobacillus rhamnosus/helveticus probiotic compared with those administered placebo. METHODS This a priori planned multicenter, randomized, double-blinded, placebo-controlled ancillary study enrolled children presenting for emergency care who received a 5-d probiotic or placebo course. Participants submitted stool specimens on days 0, 5, and 28. The primary endpoint was the change in stool sIgA concentrations on day 5 compared with baseline. RESULTS A total of 133 (n = 66 probiotic, 67 placebo) of 886 PROGUT participants (15.0%) provided all 3 specimens. Median stool sIgA concentrations did not differ between the probiotic and placebo groups at any of the study time points: day 0 median (IQR): 1999 (768, 4071) compared with 2198 (702, 5278) (P = 0.27, Cohen's d = 0.17); day 5: 2505 (1111, 5310) compared with 3207 (982, 7080) (P = 0.19, Cohen's d = 0.16); and day 28: 1377 (697, 2248) compared with 1779 (660, 3977) (P = 0.27, Cohen's d = 0.19), respectively. When comparing measured sIgA concentrations between days 0 and 5, we found no treatment allocation effects [β: -0.24 (-0.65, 0.18); P = 0.26] or interaction between treatment and specimen collection day [β: -0.003 (-0.09, 0.09); P = 0.95]. Although stool sIgA decreased between day 5 and day 28 within both groups (P < 0.001), there were no differences between the probiotic and placebo groups in the median changes in sIgA concentrations when comparing day 0 to day 5 median (IQR) [500 (-1135, 2362) compared with 362 (-1122, 4256); P = 0.77, Cohen's d = 0.075] and day 5 to day 28 [-1035 (-3130, 499) compared with -1260 (-4437, 843); P = 0.70, Cohen's d = 0.067], respectively. CONCLUSIONS We found no effect of an L. rhamnosus/helveticus probiotic, relative to placebo, on stool IgA concentrations. This trial was registered at clinicaltrials.gov as NCT01853124.
Collapse
Affiliation(s)
- Stephen B Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Department of Pediatrics, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
- Department of Emergency Medicine, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Rachael Horne
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Kathene Johnson-Henry
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Jianling Xie
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Sarah Williamson-Urquhart
- Section of Pediatric Emergency Medicine, Department of Pediatrics, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Linda Chui
- Alberta Precision Laboratories - ProvLab, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Xiao-Li Pang
- Alberta Precision Laboratories - ProvLab, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Bonita Lee
- Department of Pediatrics, Faculty of Medicine & Dentistry, Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
| | - Suzanne Schuh
- Division of Emergency Medicine, Department of Paediatrics, Hospital for Sick Children, Toronto, Canada
| | - Yaron Finkelstein
- Divisions of Emergency Medicine and Clinical Pharmacology and Toxicology, Research Institute, Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Serge Gouin
- Departments of Pediatric Emergency Medicine & Pediatrics, CHU Sainte-Justine, Université de Montréal, Montreal, Canada
| | - Ken J Farion
- Departments of Pediatrics and Emergency Medicine, and Pediatric Emergency Department, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, Canada
| | - Naveen Poonai
- Division of Pediatric Emergency Medicine, Departments of Pediatrics, Internal Medicine, Epidemiology & Biostatistics, Schulich School of Medicine and Dentistry, London, Canada
| | - Katrina Hurley
- Division of Paediatric Emergency Medicine, Dalhousie University, Halifax, Canada
| | - David Schnadower
- Division of Emergency Medicine, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Philip M Sherman
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Paediatrics, and the Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | | |
Collapse
|
27
|
Shin SY, Hussain Z, Lee YJ, Park H. An altered composition of fecal microbiota, organic acids, and the effect of probiotics in the guinea pig model of postoperative ileus. Neurogastroenterol Motil 2021; 33:e13966. [PMID: 32815235 DOI: 10.1111/nmo.13966] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND The aim of this study is to investigate the altered composition of fecal microbiota, organic acids, and the effect of probiotics in the guinea pig model of the postoperative ileus (POI). METHODS A laparotomy with cecal manipulation was performed to induce POI in guinea pigs. Fecal pellets were collected before the operation (the baseline) and 1, 3, and 5 days after the operation. The extracted fecal DNA was amplified and sequenced using the Illumina MiSeq sequencing system. The same POI procedures were performed after oral pretreatment of the probiotics for 7 days before operation. The effect of the probiotics on the selected taxa and fecal acetate were evaluated, as were the butyrate levels. The colonic transit was assessed by measurement of the fecal pellet output. KEY RESULTS The communities of the baseline and POI groups indicated significantly distinct composition. The genera Bifidobacterium and Lactobacillus were more abundant in the baseline group compared with the POI groups, and Bacteroides and Blautia were more abundant in the POI groups. Decreased abundances of the species Bifidobacterium bifidum and Bifidobacterium longum after the POI procedure were significantly increased in the probiotics group. The decreased fecal butyrate level after the POI procedure was significantly increased, and colonic transit was significantly improved in the probiotics group. CONCLUSIONS AND INFERENCES POI induces gut bacterial dysbiosis. Moreover, pretreatment of probiotics before operation restores the beneficial bacterial species, butyrate production, and bowel movement. The modulation of gut microbiota may help the treatment and prevention of POI.
Collapse
Affiliation(s)
- Seung Yong Shin
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Zahid Hussain
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Ju Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
28
|
Collinson S, Deans A, Padua-Zamora A, Gregorio GV, Li C, Dans LF, Allen SJ. Probiotics for treating acute infectious diarrhoea. Cochrane Database Syst Rev 2020; 12:CD003048. [PMID: 33295643 PMCID: PMC8166250 DOI: 10.1002/14651858.cd003048.pub4] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Probiotics may be effective in reducing the duration of acute infectious diarrhoea. OBJECTIVES To assess the effects of probiotics in proven or presumed acute infectious diarrhoea. SEARCH METHODS We searched the trials register of the Cochrane Infectious Diseases Group, MEDLINE, and Embase from inception to 17 December 2019, as well as the Cochrane Controlled Trials Register (Issue 12, 2019), in the Cochrane Library, and reference lists from studies and reviews. We included additional studies identified during external review. SELECTION CRITERIA Randomized controlled trials comparing a specified probiotic agent with a placebo or no probiotic in people with acute diarrhoea that is proven or presumed to be caused by an infectious agent. DATA COLLECTION AND ANALYSIS Two review authors independently applied inclusion criteria, assessed risk of bias, and extracted data. Primary outcomes were measures of diarrhoea duration (diarrhoea lasting ≥ 48 hours; duration of diarrhoea). Secondary outcomes were number of people hospitalized in community studies, duration of hospitalization in inpatient studies, diarrhoea lasting ≥ 14 days, and adverse events. MAIN RESULTS We included 82 studies with a total of 12,127 participants. These studies included 11,526 children (age < 18 years) and 412 adults (three studies recruited 189 adults and children but did not specify numbers in each age group). No cluster-randomized trials were included. Studies varied in the definitions used for "acute diarrhoea" and "end of the diarrhoeal illness" and in the probiotic(s) tested. A total of 53 trials were undertaken in countries where both child and adult mortality was low or very low, and 26 where either child or adult mortality was high. Risk of bias was high or unclear in many studies, and there was marked statistical heterogeneity when findings for the primary outcomes were pooled in meta-analysis. Effect size was similar in the sensitivity analysis and marked heterogeneity persisted. Publication bias was demonstrated from funnel plots for the main outcomes. In our main analysis of the primary outcomes in studies at low risk for all indices of risk of bias, no difference was detected between probiotic and control groups for the risk of diarrhoea lasting ≥ 48 hours (risk ratio (RR) 1.00, 95% confidence interval (CI) 0.91 to 1.09; 2 trials, 1770 participants; moderate-certainty evidence); or for duration of diarrhoea (mean difference (MD) 8.64 hours shorter, 95% CI 29.4 hours shorter to 12.1 hours longer; 6 trials, 3058 participants; very low-certainty evidence). Effect size was similar and marked heterogeneity persisted in pre-specified subgroup analyses of the primary outcomes that included all studies. These included analyses limited to the probiotics Lactobacillus rhamnosus GG and Saccharomyces boulardii. In six trials (433 participants) of Lactobacillus reuteri, there was consistency amongst findings (I² = 0%), but risk of bias was present in all included studies. Heterogeneity also was not explained by types of participants (age, nutritional/socioeconomic status captured by mortality stratum, region of the world where studies were undertaken), diarrhoea in children caused by rotavirus, exposure to antibiotics, and the few studies of children who were also treated with zinc. In addition, there were no clear differences in effect size for the primary outcomes in post hoc analyses according to decade of publication of studies and whether or not trials had been registered. For other outcomes, the duration of hospitalization in inpatient studies on average was shorter in probiotic groups than in control groups but there was marked heterogeneity between studies (I² = 96%; MD -18.03 hours, 95% CI -27.28 to -8.78, random-effects model: 24 trials, 4056 participants). No differences were detected between probiotic and control groups in the number of people with diarrhoea lasting ≥ 14 days (RR 0.49, 95% CI 0.16 to 1.53; 9 studies, 2928 participants) or in risk of hospitalization in community studies (RR 1.26, 95% CI 0.84 to 1.89; 6 studies, 2283 participants). No serious adverse events were attributed to probiotics. AUTHORS' CONCLUSIONS Probiotics probably make little or no difference to the number of people who have diarrhoea lasting 48 hours or longer, and we are uncertain whether probiotics reduce the duration of diarrhoea. This analysis is based on large trials with low risk of bias.
Collapse
Affiliation(s)
- Shelui Collinson
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Andrew Deans
- Urgent Care, Team Medical, Paraparaumu, New Zealand
| | - April Padua-Zamora
- Department of Pediatrics, University of the Philippines Manila College of Medicine-Philippine General Hospital, Manila, Philippines
| | - Germana V Gregorio
- Department of Pediatrics, University of the Philippines Manila College of Medicine-Philippine General Hospital, Manila, Philippines
| | - Chao Li
- Tropical Clinical Trials Unit, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Leonila F Dans
- Department of Pediatrics, University of the Philippines Manila College of Medicine-Philippine General Hospital, Manila, Philippines
| | - Stephen J Allen
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
29
|
Enhancement of the Anti-inflammatory Effect of Bromelain by Its Immobilization on Probiotic Spore of Bacillus cereus. Probiotics Antimicrob Proteins 2020; 13:847-861. [PMID: 33156496 DOI: 10.1007/s12602-020-09714-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 12/22/2022]
Abstract
The therapeutic application of bromelain is limited due to its sensitivity to operating conditions such as high acidity, gastric proteases in the stomach juice, chemicals, organic solvents and elevated temperature. We hypothesized that bromelain immobilized on probiotic bacterial spores would show enhanced therapeutic activity through possible synergistic or additive effects. In this study, the oedema inhibition potential of bromelain immobilized on probiotic Bacillus spores was compared to the free enzyme using the carrageenan paw oedema model with Wistar rats. In batch A rats (carrageenan-induced inflammation 30 min after receiving oral treatments), group 7 rats treated with a lower dose of spore-immobilized bromelain suspension showed the highest oedema inhibition, 89.20 ± 15.30%, while group 4 treated with a lower dose of free bromelain had oedema inhibition of 60.25 ± 13.00%. For batch B rats (carrageenan-induced inflammation after receiving oral treatment for three days), group 7 rats treated with a lower dose of spore-immobilized bromelain suspension showed higher inhibition percentage (81.94 ± 8.86) than group 4 treated with a lower dose of free bromelain (78.45 ± 4.46) after 24 h. Our results showed that used alone, the enzyme and the spores produced oedema inhibition and improved the motility of the rats. The spore-immobilized bromelain formulation performed approximately 0.9-fold better than the free bromelain and the free spores at the lower evaluated dose.
Collapse
|
30
|
Ugwuodo CJ, Nwagu TN. Stabilizing enzymes by immobilization on bacterial spores: A review of literature. Int J Biol Macromol 2020; 166:238-250. [PMID: 33115650 DOI: 10.1016/j.ijbiomac.2020.10.171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/16/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
The ever-increasing applications of enzymes are limited by the relatively poor performance in harsh processing conditions. As a result, there are constant innovations in immobilization protocols for improving biocatalyst activity and stability. Bacterial spores are cheap to generate and highly resistant to environmental stress. The spore core is sheathed by an inner membrane, the germ cell wall, the cortex, outer membrane, spore coat and in some species the exosporium. The spore surface is anion-rich, hydrophobic and contains several reactive groups capable of interacting and stabilizing enzyme molecules through electrostatic forces, hydrophobic interactions and covalent bonding. The probiotic nature of spores obtained from non-toxic bacterial species makes them suitable carriers for the enzyme immobilization, especially food-grade enzymes or those intended for therapeutic use. Immobilization on spores is by direct adsorption, covalent attachment or surface display during the sporulation phase. Hindrances to the immobilization on spore matrix include the production rates, operational instability, and reduced catalytic properties due to conformational changes in enzyme. This paper reviews bacterial spore as a heterofunctional support matrix gives reasons why probiotic bacillus spores are better options and the diverse technologies adopted for spore-enzyme immobilization. It further suggests directions for future use and discusses the commercialization prospects.
Collapse
|
31
|
Abstract
Since the publication of the 2014 European Society for Paediatric Gastroenterology, Hepatology, and Nutrition Working Group (WG) on Probiotics and Prebiotics guidelines for the management of acute gastroenteritis (AGE), new evidence concerning the efficacy of probiotics has become available. This document provides updated recommendations on the use of probiotics for the treatment of AGE in previously presumed healthy infants and children. A systematic literature search was performed. All pooled analyses were explicitly performed for the current report. The WG graded the recommendations and assessed the certainty of the supporting evidence using the Grading of Recommendations, Assessment Development, and Evaluations tool. The recommendations were formulated if at least 2 randomized controlled trials that used a given probiotic were available. Despite the large number of identified trials, the WG could not identify 2 randomized controlled trial of high quality for any strain that provided benefit when used for treating AGE. The WG made weak recommendations for (in descending order in terms of the number of trials evaluating any given strain): Saccharomyces boulardii (low to very low certainty of evidence); Lactobacillus rhamnosus GG (very low certainty of evidence); L reuteri DSM 17938 (low to very low certainty of evidence); and L rhamnosus 19070-2 and L reuteri DSM 12246 (very low certainty of evidence). The WG made a strong recommendation against L helveticus R0052 and L rhamnosus R0011 (moderate certainty of evidence) and a weak recommendation against Bacillus clausii strains O/C, SIN, N/R, and T (very low certainty of evidence).
Collapse
|
32
|
Preidis GA, Weizman AV, Kashyap PC, Morgan RL. AGA Technical Review on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020; 159:708-738.e4. [PMID: 32531292 PMCID: PMC8018518 DOI: 10.1053/j.gastro.2020.05.060] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Geoffrey A. Preidis
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, Texas
| | - Adam V. Weizman
- Division of Gastroenterology, Mount Sinai Hospital, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Purna C. Kashyap
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Rebecca L. Morgan
- Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
33
|
Chen K, Xin J, Zhang G, Xie H, Luo L, Yuan S, Bu Y, Yang X, Ge Y, Liu C. A combination of three probiotic strains for treatment of acute diarrhoea in hospitalised children: an open label, randomised controlled trial. Benef Microbes 2020; 11:339-346. [PMID: 32720832 DOI: 10.3920/bm2020.0046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Acute diarrhoea continues to be a leading cause of morbidity, hospitalisation, and mortality worldwide, and probiotics have been proposed as a complementary therapy in the treatment of acute diarrhoea. The goal of this study is to assess the efficacy and safety of three combined probiotic strains, Bifidobacterium lactis Bi-07, Lactobacillus rhamnosus HN001, and Lactobacillus acidophilus NCFM, as an adjunct to rehydration therapy in treatment of acute watery diarrhoea in hospitalised children. Eligible diarrheal children were randomised into intervention group (IG, n=96, conventional treatment for diarrhoea in combination with probiotics) and control group (CG, n=98, conventional treatment for diarrhoea without probiotics). The primary assessments of this study were duration of diarrhoea and hospital stay and improvement in diarrhoea symptoms. Significantly more children in the IG showed improvements in diarrhoea (defined as a decrease of stool frequency to no more than four times per day and an improved stool consistency within 24-48 h after the treatment) than those in the CG (96.9 vs 79.6%, P<0.05). Children supplemented with the mixed strains had a 22.5 h shorter (121.4±13.7 h vs 143.9±19.8 h) mean duration of diarrhoea and 1.2 d shorter hospital stays (5.1±1.2 d vs 6.3±1.4 d) than children only receiving the rehydration therapy (P<0.05). The prevalence of constipation of children in the IG (3.1%) was markedly lower (P<0.05) than that of children in the CG (13.3%) after treatment. In conclusion, the mixture of three probiotic strains given to children aged 1-3 years resulted in shorter durations of diarrhoea and hospitalisation and a higher percentage of improved children.
Collapse
Affiliation(s)
- K Chen
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - J Xin
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - G Zhang
- Department of Pediatric Intensive Care Unit, Chengdu Women's & Children's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China P.R
| | - H Xie
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - L Luo
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - S Yuan
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - Y Bu
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - X Yang
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - Y Ge
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - C Liu
- School of Exercise and Nutritional Sciences, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182-7251, USA
| |
Collapse
|
34
|
De Filippis A, Ullah H, Baldi A, Dacrema M, Esposito C, Garzarella EU, Santarcangelo C, Tantipongpiradet A, Daglia M. Gastrointestinal Disorders and Metabolic Syndrome: Dysbiosis as a Key Link and Common Bioactive Dietary Components Useful for their Treatment. Int J Mol Sci 2020; 21:4929. [PMID: 32668581 PMCID: PMC7404341 DOI: 10.3390/ijms21144929] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal (GI) diseases, which include gastrointestinal reflux disease, gastric ulceration, inflammatory bowel disease, and other functional GI disorders, have become prevalent in a large part of the world population. Metabolic syndrome (MS) is cluster of disorders including obesity, hyperglycemia, hyperlipidemia, and hypertension, and is associated with high rate of morbidity and mortality. Gut dysbiosis is one of the contributing factors to the pathogenesis of both GI disorder and MS, and restoration of normal flora can provide a potential protective approach in both these conditions. Bioactive dietary components are known to play a significant role in the maintenance of health and wellness, as they have the potential to modify risk factors for a large number of serious disorders. Different classes of functional dietary components, such as dietary fibers, probiotics, prebiotics, polyunsaturated fatty acids, polyphenols, and spices, possess positive impacts on human health and can be useful as alternative treatments for GI disorders and metabolic dysregulation, as they can modify the risk factors associated with these pathologies. Their regular intake in sufficient amounts also aids in the restoration of normal intestinal flora, resulting in positive regulation of insulin signaling, metabolic pathways and immune responses, and reduction of low-grade chronic inflammation. This review is designed to focus on the health benefits of bioactive dietary components, with the aim of preventing the development or halting the progression of GI disorders and MS through an improvement of the most important risk factors including gut dysbiosis.
Collapse
Affiliation(s)
- Anna De Filippis
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Hammad Ullah
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Alessandra Baldi
- TefarcoInnova, National Inter-University Consortium of Innovative Pharmaceutical Technologies—Parma, 43124 Parma, Italy;
| | - Marco Dacrema
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Cristina Esposito
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Emanuele Ugo Garzarella
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Cristina Santarcangelo
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Ariyawan Tantipongpiradet
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
| | - Maria Daglia
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy; (A.D.F.); (H.U.); (M.D.); (C.E.); (E.U.G.); (C.S.); (A.T.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
35
|
Nwagu TN, Ugwuodo CJ, Onwosi CO, Inyima O, Uchendu OC, Akpuru C. Evaluation of the probiotic attributes of Bacillus strains isolated from traditional fermented African locust bean seeds (Parkia biglobosa), “daddawa”. ANN MICROBIOL 2020. [DOI: 10.1186/s13213-020-01564-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
Background
The involvement of probiotic cultures in food fermentation guarantees enhanced organoleptic properties and maximum consumer health benefits. In this study, isolated Bacillus cultures used in the fermentation of African locust bean seeds “Parkia biglobosa” into the food condiment “daddawa” were evaluated for probiotic attributes. Bacillus cereus strains BC1 and BC2 were tested for tolerance to acid, common salt (NaCl), and bile salt. Auto-aggregation and adhesion to epithelial cells, antibiotic sensitivity profile, hemolytic pattern, and antibacterial activity were also evaluated. To demonstrate further health benefit, spores of strain BC1 were investigated for anti-inflammatory potential employing the rat paw edema technique.
Results
Both Bacillus cereus strains showed antagonistic activity against pathogenic Escherichia coli and Staphylococcus aureus. BC1 was more acid-stress tolerant than BC2, maintaining 107.6% viability after 3 h incubation in MRS broth of pH 2.5. However, at 97.74% viability after incubation for 3 h, BC2 was more tolerant to 0.4 % bile salt. The Bacillus cereus strains were susceptible to all antibiotics tested with the exception of norfloxacin and thrived under high saline stress. Both strains were protease producers and non-hemolytic on sheep blood agar. The edema inhibition study revealed that spores of Bacillus cereus strain BC1 had anti-inflammation potential and produced no physiological toxicity on the animals.
Conclusion
These results indicate that the Bacillus cultures for “daddawa” production are good candidates for probiotics and have the potential for application in both animal and human formulations for increased health benefit to consumers.
Collapse
|
36
|
Chelliah R, Saravanakumar K, Daliri EBM, Kim JH, Lee JK, Jo HY, Kim SH, Ramakrishnan SR, Madar IH, Wei S, Rubab M, Barathikannan K, Ofosu FK, Subin H, Eun-Ji P, Yeong JD, Elahi F, Wang MH, Park JH, Ahn J, Kim DH, Park SJ, Oh DH. Unveiling the potentials of bacteriocin (Pediocin L50) from Pediococcus acidilactici with antagonist spectrum in a Caenorhabditis elegans model. Int J Biol Macromol 2019; 143:555-572. [PMID: 31785295 DOI: 10.1016/j.ijbiomac.2019.10.196] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022]
Abstract
Human-milk-based probiotics play a major role in the early colonization and protection of infants against gastrointestinal infection. We investigated potential probiotics in human milk. Among 41 Lactic acid bacteria (LAB) strains, four strains showed high antimicrobial activity against Escherichia coli 0157:H7, Listeria monocytogenes ATCC 15313, Bacillus cereus ATCC 14576, Staphylococcus aureus ATCC 19095, and Helicobacter pylori. The selected LAB strains were tested in simulated gastrointestinal conditions for their survival. Four LAB strains showed high resistance to pepsin (82%-99%), bile with pancreatine stability (96%-100%), and low pH (80%-94%). They showed moderate cell surface hydrophobicity (22%-46%), auto-aggregation abilities (12%-34%), and 70%-80% co-aggregation abilities against L. monocytogenes ATCC 15313, S. aureus ATCC 19095, B. cereus ATCC 14576, and E. coli 0157:H7. All four selected isolates were resistant to gentamicin, imipenem, novobiocin, tetracycline, clindamycin, meropenem, ampicillin, and penicillin. The results show that Pediococcus acidilatici is likely an efficient probiotic strain to produce < 3 Kda pediocin-based antimicrobial peptides, confirmed by applying amino acid sequences), using liquid chromatography mass spectrometry and HPLC with the corresponding sequences from class 2 bacteriocin, and based on the molecular docking, the mode of action of pediocin was determined on LipoX complex, further the 13C nuclear magnetic resonance structural analysis, which confirmed the antimicrobial peptide as pediocin.
Collapse
Affiliation(s)
- Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Kandasamy Saravanakumar
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Joong-Hark Kim
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea; Erom, Co., Ltd, Chuncheon, Gangwon-do 24427, South Korea
| | - Jung-Kun Lee
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea; Erom, Co., Ltd, Chuncheon, Gangwon-do 24427, South Korea
| | - Hyeon-Yeong Jo
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Se-Hun Kim
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | | | - Inamul Hasan Madar
- Department of Biochemistry, School of Life Science, Bharathidasan University, Thiruchirappalli, Tamilnadu, India
| | - Shuai Wei
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea; Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Momna Rubab
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Kaliyan Barathikannan
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Fred Kwame Ofosu
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Hwang Subin
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Park Eun-Ji
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Jung Da Yeong
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Fazle Elahi
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Myeong-Hyeon Wang
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Jong Hwan Park
- Laboratory Animal Medicine, Chonnam National University, Gwangju, South Korea
| | - Juhee Ahn
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Dong-Hwan Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Sung Jin Park
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon, Gangwon-do 24341, South Korea.
| |
Collapse
|
37
|
Patro-Gołąb B, Szajewska H. Systematic Review with Meta-Analysis: Lactobacillus reuteri DSM 17938 for Treating Acute Gastroenteritis in Children. An Update. Nutrients 2019; 11:nu11112762. [PMID: 31739457 PMCID: PMC6893691 DOI: 10.3390/nu11112762] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
The effectiveness of Lactobacillus reuteri DSM 17938 (L. reuteri) for the management of acute gastroenteritis (AGE) has been recently questioned. We performed a systematic review to update evidence on L. reuteri for treating AGE in children. We searched MEDLINE, EMBASE, the Cochrane Library databases, and additional data sources from January 2016 (end of search for our 2016 systematic review) to August 2019. The primary outcomes were stool volume and duration of diarrhea. Four RCTs were included. None of them evaluated stool volume. Compared with placebo or no treatment, L. reuteri reduced diarrhea duration (four RCTs, n = 347, mean difference, MD −0.87 days, 95% CI [−1.43, −0.31]). L. reuteri use was also associated with a reduced duration of hospitalization (three RCTs, n = 284, MD −0.54 days, 95% CI [−1.09, 0.0]). The small effect sizes of limited clinical relevance and methodological limitations of the included trials should be noted when interpreting these findings.
Collapse
|
38
|
The Effectiveness of Synbiotics in Preventing Antibiotic-Associated Diarrhea in Children: A Double-Blind Randomized Clinical Trial. ARCHIVES OF PEDIATRIC INFECTIOUS DISEASES 2019. [DOI: 10.5812/pedinfect.89707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Xiang R, Tang Q, Chen XQ, Li MY, Yang MX, Yun X, Huang L, Shan QW. Effects of Zinc Combined with Probiotics on Antibiotic-associated Diarrhea Secondary to Childhood Pneumonia. J Trop Pediatr 2019; 65:421-426. [PMID: 30521044 DOI: 10.1093/tropej/fmy069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
AIM The aim of this study was to evaluate the impact of zinc combined with probiotics (Bifico) on antibiotic-associated diarrhea (AAD) secondary to pneumonia. METHODS A total of 50 patients with AAD secondary to pneumonia were randomly divided into a probiotics group (Bifico) and a combined group (zinc combined with Bifico) and 25 pneumonia patients without AAD as the control group. Serum levels of zinc, diamine oxidase (DAO) activity, D-lactate and intestinal flora [Bifidobacterium, Escherichia coli and Bifidobacterium/E. coli (B/E) ratio] were detected before and after intervention. RESULTS The results showed that zinc combined with Bifico had significantly higher overall efficiency than Bifico alone for treatment of AAD secondary to pneumonia. Notably, the combined treatment increased the population of Bifidobacterium, while the number of E. coli was reduced, the B/E value was improved and DAO activity and D-lactate levels were markedly reduced. CONCLUSION Patients with AAD secondary to pneumonia benefit from zinc supplementation of probiotic treatment.
Collapse
Affiliation(s)
- Rong Xiang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Qing Tang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Xiu-Qi Chen
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Mu-Yan Li
- The Medical Science Experiment Center, Guangxi Medical University, Nanning city, China
| | - Mei-Xiong Yang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Xiang Yun
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Li Huang
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| | - Qing-Wen Shan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning city, China
| |
Collapse
|
40
|
Yang B, Lu P, Li MX, Cai XL, Xiong WY, Hou HJ, Ha XQ. A meta-analysis of the effects of probiotics and synbiotics in children with acute diarrhea. Medicine (Baltimore) 2019; 98:e16618. [PMID: 31517810 PMCID: PMC6750275 DOI: 10.1097/md.0000000000016618] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
OBJECTIVE This meta-analysis assessed the effectiveness of probiotics and synbiotics for acute diarrhea (AD) in children and investigated probiotic formulations, types of interventions, and country factors. METHODS Randomized, double-blind, placebo-controlled trials evaluating the effects of probiotics or synbiotics on AD were analyzed. We followed the recommendations of the Cochrane Handbook and the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) statement. The risks of systematic errors (bias) and random errors were assessed, and the overall quality of the evidence was evaluated using the Grades of Recommendations Assessment, Development, and Evaluation (GRADE) approach. RESULTS The meta-analysis included 34 studies with 4911 patients. Five and 29 studies presented the results of synbiotic and probiotic interventions, respectively. After intervention, the durations of diarrhea (weighted mean difference (WMD) = -16.63 [-20.16; -12.51]) and hospitalization (risk ratio (RR) = 0.59 [0.48; 0.73]) were shorter, the stool frequency on day 3 (WMD = -0.98 [-1.55; -0.40]) was decreased, and the incidence of diarrhea lasting 3 days was lower in the probiotic and synbiotic groups than in the control groups. Furthermore, in the subgroup analyses, synbiotics were more effective than probiotics at reducing the durations of diarrhea and hospitalization, and Saccharomyces and Bifidobacterium were more effective than Lactobacillus at reducing the duration of diarrhea. CONCLUSION This meta-analysis supports the potential beneficial roles of probiotics and synbiotics for AD in children. Further research is needed to determine problems associated with probiotic/synbiotic mixtures and appropriate dosages.
Collapse
Affiliation(s)
- Bo Yang
- Department of Clinical Laboratory, The People's Liberation Arimy Joint Service Support Unit 940 Hospital
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine (TCM), Lanzhou
| | - Ping Lu
- School of Clinical Medicine, Capital Medical University, Beijing
| | - Mei-Xuan Li
- Evidence Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Xiao-Ling Cai
- Department of Clinical Laboratory, The People's Liberation Arimy Joint Service Support Unit 940 Hospital
| | - Wan-Yuan Xiong
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine (TCM), Lanzhou
| | - Huai-Jing Hou
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine (TCM), Lanzhou
| | - Xiao-Qin Ha
- Department of Clinical Laboratory, The People's Liberation Arimy Joint Service Support Unit 940 Hospital
| |
Collapse
|
41
|
De Almeida CV, Lulli M, di Pilato V, Schiavone N, Russo E, Nannini G, Baldi S, Borrelli R, Bartolucci G, Menicatti M, Taddei A, Ringressi MN, Niccolai E, Prisco D, Rossolini GM, Amedei A. Differential Responses of Colorectal Cancer Cell Lines to Enterococcus faecalis' Strains Isolated from Healthy Donors and Colorectal Cancer Patients. J Clin Med 2019; 8:388. [PMID: 30897751 PMCID: PMC6463247 DOI: 10.3390/jcm8030388] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 02/06/2023] Open
Abstract
The metabolites produced by the host's gut microbiota have an important role in the maintenance of intestinal homeostasis, but can also act as toxins and induce DNA damage in colorectal epithelial cells increasing the colorectal cancer (CRC) chance. In this scenario, the impact of some of the components of the natural human gastrointestinal microbiota, such as Enterococcus faecalis (E. faecalis), at the onset of CRC progression remains controversial. Since under dysbiotic conditions it could turn into a pathogen, the aim of this study was to compare the effect of E. faecalis' strains (isolated from CRC patients and healthy subjects' stools) on the proliferation of different colorectal cells lines. First, we isolated and genotyping characterized the Enterococcus faecalis' strains. Then, we analyzed the proliferation index (by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay) of three tumor and one normal intestinal cell lines, previously exposed to E. faecalis strains pre-cultured medium. Stool samples of CRC patients demonstrated a reduced frequency of E. faecalis compared to healthy subjects. In addition, the secreted metabolites of E. faecalis' strains, isolated from healthy donors, decreased the human ileocecal adenocarcinoma cell line HCT-8 and human colon carcinoma cell line HCT-116 cell proliferation without effects on human colorectal adenocarcinoma cell line SW620 and on normal human diploid cell line CLR-1790. Notably, the metabolites of the strains isolated from CRC patients did not influence the cell growth of CRC cell lines. Our results demonstrated a new point of view in the investigation of E. faecalis' role in CRC development, which raises awareness of the importance of not only associating the presence/absence of a unique microorganism, but also in defining the specific characteristics of the different investigated strains.
Collapse
Affiliation(s)
| | - Matteo Lulli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy.
| | - Vincenzo di Pilato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Nicola Schiavone
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy.
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Rossella Borrelli
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, 50139 Florence, Italy.
| | - Marta Menicatti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, 50139 Florence, Italy.
| | - Antonio Taddei
- Department of Surgery and Translational Medicine, University of Florence, 50134 Florence, Italy.
| | - Maria Novella Ringressi
- Department of Surgery and Translational Medicine, University of Florence, 50134 Florence, Italy.
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Domenico Prisco
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
- Department of Microbiology and Virology Unit, Florence Careggi University Hospital, 50134 Florence, Italy.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
- Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy.
| |
Collapse
|
42
|
Industry funding effect on positive results of probiotic use in the management of acute diarrhea: a systematized review. Eur J Gastroenterol Hepatol 2019; 31:289-302. [PMID: 30557228 DOI: 10.1097/meg.0000000000001322] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Several investigations have found that industry-funded studies tend to inform results favoring the sponsored products. The pressure to demonstrate that a drug or a product causes a favorable outcome may result in investigation biases from industry-funded research. One example of this could be found in the probiotic research funded by the industry. The aim of this study was to assess the effect of industry funding on positive outcomes of the use of probiotics in the management of acute diarrhea. A systematized review of clinical trials on the use of probiotics in the management of acute diarrhea was performed. The associations between the source of funding, clinical outcomes, probiotic genus, and quality of the study were assessed using the χ-test and Fisher's exact test. Sixty-six clinical trials were included; 27 were industry funded, 18 were nonindustry funded, and 21 did not disclose their funding source. There were 48 positive and 30 negative clinical outcomes. There was no significant association between the source of funding and clinical outcomes (P=0.491). No association between the rest of the studied variables and outcomes was observed either (P>0.05). In clinical trials on the use of probiotics in the management of acute diarrhea, the source of funding has no influence on positive clinical outcomes.
Collapse
|
43
|
Ling Z, Liu X, Guo S, Cheng Y, Shao L, Guan D, Cui X, Yang M, Xu X. Role of Probiotics in Mycoplasma pneumoniae Pneumonia in Children: A Short-Term Pilot Project. Front Microbiol 2019; 9:3261. [PMID: 30687259 PMCID: PMC6334620 DOI: 10.3389/fmicb.2018.03261] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022] Open
Abstract
Mycoplasma pneumoniae is one of the most common pathogens causing community-acquired pneumonia in children. Mycoplasma pneumoniae pneumonia (MPP) can be successfully treated with azithromycin; however, antibiotic-associated diarrhea (AAD) is a common adverse effect. Increasing evidence suggests that some probiotics may prevent the development of AAD. The present study determined the effects of probiotics (live Clostridium butyricum plus Bifidobacterium infantis) on the prevention and treatment of AAD in children with MPP when co-administered with intravenous azithromycin. Fifty-five children with MPP were enrolled and received azithromycin (10 mg/kg/day; once daily for 7 days) combined with probiotics (starting on the third day of azithromycin treatment; 1,500 mg three times daily); 50 healthy children served as controls. At the end of the trial, the incidence of AAD, fecal microbiota, intestinal mucosal barriers, and systemic inflammation were analyzed using recommended systems biology techniques. No cases of AAD or other adverse events occurred in children with MPP after co-administration of probiotics with azithromycin. A live C. butyricum plus B. infantis preparation partly reconstructed the gut microbiota, especially restoration of bacterial diversity. The indicators of intestinal mucosal barrier function, such as D-lactate, endotoxin, and diamine oxidase, were significantly improved and the systemic inflammation (interleukin 10) was attenuated after probiotic therapy. The present study indicated that co-administration of probiotics with azithromycin is a promising therapy for MPP treatment which could prevent and treat AAD effectively.
Collapse
Affiliation(s)
- Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shu Guo
- Department of Gastroenterology, Affiliated Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Li Shao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dexiu Guan
- Department of Gastroenterology, Affiliated Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Xiaoshuang Cui
- Department of Gastroenterology, Affiliated Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Mingming Yang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiwei Xu
- Department of Gastroenterology, Affiliated Beijing Children’s Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
44
|
Fernandez-Duarte KP, Olaya-Galán NN, Salas-Cárdenas SP, Lopez-Rozo J, Gutierrez-Fernandez MF. Bifidobacterium adolescentis (DSM 20083) and Lactobacillus casei (Lafti L26-DSL): Probiotics Able to Block the In Vitro Adherence of Rotavirus in MA104 Cells. Probiotics Antimicrob Proteins 2018; 10:56-63. [PMID: 28432676 PMCID: PMC5801392 DOI: 10.1007/s12602-017-9277-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Rotavirus is the leading worldwide cause of gastroenteritis in children under five years of age. Even though there are some available vaccines to prevent the disease, there are limited strategies for challenging diarrhea induced by rotavirus infection. For this reason, researchers are constantly searching for other approaches to control diarrhea by means of probiotics. In order to demonstrate the ability of some probiotic bacteria to interfere with the in vitro rotavirus infection in MA104 cells, strains of Lactobacillus sp. and Bifidobacterium sp. were tested in MA104 cells before the viral infection. As a preliminary assay, a blocking effect treatment was performed with viable bacteria. In this screening assay, four of initial ten bacteria showed a slight reduction of the viral infection (measured by percentage of infection). L. casei (Lafti L26-DSL), L. fermentum(ATCC 9338), B. adolescentis (DSM 20083), and B. bifidum (ATCC 11863) were used in further experiments. Three different treatments were tested in order to evaluate protein-based metabolites obtained from mentioned bacteria: (i) cell exposure to the protein-based metabolites before viral infection, (ii) exposure to protein-based metabolites after viral infection, and (iii) co-incubation of the virus and protein-based metabolites before viral infection to the cell culture. The best effect performed by protein-based metabolites was observed during the co-incubation assay of the virus and protein-based metabolites before adding them into the cell culture. The results showed 25 and 37% of infection in the presence of L. casei and B. adolescentis respectively. These results suggest that the antiviral effect may be occurring directly with the viral particle instead of making a blocking effect of the cellular receptors that are needed for the viral entrance.
Collapse
Affiliation(s)
- Karem Prunella Fernandez-Duarte
- Infectious Diseases Research Group, Department of Microbiology, Pontificia Universidad Javeriana, Carrera 7 #40-62, (50-123), Bogota, Colombia
| | - Nury Nathalia Olaya-Galán
- Infectious Diseases Research Group, Department of Microbiology, Pontificia Universidad Javeriana, Carrera 7 #40-62, (50-123), Bogota, Colombia
| | - Sandra Patricia Salas-Cárdenas
- Infectious Diseases Research Group, Department of Microbiology, Pontificia Universidad Javeriana, Carrera 7 #40-62, (50-123), Bogota, Colombia
| | - Jazmin Lopez-Rozo
- Infectious Diseases Research Group, Department of Microbiology, Pontificia Universidad Javeriana, Carrera 7 #40-62, (50-123), Bogota, Colombia
| | - Maria Fernanda Gutierrez-Fernandez
- Infectious Diseases Research Group, Department of Microbiology, Pontificia Universidad Javeriana, Carrera 7 #40-62, (50-123), Bogota, Colombia.
| |
Collapse
|
45
|
Li HH, Li YP, Zhu Q, Qiao JY, Wang WJ. Dietary supplementation with Clostridium butyricum helps to improve the intestinal barrier function of weaned piglets challenged with enterotoxigenic Escherichia coli K88. J Appl Microbiol 2018; 125:964-975. [PMID: 29851202 DOI: 10.1111/jam.13936] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 04/11/2018] [Accepted: 05/27/2018] [Indexed: 11/28/2022]
Abstract
AIMS The objective of this study was twofold: (i) to examine the effect of Clostridium butyricum on intestinal barrier function and (ii) to elucidate the mechanisms involved in enhanced intestinal barrier function. METHODS AND RESULTS Forty-eight weaned piglets were assigned randomly to either a basal diet or a C. butyricum-supplemented diet. On day 15, all pigs were orally challenged with enterotoxigenic Escherichia coli (ETEC) K88 or saline. Clostridium butyricum decreased serum diamine oxidase activity and d-lactic acid concentration, as well as increased intestinal tight junction proteins (ZO-1, claudin-3 and occludin) expression in ETEC K88-infected pigs. Moreover, C. butyricum decreased IL-1β and IL-18 levels in serum and gut, whereas it increased IL-10 levels. Furthermore, C. butyricum downregulated NLRP3 and caspase-1 expression in ETEC K88-challenged pig gut, but did not affect apoptosis-associated speck-like protein expression. CONCLUSIONS Clostridium butyricum enhanced intestinal barrier function and inhibited apoptosis-associated speck-like protein-independent NLRP3 inflammasome signalling pathway in weaned piglets after ETEC K88 challenge. SIGNIFICANCE AND IMPACT OF THE STUDY The novelty of this study lies in the beneficial effects of C. butyricum on intestinal health, likely by improving intestinal barrier function and alleviating inflammatory reactions.
Collapse
Affiliation(s)
- H-H Li
- Tianjin Animal Husbandry and Veterinary Research Institute, Tianjin, China
| | - Y-P Li
- Tianjin Animal Husbandry and Veterinary Research Institute, Tianjin, China
| | - Q Zhu
- Tianjin Animal Husbandry and Veterinary Research Institute, Tianjin, China
| | - J-Y Qiao
- Tianjin Animal Husbandry and Veterinary Research Institute, Tianjin, China
| | - W-J Wang
- Tianjin Animal Husbandry and Veterinary Research Institute, Tianjin, China
| |
Collapse
|
46
|
Matsuura Y, Takasaki M, Miyazawa R, Nakanishi T. Stimulatory effects of heat-killed Enterococcus faecalis on cell-mediated immunity in fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 74:1-9. [PMID: 28389237 DOI: 10.1016/j.dci.2017.03.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/24/2017] [Accepted: 03/24/2017] [Indexed: 06/07/2023]
Abstract
Intracellular bacterial and viral diseases are widespread in the aquaculture industry and cause serious economic losses. Development of effective vaccines and adjuvants that can induce cell-mediated immunity is urgently needed for prevention of these diseases. Here we report the immunostimulatory effects of probiotic bacteria ''E. faecalis'' in ginbuna crucian carp Carassius auratus langsdorfii. Intraperitoneal injection of heat-killed E. faecalis induced an increase in CD4-1+ lymphocytes, CD8α+ lymphocytes and macrophages in vivo. Expression of Th1 cytokine genes was enhanced by exposure to the bacteria in vitro. We identified the leukocyte subsets that expressed specific Th1 cytokine genes: granulocytes and macrophages produced IL12 and IFNγrel2, respectively, while lymphocytes produced IFNγs including IFNγ1 and IFNγ2. Finally, expression of Th1 cytokines was also enhanced by intraperitoneal injection of heat-killed E. faecalis in vivo, while expression of Th2 cytokine was unchanged. Together, these findings suggest that heat-killed E. faecalis can induce cell-mediated immunity in fish.
Collapse
|
47
|
Ba HV, Seo HW, Cho SH, Kim YS, Kim JH, Park BY, Kim HW, Ham JS, Seong PN. Utilisation possibility ofEnterococcus faecalisisolates from neonate's faeces for production of fermented sausages as starter cultures. Int J Food Sci Technol 2017. [DOI: 10.1111/ijfs.13440] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hoa Van Ba
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Hyun-Woo Seo
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Soo-Hyun Cho
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Yoon-Seok Kim
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Jin-Hyoung Kim
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Beom-Young Park
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Hyun-Wook Kim
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Jun-Sang Ham
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| | - Pil-Nam Seong
- Animal Products Development and Processing Division; National Institute of Animal Science; 1500 kongjwipatjwi-ro Iseo-Myeon Wanju-gun Jeollabuk-do 565-851 Korea
| |
Collapse
|
48
|
Homayoni Rad A, Vaghef Mehrabany E, Alipoor B, Vaghef Mehrabany L. The Comparison of Food and Supplement as Probiotic Delivery Vehicles. Crit Rev Food Sci Nutr 2017; 56:896-909. [PMID: 25117939 DOI: 10.1080/10408398.2012.733894] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Probiotics are live bacteria which have frequently been reported to be beneficial in preventing a wide range of diseases as well as playing a major role in treating the existing ailments. Thus far, a variety of probiotic products have been developed which can be categorized into two groups: probiotic foods and supplements. Both foods and supplements have been able to confer the health benefits claimed for them. However, it is not known which one can be clinically more efficient, and to the best of our knowledge, until now no research has been conducted to investigate this issue. The present review aims to discuss this matter, based on the evidence available in the literature. To do so, articles indexed in PubMed and ScienceDirect between 2000 and 2011 were reviewed. The articles included the clinical trials in which either foods or supplements were used to administer the probiotics to either patients suffering from different diseases or healthy subjects. Although both foods and supplements seem to have been efficient carriers for the beneficial bacteria, to generally promote public health in communities, probiotic foods appear to be preferred to probiotic supplements.
Collapse
Affiliation(s)
- Aziz Homayoni Rad
- a Faculty of Nutrition, Department of Food Science and Technology, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Elnaz Vaghef Mehrabany
- b Faculty of Nutrition, Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Beitullah Alipoor
- b Faculty of Nutrition, Department of Nutrition, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Leila Vaghef Mehrabany
- c School of Nutritional Sciences and Dietetics, Department of Clinical Nutrition, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
49
|
Moraes-Filho JP, Quigley EMM. THE INTESTINAL MICROBIOTA AND THE ROLE OF PROBIOTICS IN IRRITABLE BOWEL SYNDROME: a review. ARQUIVOS DE GASTROENTEROLOGIA 2016; 52:331-8. [PMID: 26840477 DOI: 10.1590/s0004-28032015000400015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/13/2015] [Indexed: 12/12/2022]
Abstract
Irritable bowel syndrome is a common, chronic relapsing gastrointestinal disorder that affects 7%-22% of the population worldwide. According to Rome III Criteria, the disorder is defined by the coexistence of abdominal discomfort or pain associated with an alteration in bowel habits. Its pathophysiology is not completely understood but, in addition to some important abnormalities, the disturbed intestinal microbiota has also been described supported by several strands of evidence. The treatment of irritable bowel syndrome is based upon several therapeutic approaches but few have been successful or without adverse events and more recently the gut microbiota and the use of probiotics have emerged as a factor to be considered. Probiotics are live micro-organisms which when consumed in adequate amounts confer a health benefit to the host, such as Lactic bacteria among others. An important scientific rationale has emerged for the use of probiotics in irritable bowel syndrome, although the data regarding different species are still limited. Not all probiotics are beneficial: it is important to select the specific strain which should be supported by good evidence base. The mechanisms of action of probiotics are described and the main strains are quoted.
Collapse
|
50
|
Khatri I, Sharma S, Ramya TNC, Subramanian S. Complete Genomes of Bacillus coagulans S-lac and Bacillus subtilis TO-A JPC, Two Phylogenetically Distinct Probiotics. PLoS One 2016; 11:e0156745. [PMID: 27258038 PMCID: PMC4892684 DOI: 10.1371/journal.pone.0156745] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/18/2016] [Indexed: 12/31/2022] Open
Abstract
Several spore-forming strains of Bacillus are marketed as probiotics due to their ability to survive harsh gastrointestinal conditions and confer health benefits to the host. We report the complete genomes of two commercially available probiotics, Bacillus coagulans S-lac and Bacillus subtilis TO-A JPC, and compare them with the genomes of other Bacillus and Lactobacillus. The taxonomic position of both organisms was established with a maximum-likelihood tree based on twenty six housekeeping proteins. Analysis of all probiotic strains of Bacillus and Lactobacillus reveal that the essential sporulation proteins are conserved in all Bacillus probiotic strains while they are absent in Lactobacillus spp. We identified various antibiotic resistance, stress-related, and adhesion-related domains in these organisms, which likely provide support in exerting probiotic action by enabling adhesion to host epithelial cells and survival during antibiotic treatment and harsh conditions.
Collapse
Affiliation(s)
- Indu Khatri
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - Shailza Sharma
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | - T. N. C. Ramya
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, India
| | | |
Collapse
|