1
|
Miao X, Jiang P, Zhang X, Li X, Wu Z, Jiang Y, Liu H, Xie W, Li X, Shi B, Cai J, Gong W. Lactobacillus rhamnosus HN001 facilitates the efficacy of dual PI3K/mTOR inhibition prolonging cardiac transplant survival and enhancing antitumor effect. Microbiol Spectr 2024; 12:e0183923. [PMID: 38564670 PMCID: PMC11064485 DOI: 10.1128/spectrum.01839-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/24/2023] [Indexed: 04/04/2024] Open
Abstract
UNLABELLED Solid organ transplantation is a crucial treatment for patients who have reached the end stage of heart, lung, kidney, or liver failure. However, the likelihood of developing cancer post-transplantation increases. Additionally, primary malignant tumors remain a major obstacle to the long-term survival of transplanted organs. Therefore, it is essential to investigate effective therapies that can boost the immune system's ability to combat cancer and prevent allograft rejection. We established a mouse orthotopic liver tumor model and conducted allogeneic heterotopic heart transplantation. Various treatments were administered, and survival curves were generated using the Kaplan-Meier method. We also collected graft samples and measured inflammatory cytokine levels in the serum using an inflammatory array. The specificity of the histochemical techniques was tested by staining sections. We administered a combination therapy of phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) dual inhibitor BEZ235 and Lactobacillus rhamnosus HN001 to primary liver cancer model mice with cardiac allografts. Consistent with our prior findings, L. rhamnosus HN001 alleviated the intestinal flora imbalance caused by BEZ235. Our previous research confirmed that the combination of BEZ235 and L. rhamnosus HN001 significantly prolonged cardiac transplant survival. IMPORTANCE We observed that the combination of phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) dual inhibitor BEZ235 and Lactobacillus rhamnosus HN001 notably prolonged cardiac transplant survival while also inhibiting the progression of primary liver cancer. The combination therapy was efficacious in treating antitumor immunity and allograft rejection, as demonstrated by the efficacy results. We also found that this phenomenon was accompanied by the regulation of inflammatory IL-6 expression. Our study presents a novel and effective therapeutic approach to address antitumor immunity and prevent allograft rejection.
Collapse
Affiliation(s)
- Xiaolong Miao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Jiang
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Zhang
- Medical department, Qingdao Eighth People’s Hospital, Qingdao, China
| | - Xinqiang Li
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zelai Wu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuancong Jiang
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Weixun Xie
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinwei Li
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bingfeng Shi
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinzhen Cai
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihua Gong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
2
|
Guo L, Ze X, Feng H, Liu Y, Ge Y, Zhao X, Song C, Jiao Y, Liu J, Mu S, Yao S. Identification and quantification of viable Lacticaseibacillus rhamnosus in probiotics using validated PMA-qPCR method. Front Microbiol 2024; 15:1341884. [PMID: 38298895 PMCID: PMC10828034 DOI: 10.3389/fmicb.2024.1341884] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/03/2024] [Indexed: 02/02/2024] Open
Abstract
The identification and quantification of viable bacteria at the species/strain level in compound probiotic products is challenging now. Molecular biology methods, e.g., propidium monoazide (PMA) combination with qPCR, have gained prominence for targeted viable cell counts. This study endeavors to establish a robust PMA-qPCR method for viable Lacticaseibacillus rhamnosus detection and systematically validated key metrics encompassing relative trueness, accuracy, limit of quantification, linear, and range. The inclusivity and exclusivity notably underscored high specificity of the primers for L. rhamnosus, which allowed accurate identification of the target bacteria. Furthermore, the conditions employed for PMA treatment were fully verified by 24 different L. rhamnosus including type strain, commercial strains, etc., confirming its effective discrimination between live and dead bacteria. A standard curve constructed by type strain could apply to commercial strains to convert qPCR Cq values to viable cell numbers. The established PMA-qPCR method was applied to 46 samples including pure cultures, probiotics as food ingredients, and compound probiotic products. Noteworthy is the congruity observed between measured and theoretical values within a 95% confidence interval of the upper and lower limits of agreement, demonstrating the relative trueness of this method. Moreover, accurate results were obtained when viable L. rhamnosus ranging from 103 to 108 CFU/mL. The comprehensive appraisal of PMA-qPCR performances provides potential industrial applications of this new technology in quality control and supervision of probiotic products.
Collapse
Affiliation(s)
- Lizheng Guo
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Xiaolei Ze
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Huifen Feng
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Yiru Liu
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Yuanyuan Ge
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Xi Zhao
- Microbiome Research and Application Center, BYHEALTH Institute of Nutrition & Health, Guangzhou, China
| | - Chengyu Song
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Yingxin Jiao
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Jiaqi Liu
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Shuaicheng Mu
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| | - Su Yao
- China National Research Institute of Food and Fermentation Industries Co., LTD., China Center of Industrial Culture Collection, Beijing, China
| |
Collapse
|
3
|
Transcriptome Analysis Reveals Effect of Dietary Probiotics on Immune Response Mechanism in Southern Catfish ( Silurus meridionalis) in Response to Plesiomonas shigelloides. Animals (Basel) 2023; 13:ani13030449. [PMID: 36766339 PMCID: PMC9913393 DOI: 10.3390/ani13030449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
To explore whether a probiotic complex composed of Lactobacillus rhamnosus, Lactobacillus plantarum, and Lactobacillus casei can prevent or inhibit the inflammatory response caused by the invasion of Plesiomonas shigelloides in the southern catfish, we screened differentially expressed genes and enriched inflammation-related pathways among a control and three experimental groups and conducted analysis by transcriptome sequencing after a 21-day breeding experiment. Compared with those in the PS (Plesiomonas shigelloides) group, southern catfish in the L-PS (Lactobacillus-Plesiomonas shigelloides) group had no obvious haemorrhages or ulcerations. The results also showed that inflammation-related genes, such as mmp9, cxcr4, nfkbia, socs3, il-8, pigr, tlr5, and tnfr1, were significantly upregulated in the PS group compared with those in the L-PS groups. In addition, we verified six DEGs (mmp9, cxcr4, nfkbia, socs3, rbp2, and calr) and three proteins (CXCR4, NFKBIA, and CALR) by qRT-PCR and ELISA, respectively. Our results were consistent with the transcriptome data. Moreover, significantly downregulated genes (p < 0.05) were enriched in inflammation-related GO terms (lymphocyte chemotaxis and positive regulation of inflammatory response) and immune-related pathways (intestinal immune network for IgA production and IL-17 signalling pathway) in the L-PS vs. the PS group. Our results indicate that the infection of P. shigelloides can produce an inflammatory response, and probiotics could inhibit the inflammatory response caused by P. shigelloides to some extent.
Collapse
|
4
|
Parreno V, Bai M, Liu F, Jing J, Olney E, Li G, Wen K, Yang X, Castellucc TB, Kocher JF, Zhou X, Yuan L. Probiotic as Adjuvant Significantly Improves Protection of the Lanzhou Trivalent Rotavirus Vaccine against Heterologous Challenge in a Gnotobiotic Pig Model of Human Rotavirus Infection and Disease. Vaccines (Basel) 2022; 10:vaccines10091529. [PMID: 36146607 PMCID: PMC9506166 DOI: 10.3390/vaccines10091529] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 11/25/2022] Open
Abstract
This preclinical study in the gnotobiotic (Gn) pig model of human rotavirus (HRV) infection and disease evaluates the effect of probiotic Lactobacillus rhamnosus GG (LGG) as a mucosal adjuvant on the immunogenicity and cross-protective efficacy of the Lanzhou live oral trivalent (G2, G3, G4) vaccine (TLV, aka LLR3). Gn pigs were immunized with three doses of TLV with or without concurrent administration of nine doses of LGG around the time of the first dose of the TLV vaccination, and were challenged orally with the virulent heterotypic Wa G1P[8] HRV. Three doses of TLV were highly immunogenic and conferred partial protection against the heterotypic HRV infection. LGG significantly enhanced the intestinal and systemic immune responses and improved the effectiveness of protection against the heterotypic HRV challenge-induced diarrhea and virus shedding. In conclusion, we demonstrated the immune-stimulating effects of probiotic LGG as a vaccine adjuvant and generated detailed knowledge regarding the cross-reactive and type-specific antibody and effector B and T cell immune responses induced by the TLV. Due to the low cost, ease of distribution and administration, and favorable safety profiles, LGG as an adjuvant has the potential to play a critical role in improving rotavirus vaccine efficacy and making the vaccines more cost-effective.
Collapse
Affiliation(s)
- Viviana Parreno
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- INCUINTA, Institutot de Virología e Innovaciones Tecnológicas (IVIT), Instituto Nacional de Tecnología Agropecuaria (INTA)-CONICET, Ciudad Autónoma de Buenos Aires C1033AAE, Argentina
| | - Muqun Bai
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Lanzhou Institute of Biological Products, Lanzhou 730046, China
| | - Fangning Liu
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jiqiang Jing
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Animal Medicine, Shandong Vocational College of Animal Husbandry and Veterinary Medicine, Weifang 261071, China
| | - Erika Olney
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA
| | - Guohua Li
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Ke Wen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Xingdong Yang
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Tammy Bui Castellucc
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Jacob F. Kocher
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Xu Zhou
- Lanzhou Institute of Biological Products, Lanzhou 730046, China
| | - Lijuan Yuan
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Correspondence: ; Tel.: +86-540-231-9053
| |
Collapse
|
5
|
Negi S, Hashimoto-Hill S, Alenghat T. Neonatal microbiota-epithelial interactions that impact infection. Front Microbiol 2022; 13:955051. [PMID: 36090061 PMCID: PMC9453604 DOI: 10.3389/fmicb.2022.955051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Despite modern therapeutic developments and prophylactic use of antibiotics during birth or in the first few months of life, enteric infections continue to be a major cause of neonatal mortality and morbidity globally. The neonatal period is characterized by initial intestinal colonization with microbiota and concurrent immune system development. It is also a sensitive window during which perturbations to the environment or host can significantly impact colonization by commensal microbes. Extensive research has demonstrated that these early life alterations to the microbiota can lead to enhanced susceptibility to enteric infections and increased systemic dissemination in newborns. Various contributing factors continue to pose challenges in prevention and control of neonatal enteric infections. These include alterations in the gut microbiota composition, impaired immune response, and effects of maternal factors. In addition, there remains limited understanding for how commensal microbes impact host-pathogen interactions in newborns. In this review, we discuss the recent recognition of initial microbiota-epithelial interactions that occur in neonates and can regulate susceptibility to intestinal infection. These studies suggest the development of neonatal prophylactic or therapeutic regimens that include boosting epithelial defense through microbiota-directed interventions.
Collapse
|
6
|
Vecchio AL, Nunziata F, Bruzzese D, Conelli ML, Guarino A. Rotavirus immunisation status affects the efficacy of Lacticaseibacillus rhamnosus GG for the treatment of children with acute diarrhoea: a meta-analysis. Benef Microbes 2022; 13:283-294. [PMID: 36004717 DOI: 10.3920/bm2022.0024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The efficacy of Lacticaseibacillus rhamonosus GG (LGG) for the treatment of children with acute gastroenteritis has been debated based on most recent evidence. Previous evidence demonstrated that LGG mainly benefits children with Rotavirus infection compared to other aetiologies. However, Rotavirus immunisation (RVI) has been implemented worldwide since 2006. We aimed to investigate whether the efficacy of LGG in children with gastroenteritis vary according to RVI status. The MEDLINE, Embase and Cochrane library databases were searched for relevant randomised controlled trials (RCT) up to April 2022. The duration of diarrhoea and episodes lasting >48 h were considered as primary outcomes. The date of vaccine introduction and RVI coverage were reviewed for all countries where trials were conducted. Among the 15 RCTs included in the analysis (n=3,465), only 5 showed a low risk of bias. In RCT conducted before the introduction of RVI (n=2,932), LGG was effective in reducing the duration of diarrhoea compared with placebo or standard care (Median -23.80 h, 95% confidence interval (CI) -36.59 to -11.02]). Only 2 RCTs (n=1,072) reported data of populations partially immunised against Rotavirus with an overall coverage of 44 and 67%, respectively. In this population, LGG showed no efficacy in reducing the duration of diarrhoea (Median -5.34, 95%CI -12.9 to 2.22). Similarly, LGG reduced the risk of diarrhoea lasting >48 h in children not immunised against Rotavirus (RR 0.73, 95%CI 0.54-0.99), but not in population partially immunised (RR 0.98, 95%CI 0.87 to 1.11). The implementation of RVI might affect the efficacy of LGG modifying local epidemiology and susceptibility of the target population to selected probiotics.
Collapse
Affiliation(s)
- A Lo Vecchio
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - F Nunziata
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - D Bruzzese
- Department of Public Health, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - M L Conelli
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| | - A Guarino
- Department of Translational Medical Sciences - Section of Pediatrics, University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
7
|
Nayebi A, Navashenaq JG, Soleimani D, Nachvak SM. Probiotic supplementation: A prospective approach in the treatment of COVID-19. Nutr Health 2022; 28:163-175. [PMID: 34747257 PMCID: PMC9160438 DOI: 10.1177/02601060211049631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: Despite strategies based on social distancing, the coronavirus disease 2019 (COVID-19) expands globally, and so far, many attempts have been made to achieve effective treatment for patients with COVID-19. This disease infects the lower respiratory tract and may lead to severe acute respiratory syndrome coronavirus (SARS-CoV). COVID-19 also can cause gastrointestinal infections. Therefore, COVID-19 patients with gastrointestinal symptoms are more likely to be complicated by SARS-CoV. In this disease, acquired immune responses are impaired, and uncontrolled inflammatory responses result in cytokine storms, leading to acute lung injury and thrombus formation. Probiotics are living microorganisms that contribute to the health of the host if administered in appropriate doses. Aim: This study aimed to provide evidence to show the importance of gut dysbiosis in viral disease, especially COVID-19. Therefore, we have focused on the impact of probiotics consumption on preventing severe symptoms of the disease. Methods: We have entirely searched SCOPUS, PubMed, and Google Scholar databases to collect evidence regarding the relationship between probiotics and viral infections to expand this relationship to the COVID-19. Results: It has been shown that probiotics directly counteract SARS-CoV in the gastrointestinal and respiratory tracts. Moreover, probiotics suppress severe immune responses and prevent cytokine storms to inhibit pathologic inflammatory conditions in the body via modulation of immune responses. Conclusion: According to available evidence based on their antiviral and respiratory activities, using probiotics might be an adjuvant therapy to reduce the burden and severity of this disease.
Collapse
Affiliation(s)
- Atiyeh Nayebi
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Davood Soleimani
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyyed Mostafa Nachvak
- Student Research Committee, Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nutritional Sciences Department, School of Nutrition Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Sanklecha M, Verma L, Pai U, Mishra S, Maqsood S, Birla A. Lactobacillus rhamnosus GG Evaluation in Acute Diarrhea (LEAD): An Observational Study. Cureus 2022; 14:e24594. [PMID: 35506120 PMCID: PMC9053112 DOI: 10.7759/cureus.24594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 12/28/2022] Open
Abstract
Background and objective Probiotics with documented efficacy such as Lactobacillus rhamnosus GG(LGG) and Saccharomyces boulardii might be utilized as adjuncts to rehydration for the management of acute gastroenteritis (AGE) in children. In this study, we aimed to evaluate the potential role of LGG in acute diarrhea in the Indian pediatric population. Methods An observational, cross-sectional study [Lactobacillus rhamnosus GG Evaluation in Acute Diarrhea (LEAD)] was conducted among children aged one month to 12 years with acute watery diarrhea. In addition to standard management of diarrhea, LGG was given as an adjuvant treatment at the discretion of treating physicians based on their routine practice. Observations were documented on days one, three, and five. Outcomes such as frequency and duration of diarrhea, time to change in consistency of stools, Bristol Stool Chart (BSC) reading, and global assessment by healthcare practitioners (HCPs) and patients were also recorded. Results Of the 2,080 patients enrolled, 1,900 completed the five-day follow-up. There was marked improvement observed in the number of incidences of loose stools from day one (mean: 7.33) to day five (mean: 1.6). The mean time to improvement in stool consistency was 34 hours. The mean duration of diarrhea was 44.63 hours. A relatively shorter duration of diarrhea was reported among participants in this study. There was also a significant improvement in the number of vomiting episodes. Most patients and HCPs reported the product to be excellent/very good on the global assessment scale. No adverse effects were noted in any of the groups. Conclusion Based on our findings, LGG supplementation may be a beneficial adjuvant treatment in reducing the severity and duration of acute diarrheal episodes.
Collapse
Affiliation(s)
- Mukesh Sanklecha
- Department of Pediatrics, Bombay Hospital Institute of Medical Sciences, Mumbai, IND
| | - Lalit Verma
- Department of Pediatrics, SRCC Narayana Children's Hospital, Mumbai, IND
| | - Uday Pai
- Pediatrics, Sai Kutir Clinic, Mumbai, IND
| | - Suman Mishra
- Department of Pediatrics, Asian City Hospital and Sparsh Child Care, Patna, IND
| | - Sameer Maqsood
- Department of Pediatrics, Max Super Speciality Hospital, Delhi, IND
| | - Amita Birla
- Clinical Research, ACT Life Sciences Pvt Ltd., Navi Mumbai, IND
| |
Collapse
|
9
|
Sabahi S, Homayouni Rad A, Aghebati-Maleki L, Sangtarash N, Ozma MA, Karimi A, Hosseini H, Abbasi A. Postbiotics as the new frontier in food and pharmaceutical research. Crit Rev Food Sci Nutr 2022; 63:8375-8402. [PMID: 35348016 DOI: 10.1080/10408398.2022.2056727] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Food is the essential need of human life and has nutrients that support growth and health. Gastrointestinal tract microbiota involves valuable microorganisms that develop therapeutic effects and are characterized as probiotics. The investigations on appropriate probiotic strains have led to the characterization of specific metabolic byproducts of probiotics named postbiotics. The probiotics must maintain their survival against inappropriate lethal conditions of the processing, storage, distribution, preparation, and digestion system so that they can exhibit their most health effects. Conversely, probiotic metabolites (postbiotics) have successfully overcome these unfavorable conditions and may be an appropriate alternative to probiotics. Due to their specific chemical structure, safe profile, long shelf-life, and the fact that they contain various signaling molecules, postbiotics may have anti-inflammatory, immunomodulatory, antihypertensive properties, inhibiting abnormal cell proliferation and antioxidative activities. Consequently, present scientific literature approves that postbiotics can mimic the fundamental and clinical role of probiotics, and due to their unique characteristics, they can be applied in an oral delivery system (pharmaceutical/functional foods), as a preharvest food safety hurdle, to promote the shelf-life of food products and develop novel functional foods or/and for developing health benefits, and therapeutic aims. This review addresses the latest postbiotic applications with regard to pharmaceutical formulations and commercial food-based products. Potential postbiotic applications in the promotion of host health status, prevention of disease, and complementary treatment are also reviewed.
Collapse
Affiliation(s)
- Sahar Sabahi
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Sangtarash
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahdi Asghari Ozma
- Department of Medical Bacteriology and Virology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Karimi
- Department of Food Safety and Hygiene, School of Public Health, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hedayat Hosseini
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Abbasi
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Lacticaseibacillus rhamnosus: A Suitable Candidate for the Construction of Novel Bioengineered Probiotic Strains for Targeted Pathogen Control. Foods 2022; 11:foods11060785. [PMID: 35327208 PMCID: PMC8947445 DOI: 10.3390/foods11060785] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics, with their associated beneficial effects, have gained popularity for the control of foodborne pathogens. Various sources are explored with the intent to isolate novel robust probiotic strains with a broad range of health benefits due to, among other mechanisms, the production of an array of antimicrobial compounds. One of the shortcomings of these wild-type probiotics is their non-specificity. A pursuit to circumvent this limitation led to the advent of the field of pathobiotechnology. In this discipline, specific pathogen gene(s) are cloned and expressed into a given probiotic to yield a novel pathogen-specific strain. The resultant recombinant probiotic strain will exhibit enhanced species-specific inhibition of the pathogen and its associated infection. Such probiotics are also used as vehicles to deliver therapeutic agents. As fascinating as this approach is, coupled with the availability of numerous probiotics, it brings a challenge with regard to deciding which of the probiotics to use. Nonetheless, it is indisputable that an ideal candidate must fulfil the probiotic selection criteria. This review aims to show how Lacticaseibacillus rhamnosus, a clinically best-studied probiotic, presents as such a candidate. The objective is to spark researchers’ interest to conduct further probiotic-engineering studies using L. rhamnosus, with prospects for the successful development of novel probiotic strains with enhanced beneficial attributes.
Collapse
|
11
|
Supplementation of Bacillus sp. DU-106 Alleviates Antibiotic-Associated Diarrhea in Association with the Regulation of Intestinal Microbiota in Mice. Probiotics Antimicrob Proteins 2022; 14:372-383. [DOI: 10.1007/s12602-022-09906-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
|
12
|
Ansari F, Pashazadeh F, Nourollahi E, Hajebrahimi S, Munn Z, Pourjafar H. A Systematic Review and Meta-Analysis: The Effectiveness of Probiotics for Viral Gastroenteritis. Curr Pharm Biotechnol 2020; 21:1042-1051. [PMID: 32297578 DOI: 10.2174/1389201021666200416123931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/22/2020] [Accepted: 03/07/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND Probiotics can be used for the treatment of viral gastroenteritis. OBJECTIVE This systematic review is to evaluate the evidence regarding the effect of probiotics on human cases of viral gastroenteritis. METHODS The objective of this review is to evaluate the effectiveness of probiotics against placebo or standard treatment for viral gastroenteritis. A comprehensive search of Cochrane Library, EMBASE, MEDLINE via PubMed and Ovid databases, and unpublished studies (till 27 January 2018) was conducted followed by a process of study selection and critical appraisal by two independent reviewers. Randomized controlled trials assessing probiotic administration in human subjects infected with any species of gastroenteritis viruses were considered for inclusion. Only studies with a confirmed viral cause of infection were included. This study was developed using the JBI methodology for systematic reviews, which is in accordance with the PRISMA guideline. Meta-analysis was conducted where feasible. Data were pooled using the inverse variance method with random effects models and expressed as Mean Differences (MDs) with 95% Confidence Intervals (CIs). Heterogeneity was assessed by Cochran Q statistic and quantified by the I2 statistic. We included 17 RCTs, containing 3,082 patients. RESULTS Probiotics can improve symptoms of viral gastroenteritis, including the duration of diarrhea (mean difference 0.7 days, 95% CI 0.31 to 1.09 days, n = 740, ten trials) and duration of hospitalization (mean difference 0.76 days, 95% CI 0.61 to 0.92 days, n = 329, four trials). CONCLUSION The results of this review show that the administration of probiotics in patients with viral gastroenteritis should be considered.
Collapse
Affiliation(s)
- Fereshteh Ansari
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fariba Pashazadeh
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Nourollahi
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sakineh Hajebrahimi
- Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zachary Munn
- The Joanna Briggs Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Hadi Pourjafar
- Department of Food Sciences and Nutrition, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
13
|
Barbuti RC, Schiavon LL, Oliveira CP, Alvares-DA-Silva MR, Sassaki LY, Passos MDCF, Farias AQ, Barros LL, Barreto BP, Albuquerque GBDMLD, Alves AM, Navarro-Rodriguez T, Bittencourt PL. GUT MICROBIOTA, PREBIOTICS, PROBIOTICS, AND SYNBIOTICS IN GASTROINTESTINAL AND LIVER DISEASES: PROCEEDINGS OF A JOINT MEETING OF THE BRAZILIAN SOCIETY OF HEPATOLOGY (SBH), BRAZILIAN NUCLEUS FOR THE STUDY OF HELICOBACTER PYLORI AND MICROBIOTA (NBEHPM), AND BRAZILIAN FEDERATION OF GASTROENTEROLOGY (FBG). ARQUIVOS DE GASTROENTEROLOGIA 2020; 57:381-398. [PMID: 33331485 DOI: 10.1590/s0004-2803.202000000-72] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
Over the last years, there is growing evidence that microorganisms are involved in the maintenance of our health and are related to various diseases, both intestinal and extraintestinal. Changes in the gut microbiota appears to be a key element in the pathogenesis of hepatic and gastrointestinal disorders, including non-alcoholic fatty liver disease, alcoholic liver disease, liver cirrhosis, inflammatory bowel disease, irritable bowel syndrome, and Clostridium difficile - associated diarrhea. In 2019, the Brazilian Society of Hepatology (SBH) in cooperation with the Brazilian Nucleus for the Study of Helicobacter Pylori and Microbiota (NBEHPM), and Brazilian Federation of Gastroenterology (FBG) sponsored a joint meeting on gut microbiota and the use of prebiotics, probiotics, and synbiotics in gastrointestinal and liver diseases. This paper summarizes the proceedings of the aforementioned meeting. It is intended to provide practical information about this topic, addressing the latest discoveries and indicating areas for future studies.
Collapse
Affiliation(s)
- Ricardo Correa Barbuti
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Leonardo Lucca Schiavon
- Universidade Federal de Santa Catarina, Faculdade de Medicina, Departamento de Clínica Médica, Florianópolis, SC, Brasil
| | - Cláudia P Oliveira
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Mário Reis Alvares-DA-Silva
- Universidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Medicina Interna, Porto Alegre, RS, Brasil
| | | | | | - Alberto Queiroz Farias
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Luisa Leite Barros
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Bruno Paes Barreto
- Universidade do Estado do Pará, Centro de Ciências Biológicas e da Saúde, Belém, PA, Brasil
- Centro Universitário do Estado do Pará (CESUPA), Belém, PA, Brasil
| | | | - Amanda Mandarino Alves
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | - Tomás Navarro-Rodriguez
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Gastroenterologia, São Paulo, SP, Brasil
| | | |
Collapse
|
14
|
Mai TT, Thi Thu P, Thi Hang H, Trang TTT, Yui S, Shigehisa A, Tien VT, Dung TV, Nga PB, Hung NT, Tuyen LD. Efficacy of probiotics on digestive disorders and acute respiratory infections: a controlled clinical trial in young Vietnamese children. Eur J Clin Nutr 2020; 75:513-520. [PMID: 32981933 PMCID: PMC7943424 DOI: 10.1038/s41430-020-00754-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 08/04/2020] [Accepted: 09/05/2020] [Indexed: 11/30/2022]
Abstract
Objectives To evaluate the efficacy of fermented milk containing Lactobacillus casei strain Shirota (LcS) on the incidence of constipation, diarrhea, acute respiratory infections (ARI), and nutritional status of young Vietnamese children. Methods A controlled field trial was conducted with 1003 children (3–5 years old) in Thanh Hoa province in Vietnam. The probiotic group (n = 510) consumed fermented milk 65 mL/day containing 108 CFU/mL of LcS for the 12-week intervention period, whereas the control group (n = 493) was not given any. The incidence of constipation, diarrhea, ARI, and anthropometry in children was determined at baseline, after 4, 8, and 12-week intervention, and after the 4-week follow-up period. Results Probiotic drink decreased the incidence of constipation after the 12-week intervention period (12.0% vs. 32.0%, OR = 0.28 (95% CI: 0.21–0.40), p < 0.001), tended to decrease the incidence of diarrhea (4.9% vs. 7.9%, OR = 0.60 (95% CI: 0.35–1.01), p = 0.068), and prevented the occurrence of ARI (15.9% vs. 24.5%, OR = 0.58 (95% CI: 0.42–0.79), p < 0.001), when compared with the control group. In contrast, no probiotic effects were observed for the duration of diarrhea or ARI. Weight gain was higher in the probiotic group than in the control group after 4, 8, and 12-week intervention and after the 4-week follow-up period (p < 0.05). Conclusions Daily intake of fermented milk containing LcS strongly prevented the incidence of constipation and ARI in Vietnamese children. This study also revealed the potential effects of the use of a probiotic drink on diarrhea prevention as well as nutritional status improvement.
Collapse
Affiliation(s)
- Truong Tuyet Mai
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, Vietnam.
| | - Pham Thi Thu
- Haiphong University of Medicine and Pharmacy, 72 A, Nguyen Binh Khiem, Ngo Quyen, Haiphong, Vietnam
| | - Hoang Thi Hang
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, Vietnam
| | | | - Shintaro Yui
- Yakult Central Institute, 5-11 Izumi, Kunitachi, Tokyo, 186-8650, Japan
| | - Akira Shigehisa
- Yakult Honsha European Research Center for Microbiology ESV, Technologiepark 4, 9052, Zwijnaarde, Belgium
| | - Vu Thuy Tien
- Yakult Vietnam Co., Ltd., 195, Truong Van Bang Street, Thanh My Loi Ward District 2, Ho Chi Minh, Vietnam
| | - Truong Viet Dung
- Hanoi Medical University, 1, Ton That Tung, Dong Da, Hanoi, Vietnam
| | - Phan Bich Nga
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, Vietnam
| | - Nguyen Trong Hung
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, Vietnam
| | - Le Danh Tuyen
- National Institute of Nutrition, 48B Tang Bat Ho Street, Hanoi, Vietnam
| |
Collapse
|
15
|
The effects of probiotics on reducing the colorectal cancer surgery complications: A periodic review during 2007–2017. Clin Nutr 2020; 39:2358-2367. [PMID: 31831184 DOI: 10.1016/j.clnu.2019.11.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 10/09/2019] [Accepted: 11/04/2019] [Indexed: 02/01/2023]
|
16
|
Szajewska H, Hojsak I. Health benefits of Lactobacillus rhamnosus GG and Bifidobacterium animalis subspecies lactis BB-12 in children. Postgrad Med 2020; 132:441-451. [DOI: 10.1080/00325481.2020.1731214] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Iva Hojsak
- Referral Center for Pediatric Gastroenterology and Nutrition, Children’s Hospital Zagreb, Zagreb, Croatia
- Department of Pediatrics, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Pediatrics, School of Medicine, University J.J. Strossmayer, Osijek, Croatia
| |
Collapse
|
17
|
Picó-Monllor JA, Mingot-Ascencao JM. Search and Selection of Probiotics That Improve Mucositis Symptoms in Oncologic Patients. A Systematic Review. Nutrients 2019; 11:E2322. [PMID: 31581434 PMCID: PMC6835542 DOI: 10.3390/nu11102322] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/17/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
Mucositis is a common and severe adverse effect of radiotherapy and/or chemotherapy treatments applied to oncologic patients. The development of effective therapies and adjuvant treatments to increase their efficacy and reduce adverse effect is a priority in cancer therapy. Probiotics are non-pathogenic live microorganisms that when ingested in adequate amounts can colonize the intestinal tract promoting the restoration of a healthy gut microbiota and contributing to all its functions including the maintenance of the integrity of the mucosa and the modulation of the immune system. In order to check the possible efficacy and safety of these microorganisms to prevent or ameliorate mucositis' symptoms, we have systematically searched the bibliographic databases MEDLINE (via Pubmed), EMBASE, The Cochrane library, Scopus, Web of science, and Latin American and Caribbean Literature in Health of Sciences (LILACS) using the descriptors "Mucositis", "Probiotics", "Neoplasms", "Humans", and "Clinical Trials". After applying our inclusion and exclusion criteria, 15 studies were accepted for review and critical analysis. Our analysis suggests that a combination of Bifidobacterium longum, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium infantis, and Saccharomyces boulardii could be a good combination of probiotics to reduce incident rates of mucositis or ameliorate its symptoms in chemo or radiotherapy treated patients.
Collapse
|
18
|
Li YT, Xu H, Ye JZ, Wu WR, Shi D, Fang DQ, Liu Y, Li LJ. Efficacy of Lactobacillus rhamnosus GG in treatment of acute pediatric diarrhea: A systematic review with meta-analysis. World J Gastroenterol 2019; 25:4999-5016. [PMID: 31543689 PMCID: PMC6737314 DOI: 10.3748/wjg.v25.i33.4999] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 07/04/2019] [Accepted: 07/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diarrhea is a major infectious cause of childhood morbidity and mortality worldwide. In clinical trials, Lactobacillus rhamnosus GG ATCC 53013 (LGG) has been used to treat diarrhea. However, recent randomized controlled trials (RCTs) found no evidence of a beneficial effect of LGG treatment.
AIM To evaluate the efficacy of LGG in treating acute diarrhea in children.
METHODS The EMBASE, MEDLINE, PubMed, Web of Science databases, and the Cochrane Central Register of Controlled Trials were searched up to April 2019 for meta-analyses and RCTs. The Cochrane Review Manager was used to analyze the relevant data.
RESULTS Nineteen RCTs met the inclusion criteria and showed that compared with the control group, LGG administration notably reduced the diarrhea duration [mean difference (MD) -24.02 h, 95% confidence interval (CI) (-36.58, -11.45)]. More effective results were detected at a high dose ≥ 1010 CFU per day [MD -22.56 h, 95%CI (-36.41, -8.72)] vs a lower dose. A similar reduction was found in Asian and European patients [MD -24.42 h, 95%CI (-47.01, -1.82); MD -32.02 h, 95%CI (-49.26, -14.79), respectively]. A reduced duration of diarrhea was confirmed in LGG participants with diarrhea for less than 3 d at enrollment [MD -15.83 h, 95%CI (-20.68, -10.98)]. High-dose LGG effectively reduced the duration of rotavirus-induced diarrhea [MD -31.05 h, 95%CI (-50.31, -11.80)] and the stool number per day [MD -1.08, 95%CI (-1.87, -0.28)].
CONCLUSION High-dose LGG therapy reduces the duration of diarrhea and the stool number per day. Intervention at the early stage is recommended. Future trials are expected to verify the effectiveness of LGG treatment.
Collapse
Affiliation(s)
- Ya-Ting Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Hong Xu
- Department of Orthopedics, Xiaoshan Traditional Chinese Medical Hospital, Hangzhou 310003, Zhejiang Province, China
| | - Jian-Zhong Ye
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Wen-Rui Wu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Ding Shi
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Dai-Qiong Fang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Yang Liu
- Department of Orthopedics, Clinical Sciences, Lund, Lund University, Lund 22185, Sweden
| | - Lan-Juan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
- Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
19
|
Wang G, Feng D. Therapeutic effect of Saccharomyces boulardii combined with Bifidobacterium and on cellular immune function in children with acute diarrhea. Exp Ther Med 2019; 18:2653-2659. [PMID: 31572514 PMCID: PMC6755444 DOI: 10.3892/etm.2019.7836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 07/23/2019] [Indexed: 12/30/2022] Open
Abstract
Clinical effect of Saccharomyces boulardii combined with bifidobacterium and its effect on cellular immune function in children with acute diarrhea were studied. In total 116 cases of children with acute diarrhea admitted to Xuzhou Children's Hospital from March 2015 to March 2017 were collected and analyzed retrospectively. There were 59 children treated with Saccharomyces boulardii as control group and 57 children treated with Saccharomyces boulardii combined with bifidobacterium as experimental group. The clinical effect, stool frequency in different time periods, mean antidiarrheal time, mean antipyretic time and length of stay, and immune function of children in the two groups after treatment were analyzed. The cure rate (73.68%) and the total effective rate (87.72%) in the experimental group were significantly higher than those in the control group (47.46 and 71.19%) (P<0.05). The stool frequency in the experimental group was significantly lower than that in the control group 3 days after treatment (P<0.05). The mean antidiarrheal time in the experimental group was significantly shorter than that in the control group (P<0.05). The length of stay in the control group was significantly longer than that in the experimental group (P<0.05). CD3+, CD4+ and CD4+/CD8+ increased significantly in the experimental group after treatment while CD8+ decreased significantly (P<0.05). After treatment, the ratio of Th1 and Th2 in the two groups decreased significantly compared with before treatment (P<0.05), and the experimental group was significantly lower than the control group (P<0.05). After treatment, Th1/Th2 ratio was significantly higher than that before treatment (P<0.05), and the experimental group was significantly higher than the control group (P<0.05). In conclusion, treatment of acute diarrhea in children with Saccharomyces boulardii combined with bifidobacterium can effectively shorten the duration of diarrhea and hospital stay, reduce the number of diarrhea and enhance the cellular immune function.
Collapse
Affiliation(s)
- Guangmeng Wang
- Department of Gastroenterology, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Dongjin Feng
- Department of Gastroenterology, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
20
|
Hou Y, Li X, Liu X, Zhang Y, Zhang W, Man C, Jiang Y. Transcriptomic responses of Caco-2 cells to Lactobacillus rhamnosus GG and Lactobacillus plantarum J26 against oxidative stress. J Dairy Sci 2019; 102:7684-7696. [PMID: 31255276 DOI: 10.3168/jds.2019-16332] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/20/2019] [Indexed: 12/12/2022]
Abstract
Oxidative stress is the basic reason for aging and age-related diseases. In this study, we investigated the protective effect of 2 strains of lactic acid bacteria (LAB), Lactobacillus rhamnosus GG and L. plantarum J26, against oxidative stress in Caco-2 cells, and gave an overview of the mechanisms of lactic acid bacteria antioxidant activity using digital gene expression profiling. The 2 LAB strains provided significant protection against hydrogen peroxide (H2O2)-induced reduction in superoxide dismutase activity and increase in glutathione peroxidase activity in Caco-2 cells. However, inactive bacteria had little effect on alleviating oxidation stress in Caco-2 cells. Eight genes related to oxidative stress-FOSB, TNF, PPP1R15A, NUAK2, ATF3, TNFAIP3, EGR2, and FBN2-were significantly upregulated in H2O2-induced Caco-2 cells compared with untreated Caco-2 cells. After incubation of the H2O2-induced Caco-2 cells with L. rhamnosus GG and L. plantarum J26, 5 genes (TNF, EGR2, NUAK2, FBN2, and TNFAIP3) and 2 genes (NUAK2 and FBN2) were downregulated, respectively. In addition, the Kyoto Encyclopedia of Genes and Genomes indicated that some signaling pathways associated with inflammation, immune response, and apoptosis, such as Janus kinase/signal transducers and activators of transcription (Jak-STAT), mitogen-activated protein kinase (MAPK), nuclear factor-κB, and tumor necrosis factor, were all negatively modulated by the 2 strains, especially L. rhamnosus GG. In this paper, we reveal the mechanism of LAB in relieving oxidative stress and provide a theoretical basis for the rapid screening and evaluation of new LAB resources.
Collapse
Affiliation(s)
- Yichao Hou
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030
| | - Xuesong Li
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030
| | - Xinyu Liu
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030
| | - Yashuo Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Department of Food Science, Northeast Agricultural University, Harbin, China, 150030.
| |
Collapse
|
21
|
Abstract
Lactobacillus rhamnosus GG (LGG) was the first strain belonging to the genus Lactobacillus to be patented in 1989 thanks to its ability to survive and to proliferate at gastric acid pH and in medium containing bile, and to adhere to enterocytes. Furthermore LGG is able to produces both a biofilm that can mechanically protect the mucosa, and different soluble factors beneficial to the gut by enhancing intestinal crypt survival, diminishing apoptosis of the intestinal epithelium, and preserving cytoskeletal integrity. Moreover LGG thanks to its lectin-like protein 1 and 2 inhibits some pathogens such as Salmonella species. Finally LGG is able to promote type 1 immune-responsiveness by reducing the expression of several activation and inflammation markers on monocytes and by increasing the production of interleukin-10, interleukin-12 and tumor necrosis factor-α in macrophages. A large number of research data on Lactobacillus GG is the basis for the use of this probiotic for human health. In this review we have considered predominantly randomized controlled trials, meta-analysis, Cochrane Review, guide lines of Scientific Societies and anyway studies whose results were evaluated by means of relative risk, odds ratio, weighted mean difference 95% confidence interval. The effectiveness of LGG in gastrointestinal infections and diarrhea, antibiotic and Clostridium difficile associated diarrhea, irritable bowel syndrome, inflammatory bowel disease, respiratory tract infections, allergy, cardiovascular diseases, nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, cystic fibrosis, cancer, elderly end sport were analyzed.
Collapse
|
22
|
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
|
23
|
Davoren MJ, Liu J, Castellanos J, Rodríguez-Malavé NI, Schiestl RH. A novel probiotic, Lactobacillus johnsonii 456, resists acid and can persist in the human gut beyond the initial ingestion period. Gut Microbes 2018; 10:458-480. [PMID: 30580660 PMCID: PMC6748577 DOI: 10.1080/19490976.2018.1547612] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 10/14/2018] [Accepted: 11/06/2018] [Indexed: 02/03/2023] Open
Abstract
Probiotics are considered to have multiple beneficial effects on the human gastrointestinal tract, including immunomodulation, pathogen inhibition, and improved host nutrient metabolism. However, extensive characterization of these properties is needed to define suitable clinical applications for probiotic candidates. Lactobacillus johnsonii 456 (LBJ 456) was previously demonstrated to have anti-inflammatory and anti-genotoxic effects in a mouse model. Here, we characterize its resistance to gastric and bile acids as well as its ability to inhibit gut pathogens and adhere to host mucosa. While bile resistance and in vitro host attachment properties of LBJ 456 were comparable to other tested probiotics, LBJ 456 maintained higher viability at lower pH conditions compared to other tested strains. LBJ 456 also altered pathogen adhesion to LS 174T monolayers and demonstrated contact-dependent and independent inhibition of pathogen growth. Genome analyses further revealed possible genetic elements involved in host attachment and pathogen inhibition. Importantly, we show that ingestion of Lactobacillus johnsonii 456 over a one week yogurt course leads to persistent viable bacteria detectable even beyond the period of initial ingestion, unlike many other previously described probiotic species of lactic acid bacteria.
Collapse
Affiliation(s)
- Michael J. Davoren
- Molecular Toxicology Interdepartmental Program, Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | - Jared Liu
- School of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jocelyn Castellanos
- Molecular Toxicology Interdepartmental Program, Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Robert H. Schiestl
- Molecular Toxicology Interdepartmental Program, Environmental Health Sciences, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
24
|
Radicioni M, Koirala R, Fiore W, Leuratti C, Guglielmetti S, Arioli S. Survival of L. casei DG ® (Lactobacillus paracasei CNCMI1572) in the gastrointestinal tract of a healthy paediatric population. Eur J Nutr 2018; 58:3161-3170. [PMID: 30498868 PMCID: PMC6842349 DOI: 10.1007/s00394-018-1860-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/14/2018] [Indexed: 01/17/2023]
Abstract
Purpose Ability to survive the digestive process is a major factor in determining the effectiveness of a probiotic. In this study, the ability of the probiotic L. casei DG® (Lactobacillus paracasei CNCMI1572) to survive gastrointestinal transit in healthy children was investigated for the first time. Methods Twenty children aged 3–12 years received L. casei DG® as drinkable solution of 1 × 109 colony forming units (CFU), once daily for 7 consecutive days. Recovery in faecal samples was evaluated at baseline and at different time-points during and after administration. Defecation frequency, faeces consistency, digestive function and product safety were also assessed. Results Nineteen (95%) of the 20 enrolled children presented viable L. casei DG® cells in their faeces at least once during the study, with a maximum count (mean 4.3 log10 CFU/g ± 2.3) reached between day 4 and 6 from the beginning of consumption. Notably, for 11 (57.9%) of the 19 children with viable cells, L. casei DG® survived in faecal samples up to 3 days after treatment end. Defecation frequency, faeces consistency and digestive function did not change considerably during or after study treatment. Safety of the study product was very good. Conclusions This study showed for the first time that L. casei DG® survives the gastrointestinal transit when ingested by children with a paediatric probiotic drinkable solution containing 1 × 109 CFU, and persists in the gut up to 3 days after the end of product intake, demonstrating resistance to gastric juices, hydrolytic enzymes and bile acids. Electronic supplementary material The online version of this article (10.1007/s00394-018-1860-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Milko Radicioni
- CROSS Research S.A., via F.A. Giorgioli 14, 6864, Arzo, Switzerland.
| | - Ranjan Koirala
- Department of Food, Environmental and Nutritional Science (DeFENS), University of Milan, Milan, Italy
| | - Walter Fiore
- SOFAR SpA, 20060, Milan, Trezzano Rosa, Italy.,Department of Food, Environmental and Nutritional Science (DeFENS), University of Milan, Milan, Italy
| | - Chiara Leuratti
- CROSS Research S.A., via F.A. Giorgioli 14, 6864, Arzo, Switzerland
| | - Simone Guglielmetti
- Department of Food, Environmental and Nutritional Science (DeFENS), University of Milan, Milan, Italy
| | - Stefania Arioli
- Department of Food, Environmental and Nutritional Science (DeFENS), University of Milan, Milan, Italy.
| |
Collapse
|
25
|
Westerik N, Reid G, Sybesma W, Kort R. The Probiotic Lactobacillus rhamnosus for Alleviation of Helicobacter pylori-Associated Gastric Pathology in East Africa. Front Microbiol 2018; 9:1873. [PMID: 30154777 PMCID: PMC6102400 DOI: 10.3389/fmicb.2018.01873] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/25/2018] [Indexed: 12/16/2022] Open
Abstract
The probiotic Lactobacillus rhamnosus GG (LGG) can play a role in establishing a harmless relationship with Helicobacter pylori and reduce gastric pathology in East African populations. H. pylori has the ability to inhabit the surface of the mucous layer of the human stomach and duodenum. In the developing world, an estimated 51% of the population is carrier of H. pylori, while in some Western countries these numbers dropped below 20%, which is probably associated with improved sanitation and smaller family sizes. Colonization by H. pylori can be followed by inflammation of the gastric mucus layer, and is a risk factor in the development of atrophic gastritis, peptic ulcers and gastric cancer. Notwithstanding the higher prevalence of H. pylori carriers in developing countries, no equal overall increase in gastric pathology is found. This has been attributed to a less pro-inflammatory immune response to H. pylori in African compared to Caucasian populations. In addition, a relatively low exposure to other risk factors in certain African populations may play a role, including the use of non-steroidal anti-inflammatory drugs, smoking, and diets without certain protective factors. A novel approach to the reduction of H. pylori associated gastric pathology is found in the administration of the probiotic bacterium Lactobacillus rhamnosus yoba 2012 (LRY), the generic variant of LGG. This gastro-intestinal isolate inhibits H. pylori by competition for substrate and binding sites as well as production of antimicrobial compounds such as lactic acid. In addition, it attenuates the host's H. pylori-induced apoptosis and inflammation responses and stimulates angiogenesis in the gastric and duodenal epithelium. The probiotic LRY is not able to eradicate H. pylori completely, but its co-supplementation in antibiotic eradication therapy has been shown to relieve side effects of this therapy. In Uganda, unlike other African countries, gastric pathology is relatively common, presumably resulting from the lack of dietary protective factors in the traditional diet. Supplementation with LRY through local production of probiotic yogurt, could be a solution to establish a harmless relationship with H. pylori and reduce gastric pathology and subsequent eradication therapy treatment.
Collapse
Affiliation(s)
- Nieke Westerik
- Department of Molecular Cell Biology, VU University Amsterdam, Amsterdam, Netherlands
- Yoba for Life Foundation, Amsterdam, Netherlands
| | - Gregor Reid
- Canadian R&D Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON, Canada
- Department of Microbiology and Immunology, and Surgery, Western University, London, ON, Canada
| | | | - Remco Kort
- Department of Molecular Cell Biology, VU University Amsterdam, Amsterdam, Netherlands
- Yoba for Life Foundation, Amsterdam, Netherlands
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research, Zeist, Netherlands
- ARTIS-Micropia, Amsterdam, Netherlands
| |
Collapse
|
26
|
Putta S, Yarla NS, Lakkappa DB, Imandi SB, Malla RR, Chaitanya AK, Chari BP, Saka S, Vechalapu RR, Kamal MA, Tarasov VV, Chubarev VN, Siva Kumar K, Aliev G. Probiotics: Supplements, Food, Pharmaceutical Industry. THERAPEUTIC, PROBIOTIC, AND UNCONVENTIONAL FOODS 2018:15-25. [DOI: 10.1016/b978-0-12-814625-5.00002-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
27
|
Barraclough H. Question 2 Probiotics in acute infectious diarrhoea: should we run with it? Arch Dis Child 2017; 102:782-785. [PMID: 28724708 DOI: 10.1136/archdischild-2017-313242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 11/03/2022]
|
28
|
Yang GY, Yu J, Su JH, Jiao LG, Liu X, Zhu YH. Oral Administration of Lactobacillus rhamnosus GG Ameliorates Salmonella Infantis-Induced Inflammation in a Pig Model via Activation of the IL-22BP/IL-22/STAT3 Pathway. Front Cell Infect Microbiol 2017; 7:323. [PMID: 28770173 PMCID: PMC5514694 DOI: 10.3389/fcimb.2017.00323] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/30/2017] [Indexed: 01/06/2023] Open
Abstract
The high rate of Salmonella enterica serovar Infantis (S. Infantis) infection poses significant risk for the development of non-typhoidal Salmonella gastroenteritis. However, efficient strategies to prevent or treat the infection remain elusive. Here, we explored the effect of the probiotic Lactobacillus rhamnosus GG (LGG) administration in preventing S. Infantis infection in a pig model. Probiotic LGG (1.0 × 1010 CFU/day) was orally administered to newly weaned piglets for 1 week before S. Infantis challenge. LGG pretreatment reduced the severity of diarrhea and alleviated intestinal inflammation caused by S. Infantis. Pre-administration of LGG excluded Salmonella from colonization of the jejunal mucosa but increased the abundance of Bifidobacterium in the feces. LGG promoted the expansion of CD4+ T-bet+ IFNγ+ T cells but attenuated S. Infantis-induced increases in the percentage of CD4+ IFNγ+ T cells and serum interleukin (IL)-22 levels in peripheral blood after S. Infantis challenge. In the small intestine, LGG pretreatment upregulated expression of the transcription factor T-bet but downregulated the S. Infantis-induced increase of CD4+ IFNγ+ T cells in Peyer's patches and IL-7Rα expression in the jejunum. Notably, LGG-treated pigs had enhanced expression of IL-22 and activated STAT3 in the ileum in response to S. Infantis infection. Pretreatment of pigs with LGG also elevated intestinal IL-22-binding protein production in response to S. Infantis challenge. In contrast, LGG consumption reduced the S. Infantis-induced increase in the number of CCL20-expressing cells in the jejunum. Our results suggest that the mechanism by which LGG ameliorates the intestinal inflammation caused by S. Infantis involves the upregulation of T-bet, activation of STAT3, and downregulation of CCL20.
Collapse
Affiliation(s)
| | | | | | | | | | - Yao-Hong Zhu
- College of Veterinary Medicine, China Agricultural UniversityBeijing, China
| |
Collapse
|
29
|
Suzuki T, Masui A, Nakamura J, Shiozawa H, Aoki J, Nakae H, Tsuda S, Imai J, Hideki O, Matsushima M, Mine T, Tamura A, Ohtsu T, Asami Y, Takagi A. Yogurt Containing Lactobacillus gasseri Mitigates Aspirin-Induced Small Bowel Injuries: A Prospective, Randomized, Double-Blind, Placebo-Controlled Trial. Digestion 2017; 95:49-54. [PMID: 28052291 DOI: 10.1159/000452361] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Although there is evidence about the beneficial effects of probiotics, their effects on aspirin-induced small bowel injuries have not been well examined. We evaluated the effects of the probiotic Lactobacillus gasseri OLL2716 (LG) on aspirin-induced small intestinal lesions, such as ulcers, erosions, reddened lesions, and bleeding. SUMMARY This study enrolled 64 patients who received aspirin for more than 1 month and provided written informed consent to be part of the study. The patients received 112 ml of yogurt containing LG or placebo twice daily for 6 weeks. Small bowel injuries were evaluated by capsule endoscopy before and after consuming the yogurt. The effect of LG on patient symptoms was also assessed using the Frequency Scale for the Symptoms of Gastroesophageal Reflux Disease (FSSG) and Gastrointestinal Symptom Rating Scale (GSRS) questionnaires before and after 6 weeks of treatment. There was no significant difference in any baseline characteristics and the number of small bowel mucosal breaks between the 2 groups. In contrast with the placebo group, the LG group had significantly fewer small bowel mucosal breaks and reddened lesions after 6 weeks (p < 0.01). The FSSG and GSRS scores were also significantly improved in the LG group but not in the placebo group. Key Messages: This double-blind, placebo-controlled study found that LG may be useful in reducing aspirin-induced small bowel injuries and in mitigating gastrointestinal symptoms.
Collapse
Affiliation(s)
- Takayoshi Suzuki
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University Tokyo Hospital, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Pinto JM, Petrova A. Lactobacillus acidophilus Mixture in Treatment of Children Hospitalized With Acute Diarrhea. Clin Pediatr (Phila) 2016; 55:1202-1209. [PMID: 26581358 DOI: 10.1177/0009922815616075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Despite unproven effectiveness, Lactobacillus acidophilus is a widely used probiotic in the treatment of pediatric diarrhea. In this report, we evaluated the association between length of stay (LOS) for 290 young children hospitalized with acute diarrhea and adjuvant therapy with a probiotic mixture containing 80% L acidophilus that was included in treatment for 22.4% of them. Overall, no association between LOS and use of L acidophilus was recorded after controlling for age, length of diarrhea symptoms, duration of intravenous fluids, and prior exposure to antibiotic. However, LOS was directly associated with use of L acidophilus in children with negative stool studies, and no such association was recorded in children with positive stool for rotavirus or other infections. We concluded that adjuvant therapy with L acidophilus mixture is not beneficial for young children hospitalized with acute diarrhea.
Collapse
Affiliation(s)
- Jamie M Pinto
- Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Anna Petrova
- Jersey Shore University Medical Center, Neptune, NJ, USA Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
31
|
Prodeus A, Niborski V, Schrezenmeir J, Gorelov A, Shcherbina A, Rumyantsev A. Fermented Milk Consumption and Common Infections in Children Attending Day-Care Centers: A Randomized Trial. J Pediatr Gastroenterol Nutr 2016; 63:534-543. [PMID: 27168455 PMCID: PMC5084641 DOI: 10.1097/mpg.0000000000001248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 04/14/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This multicenter, double-blind, randomized, placebo-controlled clinical trial investigated the effect of a fermented milk product containing the Lactobacillus casei National Collection of Microorganisms and Cell Cultures (CNCM) I-1518 strain on respiratory and gastrointestinal common infectious diseases (CIDs) in children attending day-care centers in Russia. METHODS Children ages 3 to 6 years received 100 g of a fermented milk product (n = 300) or a control product (n = 299) twice daily for 3 months, followed by a 1-month observation period. The primary outcome was the incidence of CIDs during the product consumption period. RESULTS There was no significant difference in the incidence of CIDs between the groups (N = 98 with fermented milk product vs N = 93 with control product). The overall number of CIDs (and no severe cases at all) in both study groups and in all 12 centers, however, was unexpectedly low resulting in underpowering of the study. No differences were found between the groups in the duration or severity of disease, duration of sick leave from day-care centers, parental missed working days, or in quality-of-life dimensions on the PedsQL questionnaire (P > 0.05).There was, however, a significantly lower incidence of the most frequently observed CID, rhinopharyngitis, in children consuming the fermented milk product compared with those consuming the control product (N = 81 vs N = 100, relative risk 0.82, 95% confidence interval 0.69-0.96, P = 0.017) when considering the entire study period. CONCLUSIONS Although no other significant differences were shown between the fermented milk and control product groups in this study, lower incidence of rhinopharyngitis may indicate a beneficial effect of this fermented milk product.
Collapse
Affiliation(s)
- Andrey Prodeus
- Children's Clinical Hospital No. 9 named after G.N. Speransky of Moscow Healthcare Department, Moscow, Russia
| | - Violeta Niborski
- Danone Nutricia Research, Centre de Recherche Daniel Carasso, Palaiseau, France
| | | | - Alexander Gorelov
- Federal Budget Institution of Science “Central Research Institute of Epidemiology” of The Federal Service on Customers’ Rights Protection and Human Well-being Surveillance
| | - Anna Shcherbina
- Federal Government Budget Institution “Federal Scientific Clinical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev” of the Ministry of Health of Russian Federation, Moscow, Russia
| | - Alexander Rumyantsev
- Federal Government Budget Institution “Federal Scientific Clinical Center of Pediatric Hematology, Oncology and Immunology named after Dmitry Rogachev” of the Ministry of Health of Russian Federation, Moscow, Russia
| |
Collapse
|
32
|
Westerik N, Wacoo AP, Sybesma W, Kort R. Novel Production Protocol for Small-scale Manufacture of Probiotic Fermented Foods. J Vis Exp 2016:54365. [PMID: 27684196 PMCID: PMC5091997 DOI: 10.3791/54365] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A novel dried bacterial consortium of Lactobacillus rhamnosus yoba 2012 and Streptococcus thermophilus C106 is cultured in 1 L of milk. This fresh starter can be used for the production of fermented milk and other fermented foods either at home or at small-scale in rural settings. For the fresh starter, 1 L of milk is pasteurized in a pan that fits into a larger pan containing water, placed on a source of heat. In this water bath, the milk is heated and incubated at 85 °C for 30 min. Thereafter, the milk is cooled down to 45 °C, transferred to a vacuum flask, inoculated with the dried bacteria and left for at least 16 hr between 30 °C and 45 °C. For the purpose of frequent home production, the fresh starter is frozen into ice cubes, which can be used for the production of small volumes of up to 2 L of fermented milk. For the purpose of small-scale production in resource-poor countries, pasteurization of up to 100 L of milk is conducted in milk cans that are placed in a large sauce pan filled with water and heated on a fire at 85 °C for 30 min, and subsequently cooled to 45 °C. Next, the 100 L batch is inoculated with the 1 L freshly prepared starter mentioned before. To assure an effective fermentation at a temperature between 30 and 45 °C, the milk can is covered with a blanket for 12 hr. For the production of non-dairy fermented foods, the fresh starter is left in a cheese cloth for 12 hr, and the drained-off whey can be subsequently used for the inoculation of a wide range of food raw materials, including vegetables and cereal-based foods.
Collapse
Affiliation(s)
- Nieke Westerik
- Yoba for Life Foundation; Department of Molecular Cell Biology, Vrije Universiteit Amsterdam (VUA)
| | - Alex Paul Wacoo
- Yoba for Life Foundation; Department of Molecular Cell Biology, Vrije Universiteit Amsterdam (VUA); Uganda Industrial Research Institute (UIRI)
| | | | - Remco Kort
- Yoba for Life Foundation; Department of Molecular Cell Biology, Vrije Universiteit Amsterdam (VUA); Micropia, Natura Artis Magistra; Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO);
| |
Collapse
|
33
|
Li C, Li X, Han H, Cui H, Peng M, Wang G, Wang Z. Effect of probiotics on metabolic profiles in type 2 diabetes mellitus: A meta-analysis of randomized, controlled trials. Medicine (Baltimore) 2016; 95:e4088. [PMID: 27368052 PMCID: PMC4937966 DOI: 10.1097/md.0000000000004088] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a prevalent metabolic disease which is imposing heavy burden on global health and economy. Recent studies indicate gut microbiota play important role on the pathogenesis and metabolic disturbance of T2DM. As an effective mean of regulating gut microbiota, probiotics are live micro-organisms that are believed to provide a specific health benefit on the host. Whether probiotic supplementation could improve metabolic profiles by modifying gut microbiota in T2DM or not is still in controversy.The aim of the study is to assess the effect of probiotic supplementation on metabolic profiles in T2DM.We searched PubMed, EMBASE, and Cochrane Library up to 12 April 2016. Two review authors independently assessed study eligibility, extracted data, and evaluated risk of bias of included studies. Data were pooled by using the random-effect model and expressed as standardized mean difference (SMD) with 95% confidence interval (CI). Heterogeneity was assessed and quantified (I).A total of 12 randomized controlled trials (RCTs) were included. Lipid profiles (n = 508) and fasting blood glucose (FBG) (n = 520) were reported in 9 trials; the homeostasis model of assessment for insulin resistance index (HOMA-IR) (n = 368) and glycosylated hemoglobin (HbA1c) (n = 380) were reported in 6 trials. Probiotics could alleviate FBG (SMD -0.61 mmol/L, 95% CI [-0.92, -0.30], P = 0.0001). Probiotics could increase high-density lipoprotein-cholesterol (HDL-C) (SMD 0.42 mmol/L, 95% CI [0.08, 0.76], P = 0.01). There were no significant differences in low-density lipoprotein-cholesterol (LDL-C), total cholesterol (TC), triglyceride (TG), HbA1c and HOMA-IR between the treatment group and the control group.Probiotics may improve glycemic control and lipid metabolism in T2DM. Application of probiotic agents might become a new method for glucose management in T2DM.
Collapse
Affiliation(s)
| | - Xin Li
- Department of Cardiothoracic Surgery
| | - Hongqiu Han
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Correspondence: Zhiqiang Wang, Hongqiu Han, Heping District, Tianjin, China (e-mail: ; )
| | | | | | | | - Zhiqiang Wang
- Intensive Care Unit
- Correspondence: Zhiqiang Wang, Hongqiu Han, Heping District, Tianjin, China (e-mail: ; )
| |
Collapse
|
34
|
Anti-infective activities of lactobacillus strains in the human intestinal microbiota: from probiotics to gastrointestinal anti-infectious biotherapeutic agents. Clin Microbiol Rev 2016; 27:167-99. [PMID: 24696432 DOI: 10.1128/cmr.00080-13] [Citation(s) in RCA: 228] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A vast and diverse array of microbial species displaying great phylogenic, genomic, and metabolic diversity have colonized the gastrointestinal tract. Resident microbes play a beneficial role by regulating the intestinal immune system, stimulating the maturation of host tissues, and playing a variety of roles in nutrition and in host resistance to gastric and enteric bacterial pathogens. The mechanisms by which the resident microbial species combat gastrointestinal pathogens are complex and include competitive metabolic interactions and the production of antimicrobial molecules. The human intestinal microbiota is a source from which Lactobacillus probiotic strains have often been isolated. Only six probiotic Lactobacillus strains isolated from human intestinal microbiota, i.e., L. rhamnosus GG, L. casei Shirota YIT9029, L. casei DN-114 001, L. johnsonii NCC 533, L. acidophilus LB, and L. reuteri DSM 17938, have been well characterized with regard to their potential antimicrobial effects against the major gastric and enteric bacterial pathogens and rotavirus. In this review, we describe the current knowledge concerning the experimental antibacterial activities, including antibiotic-like and cell-regulating activities, and therapeutic effects demonstrated in well-conducted, placebo-controlled, randomized clinical trials of these probiotic Lactobacillus strains. What is known about the antimicrobial activities supported by the molecules secreted by such probiotic Lactobacillus strains suggests that they constitute a promising new source for the development of innovative anti-infectious agents that act luminally and intracellularly in the gastrointestinal tract.
Collapse
|
35
|
Zhang Q, Wu Y, Fei X. Effect of probiotics on body weight and body-mass index: a systematic review and meta-analysis of randomized, controlled trials. Int J Food Sci Nutr 2016; 67:571-80. [DOI: 10.1080/09637486.2016.1181156] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
36
|
Tamang JP, Shin DH, Jung SJ, Chae SW. Functional Properties of Microorganisms in Fermented Foods. Front Microbiol 2016; 7:578. [PMID: 27199913 PMCID: PMC4844621 DOI: 10.3389/fmicb.2016.00578] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Accepted: 04/08/2016] [Indexed: 12/25/2022] Open
Abstract
Fermented foods have unique functional properties imparting some health benefits to consumers due to presence of functional microorganisms, which possess probiotics properties, antimicrobial, antioxidant, peptide production, etc. Health benefits of some global fermented foods are synthesis of nutrients, prevention of cardiovascular disease, prevention of cancer, gastrointestinal disorders, allergic reactions, diabetes, among others. The present paper is aimed to review the information on some functional properties of the microorganisms associated with fermented foods and beverages, and their health-promoting benefits to consumers.
Collapse
Affiliation(s)
- Jyoti P. Tamang
- Department of Microbiology, School of Life Sciences, Sikkim UniversityGangtok, India
| | - Dong-Hwa Shin
- Shindonghwa Food Research InstituteJeonju, South Korea
- Clinical Trial Center for Functional Foods, Chonbuk National University HospitalJeonju, South Korea
| | - Su-Jin Jung
- Clinical Trial Center for Functional Foods, Chonbuk National University HospitalJeonju, South Korea
| | - Soo-Wan Chae
- Clinical Trial Center for Functional Foods, Chonbuk National University HospitalJeonju, South Korea
- Division of Pharmacology, Chonbuk National University Medical SchoolJeonju, South Korea
| |
Collapse
|
37
|
Lactobacillus rhamnosus GG Intake Modifies Preschool Children's Intestinal Microbiota, Alleviates Penicillin-Associated Changes, and Reduces Antibiotic Use. PLoS One 2016; 11:e0154012. [PMID: 27111772 PMCID: PMC4844131 DOI: 10.1371/journal.pone.0154012] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 04/07/2016] [Indexed: 01/12/2023] Open
Abstract
Antibiotic use is considered among the most severe causes of disturbance to children’s developing intestinal microbiota, and frequently causes adverse gastrointestinal effects ranging from mild and transient diarrhoea to life-threatening infections. Probiotics are commonly advocated to help in preventing antibiotic-associated gastrointestinal symptoms. However, it is currently unknown whether probiotics alleviate the antibiotic-associated changes in children’s microbiota. Furthermore, it is not known how long-term probiotic consumption influences the developing microbiota of children. We analysed the influence of long-term Lactobacillus rhamnosus GG intake on preschool children’s antibiotic use, and antibiotic-associated gastrointestinal complaints in a double blind, randomized placebo-controlled trial with 231 children aged 2–7. In addition, we analysed the effect of L. rhanmosus GG on the intestinal microbiota in a subset of 88 children. The results show that long-term L. rhamnosus GG supplementation has an influence on the composition of the intestinal microbiota in children, causing an increase in the abundance of Prevotella, Lactococcus, and Ruminococcus, and a decrease in Escherichia. The treatment appeared to prevent some of the changes in the microbiota associated with penicillin use, but not those associated with macrolide use. The treatment, however, did reduce the frequency of gastrointestinal complaints after a macrolide course. Finally, the treatment appeared to prevent certain bacterial infections for up to 3 years after the trial, as indicated by reduced antibiotic use. Trial Registration: ClinicalTrials.gov NCT01014676
Collapse
|
38
|
Liévin-Le Moal V. A gastrointestinal anti-infectious biotherapeutic agent: the heat-treated Lactobacillus LB. Therap Adv Gastroenterol 2016; 9:57-75. [PMID: 26770268 PMCID: PMC4699271 DOI: 10.1177/1756283x15602831] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Experimental in vitro and in vivo studies support the hypothesis that heat-treated, lyophilized Lactobacillus acidophilus LB cells and concentrated, neutralized spent culture medium conserve the variety of pharmacological, antimicrobial activities of the live probiotic strain against several infectious agents involved in well-established acute and persistent watery diarrhoea and gastritis. Heat-treated cells and heat-stable secreted molecules trigger multiple strain-specific activities explaining the therapeutic efficacy of L. acidophilus LB. This review discusses the current body of knowledge on the antimicrobial mechanisms of action exerted by L. acidophilus LB demonstrated in in vitro and in vivo experimental studies, and the evidence for the therapeutic efficacy of this anti-infectious biotherapeutic agent proved in randomized clinical trials for the treatment of acute and persistent watery diarrhoea associated with several intestinal infectious diseases in humans.
Collapse
|
39
|
Lactobacillus rhamnosus GG Dosage Affects the Adjuvanticity and Protection Against Rotavirus Diarrhea in Gnotobiotic Pigs. J Pediatr Gastroenterol Nutr 2015; 60:834-43. [PMID: 25564808 DOI: 10.1097/mpg.0000000000000694] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES The use of immunostimulatory strains of probiotics as adjuvants has been increasingly recognized as a promising approach in enhancing vaccine immunogenicity; however, dose effects of probiotic adjuvants are not well defined. In the present study, we examined dose effects of a commonly used probiotic strain, Lactobacillus rhamnosus GG (LGG), on immunomodulation with 2 different dosages. METHODS Neonatal gnotobiotic pigs were inoculated with 2 oral doses of attenuated human rotavirus (AttHRV) vaccines and fed with 5 doses (LGG5X; total 2.1 × 10(6) colony-forming units) or 9 doses (LGG9X; total 3.2 × 10(6) colony-forming units) of LGG, starting at 3 days of age. RESULTS Both LGG feeding regimens enhanced the protection rate of AttHRV vaccine against diarrhea on virulent human rotavirus challenge. LGG5X, but not LGG9X, significantly enhanced rotavirus-specific intestinal memory B-cell responses to AttHRV; LGG5X also significantly enhanced virus-specific intestinal immunoglobulin A (IgA) antibody-secreting cell responses. Both regimens significantly enhanced rotavirus-specific serum IgA antibody responses to AttHRV. They also enhanced rotavirus-specific interferon-γ-producing effector/memory T-cell responses to AttHRV vaccine, with LGG9X being more effective than LGG5X, and both regimens downregulated CD4+CD25-FoxP3+ regulatory T (Treg) cell responses in most lymphoid tissues examined prechallenge and postchallenge and maintained the CD4+CD25+FoxP3+ Treg population in the ileum and intraepithelial lymphocyte postchallenge. LGG9X, however, did not significantly reduce total CD4+CD25-FoxP3+ Treg frequencies in the intestine and transforming growth factor-β-producing and interleukin (IL)-10-producing Treg frequencies in the blood. CONCLUSIONS These results indicate that LGG at both dosages functioned as effective probiotic adjuvant for AttHRV vaccine, but different dosages differentially modulated immune responses to favor either the mucosal IgA response (LGG5X) or the T-cell response (LGG9X).
Collapse
|
40
|
Yang JX, Yang JC. Mechanisms underlying protective effects of probiotics on intestinal epithelial. Shijie Huaren Xiaohua Zazhi 2015; 23:577-583. [DOI: 10.11569/wcjd.v23.i4.577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Intestinal epithelial cells (IECs), the first line of defense against pathogens, are an initial point of contact between the host and intestinal microbes. Growing evidence suggests that the interactions between the host and intestinal microbes may lead to dysregulated immune responses, while probiotics can reinforce the barrier function and exert a modest stimulation of the immune system to prevent this situation. On one hand, probiotics exert antagonistic functions via competition for nutrients, metabolites, and occupying effect. Therefore, probiotics can regulate the endogenous gastrointestinal flora and restrain exogenous pathogenic bacteria. On the other hand, IECs recognize probiotics and their metabolites through pattern recognition receptors to stimulate the non-specific immune responses. In addition, probiotics can induce IECs to produce the mucus layer covering the entire intestinal tract and prevent attachment and invasion of various bacterial pathogens. Clinical trials have also shown beneficial effects of probiotics as a potential preventive method for inflammatory bowel disease such as Crohn disease and ulcerative colitis.
Collapse
|
41
|
Dormitzer PR. Rotaviruses. MANDELL, DOUGLAS, AND BENNETT'S PRINCIPLES AND PRACTICE OF INFECTIOUS DISEASES 2015:1854-1864.e4. [DOI: 10.1016/b978-1-4557-4801-3.00152-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
42
|
Burgain J, Scher J, Lebeer S, Vanderleyden J, Cailliez-Grimal C, Corgneau M, Francius G, Gaiani C. Significance of bacterial surface molecules interactions with milk proteins to enhance microencapsulation of Lactobacillus rhamnosus GG. Food Hydrocoll 2014. [DOI: 10.1016/j.foodhyd.2014.03.029] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Wu CT, Chen PJ, Lee YT, Ko JL, Lue KH. Effects of immunomodulatory supplementation with Lactobacillus rhamnosus on airway inflammation in a mouse asthma model. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2014; 49:625-635. [PMID: 25440975 DOI: 10.1016/j.jmii.2014.08.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 07/15/2014] [Accepted: 08/07/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Asthma is a common allergic disease. In previous studies, probiotics improved the balance of intestinal microbes, reduced inflammation, and promoted mucosal tolerance. This study investigated whether oral administrations of Lactobacillus rhamnosus GG (LGG) inhibited allergen (ovalbumin or OVA)-induced airway inflammation in a mouse asthma model. METHODS The allergy/asthma animal model in this study was sensitization with OVA. After intranasal challenge with OVA, the airway inflammation and hyper-responsiveness were determined by a Buxco system, bronchoalveolar lavage fluid analysis with Liu stain, and enzyme-linked immunosorbent assay. Histopathologic changes in the lung were detected by hematoxylin and eosin staining and immunohistochemistry staining. RESULTS Both pre- and post-treatment with LGG suppressed the airway hyper-responsiveness to methacholine and significantly decreased the number of infiltrating inflammatory cells and Th2 cytokines in bronchoalveolar lavage fluid and serum compared with the OVA-sensitized mice. In addition, LGG reduced OVA-specific IgE levels in serum. Oral LGG decreased matrix metalloproteinase 9 expression in lung tissue and inhibited inflammatory cell infiltration. CONCLUSION LGG had an anti-inflammatory effect on OVA-induced airway inflammation and might be an additional or supplementary therapy for allergic airway diseases.
Collapse
Affiliation(s)
- Chia-Ta Wu
- Department of Emergency Medicine, Changhua Christian Hospital, Changhua, Taiwan; Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Peng-Jung Chen
- School of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Tzu Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Liang Ko
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ko-Haung Lue
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
44
|
Franz CM, Huch M, Mathara JM, Abriouel H, Benomar N, Reid G, Galvez A, Holzapfel WH. African fermented foods and probiotics. Int J Food Microbiol 2014; 190:84-96. [DOI: 10.1016/j.ijfoodmicro.2014.08.033] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/25/2014] [Accepted: 08/23/2014] [Indexed: 12/24/2022]
|
45
|
Khalesi S, Sun J, Buys N, Jayasinghe R. Effect of probiotics on blood pressure: a systematic review and meta-analysis of randomized, controlled trials. Hypertension 2014; 64:897-903. [PMID: 25047574 DOI: 10.1161/hypertensionaha.114.03469] [Citation(s) in RCA: 363] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/02/2014] [Indexed: 12/20/2022]
Abstract
Previous human clinical trials have shown that probiotic consumption may improve blood pressure (BP) control. The aim of the present systematic review was to clarify the effects of probiotics on BP using a meta-analysis of randomized, controlled trials. PubMed, Scopus, Cochrane Library (Central), Physiotherapy Evidence Database, and Clinicaltrial.gov databases were searched until January 2014 to identify eligible articles. Meta-analysis using a random-effects model was chosen to analyze the impact of combined trials. Nine trials were included. Probiotic consumption significantly changed systolic BP by -3.56 mm Hg (95% confidence interval, -6.46 to -0.66) and diastolic BP by -2.38 mm Hg (95% confidence interval, -2.38 to -0.93) compared with control groups. A greater reduction was found with multiple as compared with single species of probiotics, for both systolic and diastolic BP. Subgroup analysis of trials with baseline BP ≥130/85 mm Hg compared with <130/85 mm Hg found a more significant improvement in diastolic BP. Duration of intervention <8 weeks did not result in a significant reduction in systolic or diastolic BP. Furthermore, subgroup analysis of trials with daily dose of probiotics <10(11) colony-forming units did not result in a significant meta-analysis effect. The present meta-analysis suggests that consuming probiotics may improve BP by a modest degree, with a potentially greater effect when baseline BP is elevated, multiple species of probiotics are consumed, the duration of intervention is ≥8 weeks, or daily consumption dose is ≥10(11) colony-forming units.
Collapse
Affiliation(s)
- Saman Khalesi
- From the Griffith Health Institute (S.K., J.S., N.B.) and School of Medicine (S.K., J.S., R.J.), Griffith University, Australia; and Australia and Cardiac Services/Cardiology, Gold Coast Health, Australia (R.J.)
| | - Jing Sun
- From the Griffith Health Institute (S.K., J.S., N.B.) and School of Medicine (S.K., J.S., R.J.), Griffith University, Australia; and Australia and Cardiac Services/Cardiology, Gold Coast Health, Australia (R.J.)
| | - Nicholas Buys
- From the Griffith Health Institute (S.K., J.S., N.B.) and School of Medicine (S.K., J.S., R.J.), Griffith University, Australia; and Australia and Cardiac Services/Cardiology, Gold Coast Health, Australia (R.J.)
| | - Rohan Jayasinghe
- From the Griffith Health Institute (S.K., J.S., N.B.) and School of Medicine (S.K., J.S., R.J.), Griffith University, Australia; and Australia and Cardiac Services/Cardiology, Gold Coast Health, Australia (R.J.)
| |
Collapse
|
46
|
Segers ME, Lebeer S. Towards a better understanding of Lactobacillus rhamnosus GG--host interactions. Microb Cell Fact 2014; 13 Suppl 1:S7. [PMID: 25186587 PMCID: PMC4155824 DOI: 10.1186/1475-2859-13-s1-s7] [Citation(s) in RCA: 292] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus rhamnosus GG (LGG) is one of the most widely used probiotic strains. Various health effects are well documented including the prevention and treatment of gastro-intestinal infections and diarrhea, and stimulation of immune responses that promote vaccination or even prevent certain allergic symptoms. However, not all intervention studies could show a clinical benefit and even for the same conditions, the results are not univocal. Clearly, the host phenotype governed by age, genetics and environmental factors such as the endogenous microbiota, plays a role in whether individuals are responders or non-responders. However, we believe that a detailed knowledge of the bacterial physiology and the LGG molecules that play a key role in its host-interaction capacity is crucial for a better understanding of its potential health benefits. Molecules that were yet identified as important factors governing host interactions include its adhesive pili or fimbriae, its lipoteichoic acid molecules, its major secreted proteins and its galactose-rich exopolysaccharides, as well as specific DNA motifs. Nevertheless, future studies are needed to correlate specific health effects to these molecular effectors in LGG, and also in other probiotic strains.
Collapse
|
47
|
Freedman SB, Williamson-Urquhart S, Schuh S, Sherman PM, Farion KJ, Gouin S, Willan AR, Goeree R, Johnson DW, Black K, Schnadower D, Gorelick MH. Impact of emergency department probiotic treatment of pediatric gastroenteritis: study protocol for the PROGUT (Probiotic Regimen for Outpatient Gastroenteritis Utility of Treatment) randomized controlled trial. Trials 2014; 15:170. [PMID: 24885220 PMCID: PMC4037747 DOI: 10.1186/1745-6215-15-170] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/29/2014] [Indexed: 01/01/2023] Open
Abstract
Background The burden of acute gastroenteritis on children and their families continues to be enormous. Probiotics, defined as viable microbial preparations that have a beneficial effect on the health of the host, represent a rapidly expanding field. Although clinical trials in children with gastroenteritis have been performed, most have significant flaws, and guidelines do not consistently endorse their use. Methods/Design PROGUT is a randomized, placebo-controlled, double-blind, five-center, Canadian, emergency department trial. Children aged 3 months to 48 months who present between November 2013 and June 2017 with <72 hours of gastroenteritis symptoms will be assessed for eligibility. A total of 886 children will be randomized (1:1 allocation via an internet based, third party, randomization service) to receive 5 days of a combination probiotic agent (Lactobacillus rhamnosus and L. helveticus) or placebo. All participants, caregivers, and outcome assessors will be blinded to group assignment. The study includes three key outcomes: 1) clinical - the development of moderate to severe disease following an emergency department (ED) evaluation that employs a validated clinical score (Modified Vesikari Scale); 2) safety - side effect; and 3) mechanism - fecal secretory immunoglobulin A levels. Discussion Definitive data are lacking to guide the clinical use of probiotics in children with acute gastroenteritis. Hence, probiotics are rarely prescribed by North American physicians. However, the following current trends obligate an urgent assessment: 1) probiotics are sold as food supplements, and manufacturers can encourage their use while their relevance has yet to be established; 2) North American and European government agencies remain concerned about their value and safety; 3) some institutions are now recommending the routine use of probiotics; and 4) parents of affected children are often providing probiotics. With probiotic consumption increasing in the absence of solid evidence, there is a need to conduct this definitive trial to overcome the limitations of prior work in this field. Trial registration ClinicalTrials.gov: NCT01853124; first registered 9 May 2013.
Collapse
Affiliation(s)
- Stephen B Freedman
- Sections of Paediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, University of Calgary, 2888 Shaganappi Trail NW, Calgary, AB T3B 6A8, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Freedman SB, Ali S, Oleszczuk M, Gouin S, Hartling L. Treatment of acute gastroenteritis in children: an overview of systematic reviews of interventions commonly used in developed countries. ACTA ACUST UNITED AC 2014; 8:1123-37. [PMID: 23877938 DOI: 10.1002/ebch.1932] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Acute gastroenteritis (AGE) is an extremely common paediatric condition, which results in significant morbidity in children and is a financial burden to the society. OBJECTIVE The purpose of this overview is to critically evaluate the evidence currently available in the Cochrane Database of Systematic Reviews (CDSR) regarding the efficacy and safety of commonly considered treatment options in children with AGE. METHODS All Cochrane reviews evaluating the following treatments in children with AGE were eligible for inclusion: oral rehydration therapy, anti-emetics and probiotics. We excluded those focusing on the treatment of antibiotic associated or nosocomial diarrhoea, persistent (chronic) diarrhoea and the prevention of gastroenteritis. We focused on the following outcomes that were selected a priori as clinically important: rate of admission to the hospital; length of stay in hospital; rate of return visits; administration of intravenous (IV) therapy owing to failure of oral rehydration therapy; adverse events and dysnatremia. MAIN RESULTS Children who received oral rehydration therapy had a shorter length of stay in hospital compared with children who received IV therapy [mean difference, MD = -1.20 days (-2.38, -0.02)]; however, the result was no longer significant when an outlying study was removed. Children who received IV therapy were at increased risk of developing phlebitis [risk difference, RD= - 0.02 (-0.04, -0.01)], while paralytic ileus was more common in children receiving ORT [RD = 0.03 (confidence interval, CI 0.01-0.05)]. Children who received oral ondansetron had lower hospital admission rates to the emergency department (ED) and lower rates of IV rehydration during their ED stay compared with children receiving placebo [risk ration, RR = 0.40 (CI 0.19-0.83) and RR = 0.41 (CI 0.29-0.59), respectively]. Children receiving IV ondansetron had lower hospital admission rates to the ED than patients receiving placebo [RR = 0.21 (0.05, 0.93)]. Probiotic use amongst children hospitalized following AGE reduced the mean duration of hospitalization by 1.12 days (CI -1.16, -0.38). CONCLUSIONS Given that oral rehydration is less invasive than IV rehydration with no evidence of important clinical differences, it is the first choice for rehydration in children with AGE and mild-to-moderate dehydration. As the vast majority of children with AGE do not require IV rehydration, oral ondansetron administration to children with significant vomiting should be performed to reduce the use of IV rehydration and the need for hospital admission. In children deemed too unwell to receive oral rehydration therapy, IV ondansetron administration is an option, as its use is associated with lower hospital admission rates. Although probiotics appear to be an effective option for the treatment of AGE amongst hospitalized children, outpatient data is lacking and more studies are urgently needed to determine the optimal organism, dosing and duration of treatment.
Collapse
Affiliation(s)
- Stephen B Freedman
- Sections of Pediatric Emergency Medicine and Gastroenterology, Alberta Children's Hospital, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | |
Collapse
|
49
|
Live and heat-killed Lactobacillus rhamnosus GG upregulate gene expression of pro-inflammatory cytokines in 5-fluorouracil-pretreated Caco-2 cells. Support Care Cancer 2014; 22:1647-54. [PMID: 24500789 DOI: 10.1007/s00520-014-2137-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Accepted: 01/13/2014] [Indexed: 01/16/2023]
Abstract
PURPOSE This study investigates whether post-chemotherapeutic use of live and heat-killed Lactobacillus rhamnosus GG can modulate the expression of three pro-inflammatory cytokines in 5-fluorouracil (5-FU)-induced intestinal mucositis in vitro. METHODS Live L. rhamnosus GG and heat-killed L. rhamnosus GG were observed using scanning electron microscopy. To establish the duration required for optimal expression of tumor necrosis factor-α (TNF-α), monocyte chemotactic protein-1 (MCP-1), and interleukin-12 (IL-12), 5 μM of 5-FU was selected to treat 10-day-old Caco-2 cells for 4, 6, 8, and 24 h. Caco-2 cells were treated with 5-FU (5 μM) for 4 h, followed by the administration of live L. rhamnosus GG (multiplicity of infection = 25), and heat-killed L. rhamnosus GG for 2 and 4 h. Finally, total cellular RNA was isolated to quantify mRNA expression of TNF-α, MCP-1, and IL-12 using real-time PCR. RESULTS The results demonstrated that heat-killed L. rhamnosus GG remained structurally intact with elongation. A biphasic upregulated expression of TNF-α, MCP-1, and IL-12 was observed in 5-FU-treated Caco-2 cells at 4 and 24 h. Compared to non-L. rhamnosus GG controls in 5-FU-pretreated Caco-2 cells, a 2-h treatment of heat-killed L. rhamnosus GG significantly upregulated the MCP-1 expression (p < 0.05), and both live and heat-killed L. rhamnosus GG treatments lasting 4 h upregulated the TNF-α and MCP-1 expression (p < 0.05). Only live L. rhamnosus GG upregulated the IL-12 expression (p < 0.05). CONCLUSIONS Post-chemotherapeutic use of live or heat-killed L. rhamnosus GG can upregulate the gene expression of 5-FU-induced pro-inflammatory cytokines in Caco-2 cells. Human intestinal epithelium may be vulnerable to the post-chemotherapeutic use of L. rhamnosus GG in 5-FU-induced mucositis that requires further in vivo studies for clarification.
Collapse
|
50
|
Wanke M, Szajewska H. Probiotics for preventing healthcare-associated diarrhea in children: A meta-analysis of randomized controlled trials. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.pepo.2013.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|