1
|
Uttarwar RG, Mekonnen SA, Van Beeck W, Wang A, Finnegan P, Roberts RF, Merenstein D, Slupsky CM, Marco ML. Effects of Bifidobacterium animalis subsp. lactis BB-12 and yogurt on mice during oral antibiotic administration. Microbiol Res 2024; 286:127794. [PMID: 38852301 DOI: 10.1016/j.micres.2024.127794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024]
Abstract
Probiotics have the potential to prevent disruptions to normal gastrointestinal function caused by oral antibiotic use. In this study, we examined the capacity of Bifidobacterium animalis subspecies lactis BB-12 (BB-12) and yogurt, separately and combined, to mitigate the effects of the antibiotic amoxicillin-clavulanate (AMC) on the gut microbiota and metabolomes of C57BL/6 J mice. Male and female mice were administered either BB-12, yogurt, BB-12 in yogurt, or saline for 10 days concurrent with the inclusion of AMC in the drinking water. Male mice exposed to AMC exhibited significant reductions (p<0.05) in body weight over the course of the study compared to sham (no AMC) controls whereas no such effects were observed for female mice. AMC administration resulted in rapid alterations to the intestinal microbiota in both sexes irrespective of BB-12 or yogurt treatment, including significant (p<0.05) losses in bacterial cell numbers and changes in microbial alpha-diversity and beta-diversity in the feces and cecal contents. The effects of AMC on the gut microbiota were observed within one day of administration and the bacterial contents continued to change over time, showing a succession marked by rapid reductions in Muribaculaceae and Lachnospiraceae and temporal increases in proportions of Acholeplasmataceae (day 1) and Streptococcaceae and Leuconostocaceae (day 5). By day 10 of AMC intake, high proportions of Gammaproteobacteria assigned as Erwiniaceae or Enterobacteriaceae (average of 63 %), were contained in the stools and were similarly enriched in the cecum. The cecal contents of mice given AMC harbored significantly reduced concentrations of (branched) short-chain fatty acids (SCFA), aspartate, and other compounds, whereas numerous metabolites, including formate, lactate, and several amino acids and amino acid derivatives were significantly enriched. Despite the extensive impact of AMC, starting at day 7 of the study, the body weights of male mice given yogurt or BB-12 (in saline) with AMC were similar to the healthy controls. BB-12 (in saline) and yogurt intake was associated with increased Streptococcaceae and both yogurt and BB-12 resulted in lower proportions of Erwiniaceae in the fecal and cecal contents. The cecal contents of mice fed BB-12 in yogurt contained levels of formate, glycine, and glutamine that were equivalent to the sham controls. These findings highlight the potential of BB-12 and yogurt to mitigate antibiotic-induced gut dysbiosis.
Collapse
Affiliation(s)
- Ruchita G Uttarwar
- Department of Food Science & Technology, University of California, Davis, USA
| | - Solomon A Mekonnen
- Department of Food Science & Technology, University of California, Davis, USA
| | - Wannes Van Beeck
- Department of Food Science & Technology, University of California, Davis, USA
| | - Aidong Wang
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Peter Finnegan
- Department of Food Science & Technology, University of California, Davis, USA
| | | | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Carolyn M Slupsky
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, USA.
| |
Collapse
|
2
|
Liu S, Zhao S, Cheng Z, Ren Y, Shi X, Mu J, Ge X, Dai Y, Li L, Zhang Z. Akkermansia muciniphila Protects Against Antibiotic-Associated Diarrhea in Mice. Probiotics Antimicrob Proteins 2024; 16:1190-1204. [PMID: 37314693 DOI: 10.1007/s12602-023-10101-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 06/15/2023]
Abstract
Probiotics are used to prevent antibiotic-associated diarrhea (AAD) via the restoration of the gut microbiota. However, the precise effects of Akkermansia muciniphila (Akk), which is a promising probiotics, on AAD are unknown. Here, AAD models were established via the administration of lincomycin and ampicillin with or without pasteurized Akk or Amuc_1100 treatment. A diffusion test revealed that Akk was susceptible to the majority of the antibiotics, such as ampicillin. These effects were confirmed by the reduced Akk abundance in AAD model mice. Pasteurized Akk or Amuc_1100 significantly decreased the diarrhea status score and colon injury of AAD model mice. Additionally, these treatments significantly decreased the relative abundance of Citrobacter at genus level and reshaped the metabolic function of gut microbiota. Notably, pasteurized Akk or Amuc_1100 significantly changed the serum metabolome of AAD model mice. In addition, pasteurized Akk or Amuc_1100 suppressed intestinal inflammation by upregulating the expression of GPR109A and SLC5A8 and downregulating the expression of TNFα, IFNγ, IL1β, and IL6. Furthermore, they enhanced water and electrolyte absorption by upregulating AQP4, SLC26A3, and NHE3. Pasteurized Akk or Amuc_1100 also restored intestinal barrier function by ameliorating the downregulation of ZO-1, OCLN, CLDN4, and Muc2 in AAD model mice. In summary, optimizing intestinal health with pasteurized Akk or Amuc_1100 may serve as an approach for preventing AAD.
Collapse
Affiliation(s)
- Shenyin Liu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Suying Zhao
- Department of Laboratory Medicine, The Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Nanjing, 210009, People's Republic of China
| | - Zhiwei Cheng
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Yilin Ren
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xinyi Shi
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Jing Mu
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xiangyang Ge
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Yuan Dai
- Technical Department of Sujiu Group, Suqian, 223800, People's Republic of China
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| | - Zhan Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
3
|
Wang Q, He M, Liang J, Tan X, Wu Q, Wang J, Li X, Qiao M, Huang Z, Xie Q, Liu Z, Ren H, Wang L, Zhou H, Shao L, Shu R, Wu W, Yang W, Wang H, Sun Z, Xu X, Zhang X, Li Z, Zhang Y, Meng J, Zhu Y, Chen F, Qu R, Chen P, Li S, Shi Y, Mao X, Hu B, Zhang Y, Cao YJ, Guo Z. Chinese guidelines for integrated diagnosis and treatment of intestinal microecology technologies in tumor application (2024 Edition). J Cancer Res Ther 2024; 20:1130-1140. [PMID: 39206974 DOI: 10.4103/jcrt.jcrt_32_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
ABSTRACT Intestinal microecology (IM) is the largest and most important microecological system of the human body. Furthermore, it is the key factor for activating and maintaining the physiological functions of the intestine. Numerous studies have investigated the effects of the gut microbiota on the different tissues and organs of the human body as well as their association with various diseases, and the findings are gradually being translated into clinical practice. The gut microbiota affects the occurrence, progression, treatment response, and toxic side effects of tumors. The deepening of research related to IM and tumors has opened a new chapter in IM research driven by methods and technologies such as second-generation sequencing and bioinformatics. The IM maintains the function of the host immune system and plays a pivotal role in tumor-control drug therapy. Increasing evidence has proven that the efficacy of tumor-control drugs largely depends on the IM balance, and strategies based on the IM technology show promising application prospects in the diagnosis and treatment of tumor. The Tumor and Microecology Professional Committee of the Chinese Anti-cancer Association gathered relevant experts to discuss and propose the "Chinese guidelines for integrated diagnosis and treatment of IM technologies in tumor application (2024 Edition)," which was established based on the research progress of the application of the IM technology in tumor to provide a basis for the standardization of the diagnosis and treatment of the IM technology in the tumor.
Collapse
Affiliation(s)
- Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingxin He
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Xiaohua Tan
- Department of Oncology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jun Wang
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingqiang Qiao
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Ziming Huang
- Hubei Maternal and Child Health Care Hospital, Wuhan, China
| | - Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Zhe Liu
- Medical College, Tianjin University, Tianjin, China
| | - Hua Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rong Shu
- The Third People's Hospital of Hubei Province, Wuhan, China
| | - Wei Wu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Wenyan Yang
- Shangdong First Medical University and Shangdong Academy of Medical Sciences, Jinan, China
| | - Hua Wang
- Department of Hematology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Zhiqiang Sun
- Department of Hematology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xingding Zhang
- The School of Medicine of Sun Yat-Sen University, Shenzhen, China
| | - Zhiming Li
- Medical Department, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Shenzhen, China
| | - Jingye Meng
- Department of Hematology and Oncology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yanli Zhu
- The First Affliated Hosptial of Xinxiang Medical University, Xinxiang, China
| | - Feng Chen
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Qu
- Department of Critical Care Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Peng Chen
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuluan Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuanyuan Shi
- Shenzhen Cell Valley Biomedicine Co. LTD, Shenzhen, China
| | - Xin Mao
- Primary Health Care Foundation of China, Xiangyang, China
| | - Bichuan Hu
- Xiangyang Hospital of Integrated Traditional Chinese and Western Medicine, Xiangyang, China
| | - Yukui Zhang
- Xiangyang Hospital of Traditional Chinese Medicine, Xiangyang, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zhi Guo
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
4
|
Morán J, Kilasoniya A. Integration of Postbiotics in Food Products through Attenuated Probiotics: A Case Study with Lactic Acid Bacteria in Bread. Foods 2024; 13:2042. [PMID: 38998548 PMCID: PMC11240946 DOI: 10.3390/foods13132042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
The study examines the integration of postbiotics in food products through the use of attenuated probiotics, specifically lactic acid bacteria (LAB) in bread. Postbiotics, non-viable microorganisms or their metabolites, offer health benefits similar to probiotics without the risks associated with live bacteria. This research evaluates the regulatory aspects and safety of LAB in sourdough bread production, highlighting their historical and significant use in Europe before 1997. The study includes microbial quantification and Next-Generation Sequencing (NGS) to identify LAB in traditional sourdough, comparing them with historical and current EFSA Qualified Presumption of Safety (QPS) lists. Findings show that the LAB present in sourdough have been extensively and safely used in bread making, supporting their classification as non-novel foods under EU regulations. The stability and consistency of LAB metabolites in sourdough bread are also confirmed, ensuring quality and safety in each batch. The study concludes that LAB in sourdough, when inactivated through bread-making processes, are not considered novel foods, aligning with historical, scientific, and regulatory evidence.
Collapse
Affiliation(s)
- Javier Morán
- Department of Food Innovation, Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Alina Kilasoniya
- International PhD School, Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| |
Collapse
|
5
|
Phan J, Calvo DC, Nair D, Jain S, Montagne T, Dietsche S, Blanchard K, Treadwell S, Adams J, Krajmalnik-Brown R. Precision synbiotics increase gut microbiome diversity and improve gastrointestinal symptoms in a pilot open-label study for autism spectrum disorder. mSystems 2024; 9:e0050324. [PMID: 38661344 PMCID: PMC11097633 DOI: 10.1128/msystems.00503-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024] Open
Abstract
The efficacy of prebiotics and probiotics (synbiotics when combined) to improve symptoms associated with autism spectrum disorder (ASD) has shown considerable inter-study variation, likely due to the complex, heterogeneous nature of the disorder and its associated behavioral, developmental, and gastrointestinal symptoms. Here, we present a precision synbiotic supplementation study in 296 children and adults diagnosed with ASD versus 123 age-matched neurotypical controls. One hundred seventy ASD participants completed the study. Baseline and post-synbiotic assessment of ASD and gastrointestinal (GI) symptoms and deep metagenomic sequencing were performed. Within the ASD cohort, there were significant differences in microbes between subpopulations based on the social responsiveness scale (SRS2) survey (Prevotella spp., Bacteroides, Fusicatenibacter, and others) and gluten and dairy-free diets (Bifidobacterium spp., Lactococcus, Streptococcus spp., and others). At the baseline, the ASD cohort maintained a lower taxonomic alpha diversity and significant differences in taxonomic composition, metabolic pathways, and gene families, with a greater proportion of potential pathogens, including Shigella, Klebsiella, and Clostridium, and lower proportions of beneficial microbes, including Faecalibacterium compared to controls. Following the 3-month synbiotic supplementation, the ASD cohort showed increased taxonomic alpha diversity, shifts in taxonomy and metabolic pathway potential, and improvements in some ASD-related symptoms, including a significant reduction in GI discomfort and overall improved language, comprehension, cognition, thinking, and speech. However, the open-label study design may include some placebo effects. In summary, we found that precision synbiotics modulated the gut microbiome and could be used as supplementation to improve gastrointestinal and ASD-related symptoms. IMPORTANCE Autism spectrum disorder (ASD) is prevalent in 1 out of 36 children in the United States and contributes to health, financial, and psychological burdens. Attempts to identify a gut microbiome signature of ASD have produced varied results. The limited pre-clinical and clinical population sizes have hampered the success of these trials. To understand the microbiome associated with ASD, we employed whole metagenomic shotgun sequencing to classify microbial composition and genetic functional potential. Despite being one of the most extensive ASD post-synbiotic assessment studies, the results highlight the complexity of performing such a case-control supplementation study in this population and the potential for a future therapeutic approach in ASD.
Collapse
Affiliation(s)
- Joann Phan
- Sun Genomics, Inc., San Diego, California, USA
| | - Diana C. Calvo
- Department of Civil Engineering, Construction Management, and Environmental Engineering, Northern Arizona University, Flagstaff, Arizona, USA
| | - Divya Nair
- Sun Genomics, Inc., San Diego, California, USA
| | - Suneer Jain
- Sun Genomics, Inc., San Diego, California, USA
| | | | | | | | | | - James Adams
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, Arizona, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
6
|
Luan N, Zuo J, Niu Q, Yan W, Hung TC, Liu H, Wu Q, Wang G, Deng P, Ma X, Qin J, Li G. Probiotic Lactobacillus rhamnosus alleviates the neurotoxicity of microcystin-LR in zebrafish (Danio rerio) through the gut-brain axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168058. [PMID: 37914124 DOI: 10.1016/j.scitotenv.2023.168058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 10/13/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Microcystin-LR (MCLR) is one of the most toxic cyanobacterial toxins and is harmful to the central nervous system of fish. Probiotic additives can improve neuroendocrine function in fish. Although both MCLR and probiotics aim at the nervous system, whether they interact with each other and the mechanisms remain unexplored. In the present study, 4-month-old zebrafish were exposed to 0, 2.2, and 22 μg/L of MCLR for 28 days with or without the probiotic L. rhamnosus. We found that MCLR exposure could inhibit the swimming speed of zebrafish, while the presence of L. rhamnosus mitigated this abnormality. To elucidate the mechanism of how L. rhamnosus alleviates MCLR-induced neurotoxicity, we examined the bioaccumulation of MCLR, changes in neurotransmitters, immune biochemical indicators, and hormone content of the hypothalamic-pituitary-interrenal (HPI) axis in zebrafish along the gut-brain axis. Our results showed L. rhamnosus could reverse the abnormal swimming behavior and eventually alleviate neurotoxicity in zebrafish by modulating intestinal and brain neural signaling, neuroinflammation, and HPI axis responses. This study provides implications for the application of probiotics in the aquaculture industry.
Collapse
Affiliation(s)
- Ning Luan
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Junli Zuo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qianping Niu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Wei Yan
- Institute of Quality Standard and Testing Technology for Agro-Products, Hubei Academy of Agricultural Sciences, Hubei Key Laboratory of Nutritional Quality and Safety of Agro-products, Wuhan 430064, Hubei, China
| | - Tien-Chieh Hung
- Department of Biological and Agricultural Engineering, University of California-Davis, Davis, CA 95616, USA
| | - Haoling Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qin Wu
- Hubei Key Laboratory of Edible Wild Plants Conservation and Utilization, Huangshi Key Laboratory of Lake Biodiversity and Environmental Conservation, Hubei Normal University, Huangshi, Hubei Province 435002, China
| | - Guoao Wang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ping Deng
- Study and practical demonstratiministryon on regime shifts and optimization of ecosystem after ecological restoration project 'turning fishpond to wetland' in Chenhu Lake, Wuhan Academy of Agricultural Sciences, Wuhan 430056, China
| | - Xufa Ma
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianhui Qin
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Guangyu Li
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
7
|
Batista KS, de Albuquerque JG, de Vasconcelos MHA, Bezerra MLR, da Silva Barbalho MB, Pinheiro RO, Aquino JDS. Probiotics and prebiotics: potential prevention and therapeutic target for nutritional management of COVID-19? Nutr Res Rev 2023; 36:181-198. [PMID: 34668465 PMCID: PMC8593414 DOI: 10.1017/s0954422421000317] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 08/21/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
Scientists are working to identify prevention/treatment methods and clinical outcomes of coronavirus disease 2019 (COVID-19). Nutritional status and diet have a major impact on the COVID-19 disease process, mainly because of the bidirectional interaction between gut microbiota and lung, that is, the gut-lung axis. Individuals with inadequate nutritional status have a pre-existing imbalance in the gut microbiota and immunity as seen in obesity, diabetes, hypertension and other chronic diseases. Communication between the gut microbiota and lungs or other organs and systems may trigger worse clinical outcomes in viral respiratory infections. Thus, this review addresses new insights into the use of probiotics and prebiotics as a preventive nutritional strategy in managing respiratory infections such as COVID-19 and highlighting their anti-inflammatory effects against the main signs and symptoms associated with COVID-19. Literature search was performed through PubMed, Cochrane Library, Scopus and Web of Science databases; relevant clinical articles were included. Significant randomised clinical trials suggest that specific probiotics and/or prebiotics reduce diarrhoea, abdominal pain, vomiting, headache, cough, sore throat, fever, and viral infection complications such as acute respiratory distress syndrome. These beneficial effects are linked with modulation of the microbiota, products of microbial metabolism with antiviral activity, and immune-regulatory properties of specific probiotics and prebiotics through Treg cell production and function. There is a need to conduct clinical and pre-clinical trials to assess the combined effect of consuming these components and undergoing current therapies for COVID-19.
Collapse
Affiliation(s)
- Kamila Sabino Batista
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| | - Juliana Gondim de Albuquerque
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Pernambuco (UFPE), Cidade Universitária s/n, Recife, Brazil
- Post Graduate in Biotechnology, Division of Biological and Health Sciences, Universidad Autónoma Metropolitana (UAM), Ciudad de Mexico, Mexico
| | - Maria Helena Araújo de Vasconcelos
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| | - Maria Luiza Rolim Bezerra
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| | - Mariany Bernardino da Silva Barbalho
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| | - Rafael Oliveira Pinheiro
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| | - Jailane de Souza Aquino
- Experimental Nutrition Laboratory, Department of Nutrition, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), Cidade Universitária, s/n-Castelo Branco III, João Pessoa, PB, Brazil
| |
Collapse
|
8
|
Liang W, Gao Y, Zhao Y, Gao L, Zhao Z, He Z, Li S. Lactiplantibacillus plantarum ELF051 Alleviates Antibiotic-Associated Diarrhea by Regulating Intestinal Inflammation and Gut Microbiota. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10150-x. [PMID: 37639209 DOI: 10.1007/s12602-023-10150-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Probiotics are widely recognized for their ability to prevent and therapy antibiotic-associated diarrhea (AAD). This study was designed to evaluate Lactiplantibacillus plantarum ELF051 ability to prevent colon inflammation and its effect on gut microbial composition in a mouse model of AAD. The mice were intragastrically administered triple antibiotics for 7 days and then subjected to L. plantarum ELF051 for 14 days. The administration of L. plantarum ELF051 ameliorated the pathological changes in the colon tissue, downregulated interleukin (IL)-1β and tumor necrosis factor (TNF)-α, and upregulated IL-10, and increased the intestinal short-chain fatty acids (SCFAs) level. Lactiplantibacillus plantarum ELF051 also regulated the Toll-like receptor/myeloid differentiation primary response 88/nuclear factor kappa light chain enhancer of activated B cells (TLR4/MyD88/NF-κB) and the phosphatidylinositol 3-kinase/protein kinase B/ NF-κB (PI3K/AKT/ NF-κB) inflammatory signaling pathways. 16S rRNA analyses showed that L. plantarum ELF051 increased the abundance and diversity of gut bacteria, restoring gut microbiota imbalance. A Spearman's rank correlation analysis showed that lactobacilli are closely associated with inflammatory markers and SCFAs. This work demonstrated that L. plantarum ELF051 can attenuate antibiotic-induced intestinal inflammation in a mouse AAD model by suppressing the pro-inflammatory response and modulating the gut microbiota.
Collapse
Affiliation(s)
- Wei Liang
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yansong Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Yujuan Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| | - Lei Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zijian Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China
| | - Zhongmei He
- College of Chinese Medicinal Material, Jilin Agricultural University, Changchun, 130118, China
| | - Shengyu Li
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences, No. 1363 Sheng-Tai Street, Changchun, 130033, China.
| |
Collapse
|
9
|
Musina ON, Ott EF, Funk IA, Dorofeev RV, Filimonova TV. Study of Survival During Drying of Bacterial Cells of Starter Culture for Probiotic Fermented Milk Drinks. BIO WEB OF CONFERENCES 2023. [DOI: 10.1051/bioconf/20235705002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Five proprietary variants of bacterial starter culture with the following species composition were studied: No. 1 – Lac. diacetilactis, Lac. cremoris, Lac. acidophilus, No. 2 – Lb. plantarum, B. adolescentis, No. 3 – Lac. cremoris, No. 4 – Lac. lactis, Lac. diacetilactis, Lb. plantarum, No. 5 – Lac. lactis, Lac. diacetilactis, Lb. cremoris. Survival was assessed immediately after freeze-drying and during storage after 30, 90 and 180 days. It was found that the lactococci included in the bacterial starter culture of variants No. 1, 3-5 after drying and during storage showed the maximum survival of bacterial cells. Their number varied within the limits: after drying – 3.3-6.5 × 109 CFU/g, after 180 days – 1.0-2.0 × 109 CFU/g. A high degree of survival was noted in L. acidophilus (variant No. 1) – the number of viable cells after drying, as well as after 180 days of storage at the level of 1.0×108 CFU/g. The total amount of probiotic microflora of Lb. plantarum and B. adolescentis bacterial starter culture (variant No. 2) after drying was 3.2×109 CFU/g, and after 180 days of storage 1.0×109 CFU/g. Both cultures showed high survival of bacterial cells. The number of Lb. plantarum (variant No. 4) after drying and during storage was only 1.0×106 CFU / g, but it should be taken into account that this culture in the starter composition is additional, while the dominant microflora is represented by lactococci.
Collapse
|
10
|
Wortelboer K, Koopen AM, Herrema H, de Vos WM, Nieuwdorp M, Kemper EM. From fecal microbiota transplantation toward next-generation beneficial microbes: The case of Anaerobutyricum soehngenii. Front Med (Lausanne) 2022; 9:1077275. [PMID: 36544495 PMCID: PMC9760881 DOI: 10.3389/fmed.2022.1077275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The commensal gut microbiota is important for human health and well-being whereas deviations of the gut microbiota have been associated with a multitude of diseases. Restoration of a balanced and diverse microbiota by fecal microbiota transplantation (FMT) has emerged as a potential treatment strategy and promising tool to study causality of the microbiota in disease pathogenesis. However, FMT comes with logistical challenges and potential safety risks, such as the transfer of pathogenic microorganisms, undesired phenotypes or an increased risk of developing disease later in life. Therefore, a more controlled, personalized mixture of cultured beneficial microbes might prove a better alternative. Most of these beneficial microbes will be endogenous commensals to the host without a long history of safe and beneficial use and are therefore commonly referred to as next-generation probiotics (NGP) or live biotherapeutic products (LBP). Following a previous FMT study within our group, the commensal butyrate producer Anaerobutyricum spp. (previously named Eubacterium hallii) was found to be associated with improved insulin-sensitivity in subjects with the metabolic syndrome. After the preclinical testing with Anaerobutyricum soehngenii in mice models was completed, the strain was produced under controlled conditions and several clinical studies evaluating its safety and efficacy in humans were performed. Here, we describe and reflect on the development of A. soehngenii for clinical use, providing practical guidance for the development and testing of NGPs and reflecting on the current regulatory framework.
Collapse
Affiliation(s)
- Koen Wortelboer
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Pharmacy, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Annefleur M. Koopen
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Hilde Herrema
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
| | - Willem M. de Vos
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Laboratory of Microbiology, Wageningen University, Wageningen, Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Department of Experimental Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, Netherlands
- Department of Vascular Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
- Diabetes Center, Department of Internal Medicine, Amsterdam UMC, Location VUMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - E. Marleen Kemper
- Department of Pharmacy, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
11
|
Zhao L, Zhang D, Liu Y, Zhang YN, Meng DQ, Xu Q, Zhong J, Jiang QY, Zhao Y, Wang SJ. Quantitative PCR Assays for the Strain-Specific Identification and Enumeration of Probiotic Strain Lacticaseibacillus rhamnosus X253. Foods 2022; 11:foods11152282. [PMID: 35954048 PMCID: PMC9367767 DOI: 10.3390/foods11152282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
Probiotics are universally recognized for their health benefits, despite the fact that their effects depend on the strain. Identification and enumeration of probiotic strains are required prior to evaluating their effectiveness. Lacticaseibacillus rhamnosus X253 is a potential probiotic strain with antioxidant capacity. Comparative genomics and single nucleotide polymorphisms (SNPs) were used to identify a strain-specific locus within the holA gene for strain X253 that was distinct in 30 different L. rhamnosus strains. Using quantitative PCR, the primers and probe designed for the locus were able to distinguish L. rhamnosus X253 from the other 20 probiotic strains. The chosen locus remained stable over 19 generations. The sensitivity of the assay was 0.2 pg genomic DNA of L. rhamnosus X253, or 103 cfu/mL bacteria of this strain. In terms of repeatability and reproducibility, relative standard deviations (RSD) were less than 1% and 3%, respectively. Additionally, this assay achieved accurate enumerations of L. rhamnosus X253 in spiked milk and complex powder samples. The strain-specific assay could be used for quality control and compliance assessment of dairy products.
Collapse
Affiliation(s)
- Lei Zhao
- Key Laboratory of Milk and Dairy Products Detection and Monitoring Technology for State Market Regulation, Shanghai Institute of Quality Inspection and Technical Research, Shanghai 200233, China; (L.Z.); (Y.-N.Z.); (Q.X.)
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China; (J.Z.); (Q.-Y.J.)
| | - Dong Zhang
- Junlebao Dairy Group, Shijiazhuang 050221, China;
| | - Yang Liu
- Key Laboratory of Milk and Dairy Products Detection and Monitoring Technology for State Market Regulation, Shanghai Institute of Quality Inspection and Technical Research, Shanghai 200233, China; (L.Z.); (Y.-N.Z.); (Q.X.)
- Correspondence: (Y.L.); (S.-J.W.); Tel.: +86-021-54263408 (Y.L.); +86-0311-86266225 (S.-J.W.)
| | - Yi-Nan Zhang
- Key Laboratory of Milk and Dairy Products Detection and Monitoring Technology for State Market Regulation, Shanghai Institute of Quality Inspection and Technical Research, Shanghai 200233, China; (L.Z.); (Y.-N.Z.); (Q.X.)
| | - Dong-Qing Meng
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China; (D.-Q.M.); (Y.Z.)
| | - Qiong Xu
- Key Laboratory of Milk and Dairy Products Detection and Monitoring Technology for State Market Regulation, Shanghai Institute of Quality Inspection and Technical Research, Shanghai 200233, China; (L.Z.); (Y.-N.Z.); (Q.X.)
| | - Jiang Zhong
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China; (J.Z.); (Q.-Y.J.)
| | - Qiu-Yue Jiang
- Department of Microbiology and Microbial Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China; (J.Z.); (Q.-Y.J.)
| | - Yu Zhao
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, China; (D.-Q.M.); (Y.Z.)
| | - Shi-Jie Wang
- Junlebao Dairy Group, Shijiazhuang 050221, China;
- Correspondence: (Y.L.); (S.-J.W.); Tel.: +86-021-54263408 (Y.L.); +86-0311-86266225 (S.-J.W.)
| |
Collapse
|
12
|
Bao W, He Y, Yu J, Liu M, Yang X, Ta N, Zhang E, Liang C. Regulatory Effect of Lactiplantibacillus plantarum 2-33 on Intestinal Microbiota of Mice With Antibiotic-Associated Diarrhea. Front Nutr 2022; 9:921875. [PMID: 35757257 PMCID: PMC9218693 DOI: 10.3389/fnut.2022.921875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Diarrhea is one of the common adverse reactions in antibiotic treatment, which is usually caused by the imbalance of intestinal flora, and probiotics play an important role in the structure of intestinal flora. Therefore, this experiment studied the regulatory effect of Lactiplantibacillus plantarum 2-33 on antibiotic-associated diarrhea (AAD) mice. First, the AAD mice model was established by the mixed antibiotic solution of gentamicin sulfate and cefradine. Then, the physiological indexes and diarrhea of mice were observed and recorded by gastric perfusion of low dose (1.0 × 107 CFU/ml), medium dose (1.0 × 108CFU/ml), and high dose (1.0 × 109 CFU/ml) strain 2-33. 16S rRNA gene V3-V4 regions were sequenced in colon contents of mice in control group, model group, self-healing group, and experimental group, respectively, and the diversity of intestinal flora and gene function prediction were analyzed. The results showed that the intestinal flora of AAD mice was not significantly regulated by gastric perfusion of strain 2-33 to 7 days, but the relative abundance and diversity of intestinal flora of AAD mice were significantly improved by gastric perfusion to 14 days (p < 0.05). In addition, at the genus level, the relative abundance of Lactobacillus increased significantly, and the relative abundance of Enterococcus and Bacillus decreased significantly (p < 0.05). In addition, the regulation of strain 2-33 on intestinal flora of AAD mice was time- and dose-dependent, short-term gastric perfusion, and low dose had no significant effect (p > 0.05). Strain 2-33 can significantly increase the levels of anti-inflammatory cytokines IL-4 and IL-10, significantly decrease the levels of proinflammatory cytokines TNF-α and IFN-γ (p < 0.05), and can also adjust carbohydrate metabolism, amino acid metabolism, and energy metabolism to normal levels, thus accelerating the recovery of intestinal flora structure of AAD mice. In summary, strain 2-33 can improve the structure and diversity of intestinal flora of AAD mice, balance the level of substance and energy metabolism, and play a positive role in relieving diarrhea, maintaining and improving the intestinal microecological balance.
Collapse
Affiliation(s)
- Wuyundalai Bao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Yuxing He
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Jinghe Yu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Mingchao Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Xiaofeng Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Na Ta
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Enxin Zhang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Chengyuan Liang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
13
|
Xu B, Liang S, Zhao J, Li X, Guo J, Xin B, Li B, Huo G, Ma W. Bifidobacterium animalis subsp. lactis XLTG11 improves antibiotic-related diarrhea by alleviating inflammation, enhancing intestinal barrier function and regulating intestinal flora. Food Funct 2022; 13:6404-6418. [PMID: 35616024 DOI: 10.1039/d1fo04305f] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common side effect during antibiotic treatment. In this study, we evaluated the regulatory effect of Bifidobacterium animalis subsp. lactis XLTG11 on mouse diarrhea caused by antibiotic-induced intestinal flora disturbance. Then, two strains of Bifidobacterium animalis subsp. lactis XLTG11 and Bifidobacterium animalis subsp. lactis BB-12 were administered to AAD mice. We found that the recovery effect of using B. lactis XLTG11 was better than that of B. lactis BB-12. B. lactis XLTG11 reduced the pathological characteristics of the intestinal tract, and significantly reduced the levels of lipopolysaccharide (LPS), D-lactic acid (D-LA) and diamine oxidase (DAO) to decrease intestinal permeability. In addition, these two strains significantly increased the expression of aquaporin and tight junction proteins, and inhibited toll-like receptor 4 (TLR4)/activation of the nuclear factor-κB (NF-κB) signaling pathway, significantly increased the levels of anti-inflammatory cytokines and decreased levels of pro-inflammatory cytokines. Moreover, after treatment with B. lactis XLTG11, the contents of acetic acid, propionic acid, butyric acid and total short-chain fatty acids were significantly increased. Compared with the MC group, B. lactis XLTG11 increased the abundance and diversity of the intestinal flora and changed the composition of the intestinal flora. We found that B. lactis XLTG11 can promote the recovery of intestinal flora and mucosal barrier function, thereby effectively improving AAD-related symptoms, providing a scientific basis for future clinical applications.
Collapse
Affiliation(s)
- Baofeng Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Shengnan Liang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayi Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Xuetong Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Jiayao Guo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bowen Xin
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China. .,Food College, Northeast Agricultural University, Harbin 150030, China. .,Heilongjiang Key Laboratory of Genetic and Metabolic Engineering of Lactic Acid Bacteria, Harbin 150030, China
| | - Weiwei Ma
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Haerbin 150000, China.
| |
Collapse
|
14
|
Ali MA, Kamal MM, Rahman MH, Siddiqui MN, Haque MA, Saha KK, Rahman MA. Functional dairy products as a source of bioactive peptides and probiotics: current trends and future prospectives. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2022; 59:1263-1279. [PMID: 35250052 PMCID: PMC8882518 DOI: 10.1007/s13197-021-05091-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 02/23/2021] [Accepted: 04/04/2021] [Indexed: 12/31/2022]
Abstract
Milk is an incredibly healthy food world-wide. However, the 'lactase deficient' individuals cannot digest milk's carbohydrate lactose. A large part of the world population is depriving of highly beneficial milk proteins like casein, lactoalbumin, lactoglobulin, etc. due to lactose intolerance. Production of functional foods and bioactive peptides from milk with natural antioxidants and the addition of probiotics could be the best alternative to extend the use of milk functionalities. Among different probiotics, the lactic acid bacteria (LAB) like Lactobacillus delbrueckii sub sp. bulgaricus, Streptococcus thermophilus and some species of Bifidobacteria and their metabolites (paraprobiotics and postbiotics) have been given more preference to add in milk-derived functional foods. These species are generally considered as heat-tolerant, highly proteolytic, and peptidolytic towards milk proteins and they liberate smaller molecules of bioactive peptides during fermentation and other processes that stimulate the enzyme lactase to help people in digestion of milk carbohydrate lactose. Moreover, the incorporation of natural antioxidants in yoghurt and other dairy products prevents the rancidity of milk fat. The level of bioactive peptides produced in milk-derived functional foods can be determined by capillary zone electrophoresis, mass spectrometry, fractionation, and other modern assessment techniques. Commercial production of functional probiotic products with bioactive peptides could significantly contribute to reduce milk spoilage, enhance health benefits as well as the growth of the agro-processing industry.
Collapse
Affiliation(s)
- Md. Aslam Ali
- Department of Agro-Processing, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, 1706 Bangladesh
| | - Md. Mostafa Kamal
- Department of Food Processing and Preservation, Hajee Mohammad Danesh Science and Technology University, Dinajpur, 5200 Bangladesh
| | - Md. Hafizur Rahman
- Laboratory of Food Chemistry and Nutrition Science, Institute of Food Science and Technology, Chinese Academy of Agricultural Science, Beijing, China
| | - Md. Nurealam Siddiqui
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, 1706 Bangladesh
| | - Md. Azizul Haque
- Faculty of Science and Technology, Free University of Bozen-Bolzano, Piazza Università 1, 39100 Bolzano, Italy
| | - Khokan Kumar Saha
- Department of Agricultural Engineering, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, 1706 Bangladesh
| | - Md. Atikur Rahman
- Department of Food Processing and Preservation, Hajee Mohammad Danesh Science and Technology University, Dinajpur, 5200 Bangladesh
| |
Collapse
|
15
|
Bacillus subtilis ameliorates Escherichia coli-induced endometritis in mice via maintaining endometrial barrier and inhibiting inflammatory response. Microb Pathog 2022; 166:105487. [DOI: 10.1016/j.micpath.2022.105487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/19/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022]
|
16
|
Huang R, Wu F, Zhou Q, Wei W, Yue J, Xiao B, Luo Z. Lactobacillus and intestinal diseases: mechanisms of action and clinical applications. Microbiol Res 2022; 260:127019. [DOI: 10.1016/j.micres.2022.127019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
|
17
|
Li E, Zhu Q, Pang D, Liu F, Liao S, Zou Y. Analysis of Lactobacillus rhamnosus GG in Mulberry Galacto-Oligosaccharide Medium by Comparative Transcriptomics and Metabolomics. Front Nutr 2022; 9:853271. [PMID: 35369065 PMCID: PMC8973438 DOI: 10.3389/fnut.2022.853271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Lactobacillus rhamnosus GG (LGG) has strong acid resistance and can survive passing through the stomach to colonize the intestines, where it promotes the growth of beneficial bacteria. Prebiotics such as mulberry galacto-oligosaccharide (MGO), mulberry polysaccharide solution (MPS), and galactooligosaccharides (GOS) promote LGG proliferation, and MGO has the greatest effect. After culturing LGG with prebiotics, changes in gene expression were studied at the transcriptomic and metabolomic levels. The results showed that, in the stable 24-h growth period of cultivation, ~63 and 132% more differential genes were found after MPS and MGO were added to the MRS medium, respectively, than after GOS was added, and the numbers of up-regulated genes were about 18 and 66% higher with MPS and MGO, respectively, than GOS. Analysis using the KEGG database revealed that, when LGG was cultured with MGO, 120 genes that were up-regulated as the growth rate increased were mainly enriched in pathways such as membrane transport, amino acid metabolism, and carbohydrate metabolism. The genes gatB and gatC were up-regulated for galactose metabolism, and bglA was up-regulated in the glycolysis/gluconeogenesis pathway. The qRT-RCR results, which were in agreement with the RNA-seq, indicated the genes involved in the proliferation effect of LGG were up-regulated. UDP-glucose may be a key metabolite for MGO to promote LGG proliferation.
Collapse
|
18
|
Pan HY, Zhang CQ, Zhang XQ, Zeng H, Dong CH, Chen X, Ding K. A galacturonan from Dioscorea opposita Thunb. regulates fecal and impairs IL-1 and IL-6 expression in diarrhea mice. Glycoconj J 2022; 39:131-141. [DOI: 10.1007/s10719-022-10053-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 02/02/2022] [Accepted: 02/24/2022] [Indexed: 01/25/2023]
|
19
|
Rubin IMC, Stevnsborg L, Mollerup S, Petersen AM, Pinholt M. Bacteraemia caused by Lactobacillus rhamnosus given as a probiotic in a patient with a central venous catheter: a WGS case report. Infect Prev Pract 2022; 4:100200. [PMID: 35146406 PMCID: PMC8802096 DOI: 10.1016/j.infpip.2022.100200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/04/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Lactobacilli, especially Lactobacillus (L.) rhamnosus, are common and well-documented components of commercial probiotics [1]. Whole genome sequencing (WGS) is often used to compare bacterial genomes and their relatedness. In outbreak situations, it is used to investigate the transmission of pathogenic bacteria. WGS has also been used to determine safety in probiotics, by looking at potential virulence factors and resistance genes. Case presentation This case report describes a 56-year old multi-traumatised, immunocompetent woman who was given L. rhamnosus GG as a probiotic, and later developed a blood stream infection with L. rhamnosus GG. The patient was fed by a nasogastric tube, and she also had a central venous catheter for parenteral feeding. When the patient developed diarrhoea after long-term hospitalisation, she was given L. rhamnosus GG, as a probiotic, which was standard care on the ward where she was hospitalised. In this case report we describe the use of WGS to demonstrate that a patient fed with L. rhamnosus GG as a probiotic, developed a blood stream infection with the same strain. Conclusion In this case WGS was applied to show the relatedness of a probiotic and a pathogenic strain of L. rhamnosus GG. This case emphasises the need for caution when administering probiotics to patients with indwelling catheters. The patient was immunocompetent and she cleared the infection without the need for antibiotics.
Collapse
Affiliation(s)
- Ingrid Maria Cecilia Rubin
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Gastrounit, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Corresponding author. Address: Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.
| | - Lea Stevnsborg
- Department of Neurorehabilitation/Traumatic Brain Injury, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Sarah Mollerup
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Andreas Munk Petersen
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Gastrounit, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Mette Pinholt
- Department of Clinical Microbiology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| |
Collapse
|
20
|
Ivashkin V, Fomin V, Moiseev S, Brovko M, Maslennikov R, Ulyanin A, Sholomova V, Vasilyeva M, Trush E, Shifrin O, Poluektova E. Efficacy of a Probiotic Consisting of Lacticaseibacillus rhamnosus PDV 1705, Bifidobacterium bifidum PDV 0903, Bifidobacterium longum subsp. infantis PDV 1911, and Bifidobacterium longum subsp. longum PDV 2301 in the Treatment of Hospitalized Patients with COVID-19: a Randomized Controlled Trial. Probiotics Antimicrob Proteins 2021; 15:460-468. [PMID: 34643888 PMCID: PMC8512595 DOI: 10.1007/s12602-021-09858-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
The treatment of coronavirus disease (COVID-19) and COVID-19-associated diarrhea remains challenging. This study aimed to evaluate the efficacy of a multi-strain probiotic in the treatment of COVID-19. This was a randomized, controlled, single-center, open-label trial (NCT04854941). Inpatients with confirmed COVID-19 and pneumonia were randomly assigned to a group that received a multi-strain probiotic (PRO group) or to the control group (CON group). There were 99 and 101 patients in the PRO and CON groups, respectively. No significant differences in mortality, total duration of disease and hospital stay, incidence of intensive care unit admission, need for mechanical ventilation or oxygen support, liver injury development, and changes in inflammatory biomarker levels were observed between the PRO and CON groups among all included patients as well as among subgroups delineated based on age younger or older than 65 years, and subgroups with chronic cardiovascular diseases and diabetes. Diarrhea on admission was observed in 11.5% of patients; it resolved earlier in the PRO group than in the CON group (2 [1-4] vs. 4 [3-6] days; p = 0.049). Hospital-acquired diarrhea developed less frequently in the PRO group than in the CON group among patients who received a single antibiotic (0% vs. 12.5%; p = 0.023) unlike among those who received > 1 antibiotic (10.5% vs. 13.3%; p = 0.696). The studied probiotic had no significant effect on mortality and changes in most biomarkers in COVID-19. However, it was effective in treating diarrhea associated with COVID-19 and in preventing hospital-acquired diarrhea in patients who received a single antibiotic.
Collapse
Affiliation(s)
- Vladimir Ivashkin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| | - Victor Fomin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Sergey Moiseev
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Michail Brovko
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Roman Maslennikov
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation.
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation.
| | - Anatoly Ulyanin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| | - Victoria Sholomova
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Maria Vasilyeva
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Elizaveta Trush
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Oleg Shifrin
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
| | - Elena Poluektova
- Department of Internal Medicine, Gastroenterology and Hepatology, Sechenov University, Moscow, Russian Federation
- Scientific Community for Human Microbiome Research, Moscow, Russian Federation
| |
Collapse
|
21
|
van der Geest AM, Besseling-van der Vaart I, Schellinger-de Goede EM, van der Waal MB, Claassen E, Flach J, van de Burgwal LHM. Multispecies probiotics promote perceived human health and wellbeing: insights into the value of retrospective studies on user experiences. Benef Microbes 2021; 12:413-430. [PMID: 34455940 DOI: 10.3920/bm2020.0162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
When taking a broader perspective on the societal impact of probiotics, engagement of end-users is important to discover unmet needs, define relevant health benefits and identify key considerations for successful implementation in daily practice. This study therefore takes a retrospective approach and analyses a database of user experiences to review the effects of four multispecies probiotic formulations. The user experiences were analysed in a dependent sample manner (without control group) and complement previous randomised controlled trials that have been performed with the formulations. The database consisted of 584 evaluable user experiences regarding the impact of probiotic supplementation on perceived quality of life (QoL), gastrointestinal (GIT) symptoms and reported stool consistency after two weeks of consumption. Two different scales were used (n=344 in a 5-point scale; n=240 in a 10-point scale), which are presented as separate analyses. In the combined population of the 5-point-scale questionnaire, a significant increase in perceived QoL and a significant reduction in perceived GIT symptoms was observed. Descriptive summaries also indicate that diarrhoea- and constipation-like stool patterns are reduced following supplementation. Moreover, half of participants indicated that probiotic supplementation had a positive effect on their unmet medical need, and 64% of users were likely to continue using the product. Similar results were observed in the 10-point scale questionnaire. Considering the clinical relevance of probiotic supplementation in specific target groups, subgroup analyses were performed on participants who consumed the products for diarrhoea, constipation, Inflammatory Bowel Disease, Irritable Bowel Syndrome, and antibiotic usage. Overall, findings support the potential of probiotics to advance perceived human health and support the daily wellbeing of users. This systematic analysis of user experiences thereby contributes to the external validity of studies evaluating clinical effects of probiotics and increases knowledge on their societal impact.
Collapse
Affiliation(s)
- A M van der Geest
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | | | | | - M B van der Waal
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - E Claassen
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| | - J Flach
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands.,CR2O BV, Bisonspoor 3002-C701, 3605 LT Maarssen, the Netherlands
| | - L H M van de Burgwal
- Vrije Universiteit Amsterdam, Athena Institute, De Boelelaan 1085, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
22
|
Jo HG, Kim YS. Helicobacter pylori Eradication Therapy-associated Diarrhea. THE KOREAN JOURNAL OF HELICOBACTER AND UPPER GASTROINTESTINAL RESEARCH 2021. [DOI: 10.7704/kjhugr.2021.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Eradication of Helicobacter pylori has contributed to the treatment of peptic ulcers and mucosa-associated lymphoid tissue lymphoma. Moreover, it has possibly decreased the prevalence of gastric cancer. However, eradication therapy is associated with various adverse effects, of which diarrhea is the most common. The incidence of diarrhea after eradication treatment varies from 8% to 48%. In particular, the incidence is higher in patients who receive first-line standard triple therapy compared with those who receive second-line therapy. Both antibiotics and proton pump inhibitors, components of eradication therapy, have short-term and long-term impacts on gut microbiota. The alterations of gut microbiota may not recover until 1 year after eradication therapy. Most cases of diarrhea that occur after eradication therapy are antibiotic-associated diarrhea caused by the destruction of the normal gut microbiota. In some cases, Clostridioides difficile-associated diarrhea occurs after eradication therapy. If bloody diarrhea occurs after eradication therapy and the Clostridioides difficile toxin is not detected, antibiotic-associated hemorrhagic colitis associated with Klebsiella oxytoca infection should be suspected. It is crucial to explain the possibility of diarrhea before initiating eradication therapy to increase compliance. Furthermore, probiotics may be administered to reduce diarrhea. If severe diarrhea or symptoms other than the usual antibiotic-associated diarrhea occur during or after eradication therapy, antibiotics should be discontinued. In addition, appropriate tests to determine the cause of diarrhea should be performed. This review summarizes the alteration of the gut microbiota, the causes of diarrhea after Helicobacter pylori eradication therapy, and its management.
Collapse
|
23
|
HOSSAIN MN, RANADHEERA CS, FANG Z, AJLOUNI S. Healthy chocolate enriched with probiotics: a review. FOOD SCIENCE AND TECHNOLOGY 2021. [DOI: 10.1590/fst.11420] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
24
|
Duysburgh C, Van den Abbeele P, Morera M, Marzorati M. Lacticaseibacillus rhamnosus GG and Saccharomyces cerevisiae boulardii supplementation exert protective effects on human gut microbiome following antibiotic administration in vitro. Benef Microbes 2021; 12:59-73. [PMID: 34190033 DOI: 10.3920/bm2020.0180] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Antibiotic-induced dysbiosis of the microbial community has been associated with several gastrointestinal symptoms. The impact of repeated administration of Lacticaseibacillus rhamnosus GG (CNCM-I-4798) (formerly known as Lactobacillus rhamnosus GG), Saccharomyces cerevisiae boulardii (CNCM-I-1079) and their combination (associated in Smebiocta/Smectaflora Protect®) in supporting recovery of gut microbiota functionality and composition during and following amoxicillin:clavulanic acid administration was evaluated in vitro. Antibiotic dosage negatively affected SCFA production, coinciding with detrimental effects on Bacteroidetes, Firmicutes and Bifidobacterium spp. in the simulated proximal colon, while Akkermansia muciniphila was significantly reduced in the distal colon. L. rhamnosus GG and S. boulardii were able to thrive in both colon regions upon dosing, with S. boulardii even showing protective effects on the survival of L. rhamnosus GG during antibiotic administration. The impact of the probiotic strains on microbiome recovery revealed that supplementation with L. rhamnosus GG and/or S. boulardii resulted in a stimulating effect on the most abundant bacterial groups within the bacterial community of each donor. For one of the donors tested, co-dosing of L. rhamnosus GG and S. boulardii resulted in superior short-chain fatty acid recovery accompanied by a stronger increase in abundance of Bifidobacteriaceae. Overall, the current study provides first evidence that combined supplementation of L. rhamnosus GG and S. boulardii might be an interesting candidate in limiting detrimental effects of amoxicillin:clavulanic acid on the human gut microbiome, though further studies are warranted to confirm these findings.
Collapse
Affiliation(s)
- C Duysburgh
- ProDigest bv, Technologiepark 82, 9052 Ghent, Belgium
| | | | - M Morera
- IPSEN Consumer HealthCare SAS, 65 Quai Georges Gorse, 92650 Boulogne Billancourt Cedex, France
| | - M Marzorati
- ProDigest bv, Technologiepark 82, 9052 Ghent, Belgium.,Center of Microbial Ecology and Technology (CMET), Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| |
Collapse
|
25
|
Chen Y, Xiang Q, Liu L. Comparison of antibiotic-associated diarrhea caused by cefoperazone/sulbactam or piperacillin/tazobactam in neurosurgery patients. J Int Med Res 2021; 49:3000605211019661. [PMID: 34057835 PMCID: PMC8170296 DOI: 10.1177/03000605211019661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Objective To compare the occurrence and prognosis of antibiotic-associated diarrhea
(AAD) between patients treated with cefoperazone/sulbactam and
piperacillin/tazobactam in the neurosurgery department. Methods This study retrospectively analyzed patients who received
cefoperazone/sulbactam or piperacillin/tazobactam to prevent or treat
hospital-acquired infections in the Department of Neurosurgery of The First
Medical Center of Chinese PLA General Hospital between October 2019 and
October 2020. For patients with AAD, clinical data, antibiotic usage, the
incidence of diarrhea, treatment, and prognosis were collected and
analyzed. Results In total, 356 patients were enrolled, and 65 (18.6%) experienced AAD, 38
patients in the cefoperazone/sulbactam group and 27 patients in the
piperacillin/tazobactam group. The AAD rate did not differ between the
treatment arms. Conversely, the dosage, intensity, and duration of
antibiotic therapy differed between the groups, whereas no differences were
noted in the time to the appearance of diarrhea and prognosis. According to
regression analysis, the incidence of AAD did not differ between the groups
(odds ratio [OR] = 0.85, 95% confidence interval [CI] = 0.46–1.48). Conclusion Cefoperazone/sulbactam or piperacillin/tazobactam can lead to a similar
incidence rate of AAD. The combined application of antibiotics and empiric
therapy often occurs. The rational use of antibiotics should be
improved.
Collapse
Affiliation(s)
- Yue Chen
- Pharmacy Department, Medical Supplies Center of Chinese PLA General Hospital, Beijing, China
| | | | - Lei Liu
- Department of Neurosurgery, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
26
|
Kuwelker K, Langeland N, Löhr IH, Gidion J, Manyahi J, Moyo SJ, Blomberg B, Klingenberg C. Use of probiotics to reduce infections and death and prevent colonization with extended-spectrum beta-lactamase (ESBL)-producing bacteria among newborn infants in Tanzania (ProRIDE Trial): study protocol for a randomized controlled clinical trial. Trials 2021; 22:312. [PMID: 33926519 PMCID: PMC8082054 DOI: 10.1186/s13063-021-05251-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/07/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Extended-spectrum beta-lactamase-producing Enterobacteriaceae (ESBL-E) has emerged as an urgent global health threat and is by the World Health Organization ranked as priority 1 among pathogens in need of new treatment. Studies have shown high mortality in Tanzanian children with ESBL-E infections. Gut colonization of ESBL-E, which is a potential risk factor of ESBL-E infections, is reported to be very high among children in Tanzania. Probiotics may potentially reduce gut colonization of multidrug-resistant bacteria. However, there is limited data on whether probiotics may reduce ESBL-E carriage in infants. The ProRIDE Trial aims to evaluate whether the use of probiotics can reduce morbidity and mortality among infants in Haydom, Tanzania, and whether this effect is associated with a reduction in ESBL-E colonization and/or infections. METHODS/DESIGN This large randomized double-blinded placebo-controlled trial aims to recruit 2000 newborn infants at Haydom Lutheran Hospital and the surrounding area in the period of November 2020 to November 2021. Participants will be enrolled from days 0 to 3 after birth and randomized to receive probiotics or placebo for 4 weeks. Participants will be followed-up for 6 months, during which three visits will be made to collect clinical and demographic information, as well as rectal swabs and fecal samples which will be subjected to laboratory analysis. The primary composite outcome is the prevalence of death and/or hospitalization at 6 months of age. DISCUSSION As the use of probiotics may give a more favorable gut composition, and thereby improve health and reduce morbidity and mortality, the results may have implications for future therapy guidelines in Africa and internationally. TRIAL REGISTRATION ClinicalTrials.gov NCT04172012. Registered on November 21, 2019.
Collapse
Affiliation(s)
- Kanika Kuwelker
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Haukeland University Hospital, Haukelandsbakken, 5009 Bergen, Norway
| | - Nina Langeland
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Haukeland University Hospital, Haukelandsbakken, 5009 Bergen, Norway
- Department of Clinical Science, University of Bergen, Laboratory Building, Haukeland University Hospital, Jonas Lies veg 87, 5021 Bergen, Norway
| | - Iren Høyland Löhr
- Department of Medical Microbiology, Stavanger University Hospital, Gerd Ragna Bloch Thorsens gate, 4011 Stavanger, Norway
| | - Joshua Gidion
- Department of Paediatrics, Haydom Lutheran Hospital, Mbulu, Manyara Tanzania
| | - Joel Manyahi
- Department of Clinical Science, University of Bergen, Laboratory Building, Haukeland University Hospital, Jonas Lies veg 87, 5021 Bergen, Norway
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, MUHAS, P.O. Box 65005, Dar es Salaam, Tanzania
| | - Sabrina John Moyo
- Department of Clinical Science, University of Bergen, Laboratory Building, Haukeland University Hospital, Jonas Lies veg 87, 5021 Bergen, Norway
- Department of Microbiology and Immunology, Muhimbili University of Health and Allied Sciences, MUHAS, P.O. Box 65005, Dar es Salaam, Tanzania
| | - Bjørn Blomberg
- Norwegian National Advisory Unit on Tropical Infectious Diseases, Haukeland University Hospital, Haukelandsbakken, 5009 Bergen, Norway
- Department of Clinical Science, University of Bergen, Laboratory Building, Haukeland University Hospital, Jonas Lies veg 87, 5021 Bergen, Norway
| | - Claus Klingenberg
- Department of Paediatrics and Adolescence Medicine, University Hospital of North Norway, Tromsø, Norway
- Paediatric Research Group, Faculty of Health Sciences, University of Tromsø-Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
27
|
McFarland LV. Efficacy of Single-Strain Probiotics Versus Multi-Strain Mixtures: Systematic Review of Strain and Disease Specificity. Dig Dis Sci 2021; 66:694-704. [PMID: 32274669 DOI: 10.1007/s10620-020-06244-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/28/2020] [Indexed: 02/06/2023]
Abstract
The diversity of probiotic products makes choosing an appropriate probiotic challenging. One unanswered question is whether single-strain probiotics are more effective than multi-strain mixtures. The aim of this review is to account for both disease and strain specificity to determine whether single strains or multiple strains are equivalent or more effective. This literature review of randomized controlled trials from 1973 to 2019 was used to compare the pooled efficacy of trials with a single strain versus the probiotic mixture with same matched strain within the same type of disease indication. A total of 65 RCTs were included (41 with single strains, 22 multi-strain mixtures and 2 comparing single strain to mixture arms) for eight different disease indications (N = 10,863). Only three strains (L. rhamnosus GG, L. helveticus R52 and B. lactis Bb12) had corresponding trials with matching mixtures. Use of L. rhamnosus GG only was significantly more protective for necrotizing enterocolitis compared to two mixtures also containing different strains of B. lactis. The mixture of L. rhamnosus GG and B. lactis Bb12 was significantly more effective than L. rhamnosus GG alone for the eradication of H. pylori. In most cases, single strains were equivalent to mixtures. Choice of an appropriate probiotic should be based, not on the number of strains in the product, rather based on evidence-based trials of efficacy. In most cases, multi-strain mixtures were not significantly more effective than single-strain probiotics.
Collapse
Affiliation(s)
- Lynne V McFarland
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, 6047 38th Avenue NE, Seattle, WA, 98115, USA.
| |
Collapse
|
28
|
Imai S, Momo K, Kashiwagi H, Miyai T, Sugawara M, Takekuma Y. Probiotic Prescription Status of Pediatric Patients with Otitis Media Receiving Oral Amoxicillin or Amoxicillin/Clavulanate from April 2016 to March 2017 Using a Japanese Health Insurance Claims Database. Biol Pharm Bull 2021; 44:448-452. [PMID: 33642554 DOI: 10.1248/bpb.b20-00214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antibiotic-associated diarrhea (AAD) is a typical side effect of antibiotic treatment, especially in children. Amoxicillin (AMPC) and amoxicillin/clavulanate (AMPC/CVA) are associated with high risk of AAD; however, these antibiotics are important in the pediatric field. Recent research suggests that probiotics prevent pediatric AAD, including that caused by AMPC and AMPC/CVA. Indeed, guidelines for acute otitis media in children recommend the concomitant use of probiotics. However, the prescription status of probiotics for pediatric patients with otitis media receiving oral AMPC and AMPC/CVA remains unknown. We therefore conducted a survey to clarify the current status of these prescriptions and, in particular, to identify specific populations with a low proportion of probiotic prescriptions. Pediatric patients (≤15 years of age) newly prescribed oral AMPC or AMPC/CVA for otitis media between April 2016 and March 2017 were identified from a Japanese health insurance claims database. Eligible patients were divided into the AMPC (1303 patients) and AMPC/CVA (424 patients) groups, in which 659 (50.6%) and 293 (69.1%) patients were prescribed probiotics, respectively. Of the patients receiving probiotic prescriptions in the AMPC and AMPC/CVA groups, 632 (95.9%) and 286 (97.6%) patients received antibiotic-resistant probiotic prescriptions, respectively. When classified by the prescribing clinical department and patient age, the proportions of probiotic prescriptions in Internal Medicine and Pediatrics departments were lower than those in the Otorhinolaryngology department regardless of age. These results indicate the probability of insufficient probiotic prescriptions for pediatric patients with otitis media. Solving this issue may lead to the provision of safer antimicrobial therapy.
Collapse
Affiliation(s)
- Shungo Imai
- Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Kenji Momo
- Department of Hospital Pharmaceutics, School of Pharmacy, Showa University
| | | | | | - Mitsuru Sugawara
- Faculty of Pharmaceutical Sciences, Hokkaido University.,Department of Pharmacy, Hokkaido University Hospital
| | - Yoh Takekuma
- Department of Pharmacy, Hokkaido University Hospital
| |
Collapse
|
29
|
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 2021; 57:381-398. [PMID: 33331485 DOI: 10.1590/s0004-2803.202000000-72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [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
|
30
|
Hasosah M, Qurashi M, Balkhair A, Alzahrani Z, Alabbasi A, Alzahrani M, Alnahdi W, Shafei S, Bafaqih M, Khan M. Knowledge, attitudes, and understanding of probiotics among pediatricians in different regions of Saudi Arabia. BMC MEDICAL EDUCATION 2021; 21:68. [PMID: 33478488 PMCID: PMC7819255 DOI: 10.1186/s12909-021-02499-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 01/13/2021] [Indexed: 05/31/2023]
Abstract
BACKGROUND Probiotics are live microorganisms that, when administered in adequate amounts, confer a health benefit upon the host. Knowledge and attitudes of health professionals have been reported to be at a medium level for probiotics. The objective was to evaluate the knowledge and practice styles about probiotics among pediatricians working in different regions of Saudi Arabia. METHODS This cross-sectional study was conducted at pediatric hospitals in Saudi Arabia. A national survey of 550 pediatric providers (PPs) was conducted between January and March 2020 anonymously on their knowledge and practice styles regarding probiotics, and it was completed by pediatric residents (PRs), pediatric specialist (PSs), pediatric consultants (PCs), and pediatric gastroenterologists (PGs). RESULTS The survey had a response rate of 82%. Among the respondents, 57.7% were aware of the probiotic's definition. There were significant differences in the percentage of participants who had little knowledge of probiotics (P < 0.05), with the highest being PRs and the lowest being PGs. The most common probiotic used by all participants was Lactobacillus acidophilus (63.3%), and Mycobacterium avium was prescribed the least often (8.6%). Most PRs and PSs correctly reported that probiotics reduce the risk of antibiotic-induced diarrhea (74.9 and 80.2%, respectively), but there were no significant differences among them. CONCLUSIONS Significant differences in knowledge and practice patterns exist for probiotics. Identification of knowledge gaps may be useful to develop educational materials to improve the proper definition, knowledge, and use of probiotics.
Collapse
Affiliation(s)
- Mohammed Hasosah
- Pediatric Gastroenterology Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, Jeddah, Kingdom of Saudi Arabia.
| | - Mansour Qurashi
- Neoatology Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Abdullah Balkhair
- Pediatric Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, PO Box: 8202, Jeddah, 21482, Kingdom of Saudi Arabia
| | - Ziyad Alzahrani
- Pediatric Gastroenterology Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Abdullah Alabbasi
- Pediatric Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, PO Box: 8202, Jeddah, 21482, Kingdom of Saudi Arabia
| | - Muhanad Alzahrani
- Pediatric Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, PO Box: 8202, Jeddah, 21482, Kingdom of Saudi Arabia
| | - Wejdan Alnahdi
- Pediatric Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, PO Box: 8202, Jeddah, 21482, Kingdom of Saudi Arabia
| | - Sohaib Shafei
- Pediatric Gastroenterology Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Malak Bafaqih
- Pediatric Gastroenterology Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Muhammad Khan
- Pediatric Department, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center (KAIMRC), National Guard Hospital, PO Box: 8202, Jeddah, 21482, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Hu JS, Huang YY, Kuang JH, Yu JJ, Zhou QY, Liu DM. Streptococcus thermophiles DMST-H2 Promotes Recovery in Mice with Antibiotic-Associated Diarrhea. Microorganisms 2020; 8:microorganisms8111650. [PMID: 33114373 PMCID: PMC7693992 DOI: 10.3390/microorganisms8111650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) is the most common side effect of antibiotics and is routinely treated with probiotics in clinical. Streptococcus thermophiles, extensively utilized for producing dairy foods, has recently been regarded as a new promising probiotic candidate. In this study, the efficacy of Streptococcus thermophiles DMST-H2 (DMST-H2) for AAD treatment in mice was investigated. DMST-H2 was isolated from Chinese traditional yogurt, proved to be non-toxic, and presented tolerance against simulated gastrointestinal conditions in vitro. Additionally, genomic analysis revealed that it possessed genes related to acid tolerance, bile salt tolerance, adhesion, oxidative stress and bacteriocin production. The animal experiment results showed that both DMST-H2 treatment and natural recovery could reduce fecal water content. Compared with spontaneous recovery, DMST-H2 accelerated the recovery of the enlarged caecum and intestinal barrier injury from AAD, and further decreased endotoxin (ET), D-lactate (D-LA) and diamine oxidase (DAO) content in serum. Moreover, pro-inflammatory cytokines (TNF-α) were reduced, while interferon-γ (IFN-γ) and anti-inflammatory cytokines (IL-10) increased after treating with DMST-H2. Furthermore, DMST-H2 better restored the structure of intestinal flora. At the phylum level, Firmicutes increased and Proteobacteria decreased. These findings indicate that DMST-H2 could promote recovery in mice with antibiotic-associated diarrhea.
Collapse
|
32
|
Piper HG, Coughlin LA, Hussain S, Nguyen V, Channabasappa N, Koh AY. The Impact of Lactobacillus Probiotics on the Gut Microbiota in Children With Short Bowel Syndrome. J Surg Res 2020; 251:112-118. [DOI: 10.1016/j.jss.2020.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 01/22/2020] [Accepted: 01/31/2020] [Indexed: 12/22/2022]
|
33
|
Andrade JC, Almeida D, Domingos M, Seabra CL, Machado D, Freitas AC, Gomes AM. Commensal Obligate Anaerobic Bacteria and Health: Production, Storage, and Delivery Strategies. Front Bioeng Biotechnol 2020; 8:550. [PMID: 32582673 PMCID: PMC7291883 DOI: 10.3389/fbioe.2020.00550] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
In the last years several human commensals have emerged from the gut microbiota studies as potential probiotics or therapeutic agents. Strains of human gut inhabitants such as Akkermansia, Bacteroides, or Faecalibacterium have shown several interesting bioactivities and are thus currently being considered as food supplements or as live biotherapeutics, as is already the case with other human commensals such as bifidobacteria. The large-scale use of these bacteria will pose many challenges and drawbacks mainly because they are quite sensitive to oxygen and/or very difficult to cultivate. This review highlights the properties of some of the most promising human commensals bacteria and summarizes the most up-to-date knowledge on their potential health effects. A comprehensive outlook on the potential strategies currently employed and/or available to produce, stabilize, and deliver these microorganisms is also presented.
Collapse
Affiliation(s)
- José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Gandra, Portugal
| | - Diana Almeida
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Melany Domingos
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Catarina Leal Seabra
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Daniela Machado
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Cristina Freitas
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| | - Ana Maria Gomes
- CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Porto, Portugal
| |
Collapse
|
34
|
van Wietmarschen HA, Busch M, van Oostveen A, Pot G, Jong MC. Probiotics use for antibiotic-associated diarrhea: a pragmatic participatory evaluation in nursing homes. BMC Gastroenterol 2020; 20:151. [PMID: 32404062 PMCID: PMC7222499 DOI: 10.1186/s12876-020-01297-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Antibiotic-associated diarrhea (AAD) occurs in 2-25% of nursing home residents, which may lead to dehydration, malnutrition, severe complications and hospitalizations. Research shows that probiotics can be effective and safe in reducing AAD. However, probiotics are not routinely used in Dutch nursing homes. The objectives of this evaluation were to develop a procedure for the implementation of probiotics to prevent AAD in nursing homes, to evaluate effects on AAD occurrence, and to evaluate the implementation process of probiotics in daily care. METHODS A pragmatic participatory evaluation (PPE) design was chosen, as it seemed a suitable approach for implementation of probiotics, as well as for evaluation of its effectiveness in daily nursing home practice. Probiotics administration was implemented in three nursing homes of the Rivas Zorggroep for residents with somatic and/or psychogeriatric conditions. Ninety-three residents provided data on 167 episodes of antibiotics use, of which 84 episodes that included supplementation with probiotics and 83 episodes with no probiotics supplementation. A multispecies probiotics was administered twice daily upon start of antibiotic treatment, up to 1 week after completing the antibiotics course. The occurrence of AAD was monitored and a process evaluation was conducted to assess facilitators and barriers of probiotics implementation. RESULTS The number of episodes with AAD when using probiotics was significantly lower than when no probiotics was used (20% vs 36%; p = 0,022, Chi-square). No significant differences in the occurrence of AAD were found between the residents taking amoxicillin/clavulanic acid or ciprofloxacin. Reported facilitators for implementation were perceived benefits of probiotics and prescription by medical staff. Reported challenges were probiotics intake by residents and individual decision-making as to which resident would benefit from it. CONCLUSION Successful implementation of probiotics demonstrated the prevention of AAD in nursing home residents. TRIAL REGISTRATION ISRCTN 94786163, retrospectively registered on 3 February 2020.
Collapse
Affiliation(s)
- Herman A van Wietmarschen
- Department Nutrition & Health, Louis Bolk Institute, Kosterijland 3-5, 3981, Bunnik, AJ, The Netherlands.
| | - Martine Busch
- Van Praag Institute, Springweg 7, 3511, Utrecht, VH, The Netherlands
| | | | - Gerda Pot
- Department Nutrition & Health, Louis Bolk Institute, Kosterijland 3-5, 3981, Bunnik, AJ, The Netherlands
| | - Miek C Jong
- Department of Health Sciences, Mid Sweden University, Holmgatan 10, 851 70, Sundsvall, Sweden
| |
Collapse
|
35
|
European General Practitioners perceptions on probiotics: Results of a multinational survey. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
36
|
Mekonnen SA, Merenstein D, Fraser CM, Marco ML. Molecular mechanisms of probiotic prevention of antibiotic-associated diarrhea. Curr Opin Biotechnol 2020; 61:226-234. [PMID: 32087535 DOI: 10.1016/j.copbio.2020.01.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 02/08/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common and unintended adverse effect of antibiotic treatment. It is characterized by the disruption of the gut microbiota, decreased intestinal short chain fatty acid (SCFA) concentrations, accumulation of luminal carbohydrates and colonic bile acids, altered water absorption, and ultimately diarrhea. Probiotics were shown to prevent AAD in numerous clinical trials. This review examines what is currently known about how probiotics reduce the risk for AAD via modulating the gut microbiota, altering nutrient and bile acid metabolism, inducing epithelial solute transporter activity, supporting intestinal barrier function, and influencing the immune system. Although probiotics are frequently prescribed with antibiotic use, mechanistic evidence verifying how they confer protection against AAD is extremely limited. This information is urgently needed for improving recommendations for sustaining probiotic development and for implementing probiotics in clinical settings.
Collapse
Affiliation(s)
- Solomon A Mekonnen
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Claire M Fraser
- Department of Medicine, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, CA, USA.
| |
Collapse
|
37
|
Kessler C. The Florajen Digestion Balance Patient Experience Study. Health (London) 2020. [DOI: 10.4236/health.2020.1211107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Probiotics for Childhood Functional Gastrointestinal Disorders: Do We Know What We Advise? PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Fijan S, Frauwallner A, Varga L, Langerholc T, Rogelj I, Lorber M, Lewis P, Povalej Bržan P. Health Professionals' Knowledge of Probiotics: An International Survey. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16173128. [PMID: 31466273 PMCID: PMC6747149 DOI: 10.3390/ijerph16173128] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/24/2019] [Accepted: 08/26/2019] [Indexed: 12/18/2022]
Abstract
The objective of this study was to survey health professionals to investigate their knowledge of probiotics. An online survey was conducted to gather data on the knowledge of health professionals. The online survey was distributed via email and social media platforms using snowball sampling. A total of 1066 health professionals (859; 80.6% female) from 30 countries responded to the survey. Most of the respondents evaluated their knowledge of probiotics as medium (36.4%) or good (36.2%). Only 8.9% of the respondents rated it as excellent. No statistical difference in knowledge was found between male and female health professionals. Over 80% of pharmacists, allied health professionals, medical doctors and dentists, and other health professionals knew the correct definition of probiotics as “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host”, whereas three quarters of registered nurses and midwives and less than two thirds of psychologists identified the correct definition. Statistically, more female than male health professionals knew the correct definition of probiotics. The most frequently recognized species of bacteria containing probiotic strains were Lactobacillus acidophilus (92%), Bifidobacterium bifidum (82%), and Lactobacillus rhamnosus (62%). The opinions on when it is best to take probiotics were different (χ2 = 28.375; p < 0.001), with 90.2% of respondents identifying that probiotics have beneficial effects if taken during antibiotic therapy, 83.5% for diarrhea, 70.6% for constipation, 63.3% before traveling abroad, and 60.4% for treating allergies. Almost 79% of health professionals involved in this study have advised their patients to use probiotics and 57.5% of the respondents wanted to learn more about probiotics. All things considered, health professionals have a medium level of knowledge of probiotics, which could be improved by the implementation of targeted learning programs. As probiotics have many beneficial effects in a wide range of health areas, health professionals need to adopt the use of probiotics in clinical practice.
Collapse
Affiliation(s)
- Sabina Fijan
- Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia.
| | - Anita Frauwallner
- Institut Allergosan, Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, Gmeinstrasse 13, 8055 Graz, Austria
| | - László Varga
- Department of Food Science, Faculty of Agricultural and Food Sciences, Széchenyi István University, Lucsony u. 15-17., 9200 Mosonmagyaróvár, Hungary
| | - Tomaž Langerholc
- Department of Microbiology, Biochemistry, Molecular Biology and Biotechnology, Faculty of Agriculture and Life Sciences, University of Maribor, Pivola 10, 2311 Hoče, Slovenia
| | - Irena Rogelj
- Institute of Dairy Science and Probiotics, Biotechnical Faculty, University of Ljubljana, Groblje 3, 1230 Domžale, Slovenia
| | - Mateja Lorber
- Faculty of Health Sciences, University of Maribor, Žitna ulica 15, 2000 Maribor, Slovenia
| | - Peter Lewis
- School of Nursing and Midwifery, University of Western Sydney, Locked Bag 1797, Penrith, NSW 2751, Australia
| | - Petra Povalej Bržan
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
- Faculty of Electrical Engineering and Computer Science, University of Maribor, Koroška cesta 46, 2000 Maribor, Slovenia
| |
Collapse
|
40
|
Santacroce L, Charitos IA, Bottalico L. A successful history: probiotics and their potential as antimicrobials. Expert Rev Anti Infect Ther 2019; 17:635-645. [PMID: 31318576 DOI: 10.1080/14787210.2019.1645597] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Probiotics are living, non-pathogenic microorganisms (bacteria) that enter through diet in the human body, live during their passage through the gastrointestinal (GI) tract and are beneficial to health. They have become popular in recent years as a way of improving human health through nutrition. This review aims to discuss the efficacy of probiotics for the supportive therapy of certain clinical conditions, especially infectious diseases, as reported in a number of studies, even though some concerns about their safety still remain. Areas covered: This paper will review the history of probiotics, from ancient ages to date, and the evolution of their use in clinical practice. The study is based on both personal professional experience of the authors and a comprehensive literature analysis, including old documents from libraries, searching the related biological and clinical data on Scopus, Web of Science, PubMed, EMBASE, also using the 'cited by' and 'similar articles' options available in PubMed. Expert opinion: Not all researchers agree about the safety and real efficacy of probiotics in common conditions, especially infective diseases. However, the use of probiotics for clinical conditions that may be improved by consumption of these dietary supplements should be considered as a possible supportive therapy in select patients.
Collapse
Affiliation(s)
- Luigi Santacroce
- a Ionian Department (DJSGEM), Microbiology and Virology Laboratory, University of Bari , Bari , Italy
| | | | | |
Collapse
|
41
|
Chambers L, Avery A, Dalrymple J, Farrell L, Gibson G, Harrington J, Rijkers G, Rowland I, Spiro A, Varela‐Moreiras G, Vokes L, Younge L, Whelan K, Stanner S. Translating probiotic science into practice. NUTR BULL 2019. [DOI: 10.1111/nbu.12385] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - A. Avery
- University of Nottingham Nottingham UK
| | - J. Dalrymple
- Primary Care Society for Gastroenterology London UK
| | | | | | | | - G. Rijkers
- University College Roosevelt Middelburg The Netherlands
| | | | - A. Spiro
- British Nutrition Foundation London UK
| | | | - L. Vokes
- British Dietetic Association Birmingham UK
| | | | | | | |
Collapse
|
42
|
Yang B, Zhang Y, Li B, Zou Y, Xiao C. Fine particulate matter alters the microecology of the murine respiratory tract. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:8623-8632. [PMID: 30707384 DOI: 10.1007/s11356-019-04372-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Accepted: 01/24/2019] [Indexed: 06/09/2023]
Abstract
Fine particulate matter is a global challenge to human health. We investigated the effects and potential mechanisms of fine particulate matter on respiratory tract microecology in a lung injury mouse model. BALB/c mice were randomized into exposed and control groups. We found that the levels of soluble tumor necrosis factor receptor I was increased following the PM2.5 exposure. 16S rRNA sequencing of respiratory tract lavage fluid confirmed that the composition of the respiratory tract microecology was altered by the exposure. Lactobacillus was the most abundant of bacterial species present. Collectively, these results establish a link between exposure to fine particulate matter and alterations to the respiratory tract microecology. Elucidation of the underlying mechanisms may lead to treatment strategies in lung injury.
Collapse
Affiliation(s)
- Biao Yang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, Liao Ning, People's Republic of China
| | - Yu Zhang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, Liao Ning, People's Republic of China
| | - Bingyu Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, Liao Ning, People's Republic of China
| | - Yang Zou
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, Liao Ning, People's Republic of China
| | - Chunling Xiao
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huanghe North Street, Shenyang, Liao Ning, People's Republic of China.
| |
Collapse
|