1
|
Dricot CEMK, Erreygers I, Cauwenberghs E, De Paz J, Spacova I, Verhoeven V, Ahannach S, Lebeer S. Riboflavin for women's health and emerging microbiome strategies. NPJ Biofilms Microbiomes 2024; 10:107. [PMID: 39420006 PMCID: PMC11486906 DOI: 10.1038/s41522-024-00579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Riboflavin (vitamin B2) is an essential water-soluble vitamin that serves as a precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). FMN and FAD are coenzymes involved in key enzymatic reactions in energy metabolism, biosynthesis, detoxification and electron scavenging pathways. Riboflavin deficiency is prevalent worldwide and impacts women's health due to riboflavin demands linked to urogenital and reproductive health, hormonal fluctuations during the menstrual cycle, pregnancy, and breastfeeding. Innovative functional foods and nutraceuticals are increasingly developed to meet women's riboflavin needs to supplement dietary sources. An emerging and particularly promising strategy is the administration of riboflavin-producing lactic acid bacteria, combining the health benefits of riboflavin with those of probiotics and in situ riboflavin production. Specific taxa of lactobacilli are of particular interest for women, because of the crucial role of Lactobacillus species in the vagina and the documented health effects of other Lactobacillaceae taxa in the gut and on the skin. In this narrative review, we synthesize the underlying molecular mechanisms and clinical benefits of riboflavin intake for women's health, and evaluate the synergistic potential of riboflavin-producing lactobacilli and other microbiota.
Collapse
Affiliation(s)
- Caroline E M K Dricot
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Isabel Erreygers
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Eline Cauwenberghs
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Jocelyn De Paz
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Irina Spacova
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Veronique Verhoeven
- Department of Family Medicine and Population Health, University of Antwerp, Antwerp, Belgium
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium
| | - Sarah Ahannach
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium.
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
2
|
Wurm J, Curtis N, Zimmermann P. The effect of antibiotics on the intestinal microbiota in children - a systematic review. FRONTIERS IN ALLERGY 2024; 5:1458688. [PMID: 39435363 PMCID: PMC11491438 DOI: 10.3389/falgy.2024.1458688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Background Children are the age group with the highest exposure to antibiotics (ABX). ABX treatment changes the composition of the intestinal microbiota. The first few years of life are crucial for the establishment of a healthy microbiota and consequently, disturbance of the microbiota during this critical period may have far-reaching consequences. In this review, we summarise studies that have investigated the effect of ABX on the composition of the intestinal microbiota in children. Methods According to the PRISMA guidelines, a systematic search was done using MEDLINE and Embase to identify original studies that have investigated the effect of systemic ABX on the composition of the intestinal microbiota in children. Results We identified 89 studies investigating a total of 9,712 children (including 4,574 controls) and 14,845 samples. All ABX investigated resulted in a reduction in alpha diversity, either when comparing samples before and after ABX or children with ABX and controls. Following treatment with penicillins, the decrease in alpha diversity persisted for up to 6-12 months and with macrolides, up to the latest follow-up at 12-24 months. After ABX in the neonatal period, a decrease in alpha diversity was still found at 36 months. Treatment with penicillins, penicillins plus gentamicin, cephalosporins, carbapenems, macrolides, and aminoglycosides, but not trimethoprim/sulfamethoxazole, was associated with decreased abundances of beneficial bacteria including Actinobacteria, Bifidobacteriales, Bifidobacteriaceae, and/or Bifidobacterium, and Lactobacillus. The direction of change in the abundance of Enterobacteriaceae varied with ABX classes, but an increase in Enterobacteriaceae other than Escherichia coli was frequently observed. Conclusion ABX have profound effects on the intestinal microbiota of children, with notable differences between ABX classes. Macrolides have the most substantial impact while trimethoprim/sulfamethoxazole has the least pronounced effect.
Collapse
Affiliation(s)
- Juliane Wurm
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Health Science and Medicine, University Lucerne, Lucerne, Switzerland
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Infectious Diseases Unit, The Royal Children’s Hospital Melbourne, Parkville, VIC, Australia
| | - Petra Zimmermann
- Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
3
|
Sakaryalı Uyar D, Üsküdar Güçlü A, Çelik E, Memiş Özgül B, Altay Koçak A, Başustaoğlu AC. Evaluation of probiotics' efficiency on cariogenic bacteria: randomized controlled clinical study. BMC Oral Health 2024; 24:886. [PMID: 39095860 PMCID: PMC11297621 DOI: 10.1186/s12903-024-04659-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Probiotics are live beneficial bacteria to human health and their efficiency on oral health is still being investigated. The purpose of this study was to evaluate the level of Streptococcus mutans and Lactobacillus species with and without the use of probiotics for six-months after the treatment of all dental caries under general anesthesia. METHODS Fifty-eight pediatric patients without any systemic diseases, whose dental treatments were completed under general anesthesia (GA), were included in the study. The patients were recruited in two-groups; Group A: Patients started using probiotics after GA and Group B: Patients did not use probiotics after GA. Saliva samples were taken from all patients on the day before GA (T0), at one-month (T1), three-month (T2) and six-month (T3) follow-up after GA. The counts of cariogenic bacteria were determined by the analysis of saliva samples using real-time polymerase chain reaction. Statistical significance level was accepted as p < 0.05. RESULTS There was statistically significant difference between Group A and B for T0, T1, T2 and T3 regarding S. mutans (p = 0.001, p = 0.04, p = 0.04, p = 0.03; p < 0.05). However, there was no statistically significant difference between groups regarding Lactobacillus species (p ≥ 0.05). CONCLUSIONS Probiotic use and treatment of all caries significantly reduced the level of S. mutans but not Lactobacillus species. Furthermore, S. mutans decreased after cessation of probiotics, but it was not statistically significant. TRIAL REGISTRATION Study was registered as "Effects of Probiotics on Streptococcus mutans and Lactobacillus species" with the registration number of NCT05859646 (16/05/2023) at https://www. CLINICALTRIALS gov Protocol Registration and Results System.
Collapse
Affiliation(s)
- Didem Sakaryalı Uyar
- Department of Pediatric Dentistry, Faculty of Dentistry, Başkent University, Ankara, Türkiye.
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye.
| | - Aylin Üsküdar Güçlü
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| | - Ekin Çelik
- Department of Medical Biology, Faculty of Medicine, Kırşehir Ahi Evran University, Kırşehir, Türkiye
| | - Betül Memiş Özgül
- Department of Pediatric Dentistry, Faculty of Dentistry, Lokman Hekim University, Ankara, Türkiye
| | - Aylin Altay Koçak
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| | - Ahmet Celal Başustaoğlu
- Department of Medical Microbiology, Faculty of Medicine, Başkent University, Ankara, Türkiye
| |
Collapse
|
4
|
Kizilbash SJ, Connolly H, Bartosh S, Zahr R, Al-Akash S, Chishti A, Mansuri A, Tawadrous H, Jain NG. Probiotic use in pediatric kidney transplant recipients: What are current practices, and are they evidence-based? A pediatric nephrology research consortium study. Pediatr Transplant 2024; 28:e14790. [PMID: 38837638 DOI: 10.1111/petr.14790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/06/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Probiotics are living microorganisms that may confer health benefits to their host if administered in sufficient quantities. However, data on the use of probiotics in transplant recipients are scarce. METHOD This multi-center survey of pediatric nephrologists aimed to examine variations in practice regarding the use of probiotics in pediatric kidney transplant recipients. The survey was conducted via a 10-item questionnaire (developed in Survey Monkey) administered to pediatric nephrologists participating in the Pediatric Nephrology Research Consortium meeting in April 2023. RESULTS Sixty-four pediatric nephrologists completed the survey. Twenty-seven (42.2%) respondents reported having prescribed probiotics to pediatric kidney transplant recipients. The primary reason for probiotic use was the treatment of antibiotic-associated diarrhea (n = 20), with other reasons including recurrent Clostridium difficile infection (n = 15), general gut health promotion (n = 12), recurrent urinary tract infections (n = 8), and parental request (n = 1). Of those who prescribed probiotics, 48.1% held them during periods of neutropenia and 14.8% during central venous line use. Of the 64 respondents, 20 reported the lack of safety data as a concern for using probiotics in kidney transplant recipients. CONCLUSION Pediatric nephrologists are increasingly prescribing probiotics to pediatric kidney transplant recipients; nevertheless, substantial practice variations exist. The paucity of safety data is a significant deterrent to probiotic use in this population.
Collapse
Affiliation(s)
- S J Kizilbash
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - H Connolly
- Department of Pediatrics, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - S Bartosh
- Department of Pediatrics, University of Wisconsin, Madison, Wisconsin, USA
| | - R Zahr
- Department of Pediatrics, University of Tennessee Health Science Center Memphis, Memphis, Tennessee, USA
| | - S Al-Akash
- Department of Pediatrics, McGovern Medical School at UTHealth Houston, Children's Memorial Hermann Hospital, Houston, Texas, USA
| | - A Chishti
- Department of Pediatrics, University of Kentucky, Lexington, Kentucky, USA
| | - A Mansuri
- Department of Pediatrics, Children's Hospital of Georgia, Medical college of Georgia, Augusta University, Augusta, Georgia, USA
| | - H Tawadrous
- Department of Pediatrics, Connecticut Children's Medical Center, Hartford, Connecticut, USA
| | - N G Jain
- Department of Pediatrics, Hackensack University Medical Center, Hackensack, New Jersey, USA
| |
Collapse
|
5
|
Mego M, Kasperova B, Chovanec J, Danis R, Reckova M, Bystricky B, Konkolovsky P, Jurisova S, Porsok S, Vaclav V, Wagnerova M, Stresko M, Brezinova B, Sutekova D, Ciernikova S, Svetlovska D, Drgona L. The beneficial effect of probiotics in the prevention of irinotecan-induced diarrhea in colorectal cancer patients with colostomy: a pooled analysis of two probiotic trials (Probio-SK-003 and Probio-SK-005) led by Slovak Cooperative Oncology Group. Front Oncol 2024; 14:1438657. [PMID: 39104721 PMCID: PMC11298351 DOI: 10.3389/fonc.2024.1438657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Background Probiotics could decrease irinotecan-induced diarrhea due to the reduction of intestinal beta-d-glucuronidase activity. This study included a combined analysis of two clinical trials aimed to determine the effectiveness of the probiotics in the prophylaxis of irinotecan-induced diarrhea in metastatic colorectal cancer (CRC) patients. Methods This combined analysis included 46 patients with CRC enrolled in the Probio-SK-003 (NCT01410955) and 233 patients from Probio-SK-005 (NCT02819960) starting a new line of irinotecan-based therapy with identical eligibility criteria. Patients were randomized in a ratio 1:1 to probiotic formulas vs. placebo administered for 12 and 6 weeks, respectively. Due to the different durations of study treatments, only the first 6 weeks of therapy were used for analysis. Results In total, 279 patients were randomized, including 142 patients in the placebo and 137 participants in the probiotic arm. Administration of probiotics did not significantly reduce the incidence of grade 3/4 diarrhea compared to placebo (placebo 12.7% vs. probiotics 6.6%, p = 0.11). Neither the overall incidence of diarrhea (placebo 48.6% vs. probiotics 41.6%, p = 0.28) nor the incidence of enterocolitis (placebo 4.2% vs. probiotics 0.7%, p = 0.12) was different in the placebo vs. probiotic arm. However, subgroup analysis revealed that patients with a colostomy who received a placebo had a significantly higher incidence of any diarrhea (placebo 51.2% vs. probiotics 25.7%, p = 0.028) and grade 3/4 diarrhea (placebo 14.6% vs. probiotics 0.0%, p = 0.03) compared to the probiotic arm. Conclusions This combined analysis suggests that probiotics could be beneficial in the prevention of irinotecan-induced diarrhea in colorectal cancer patients with colostomy.
Collapse
Affiliation(s)
- Michal Mego
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Barbora Kasperova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Jozef Chovanec
- Department of Oncology, St. Jacob Hospital, Bardejov, Slovakia
| | - Radoslav Danis
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Maria Reckova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
- Department of Oncology, Regional Cancer Center, Poprad, Slovakia
| | | | | | - Silvia Jurisova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Stefan Porsok
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Vladimir Vaclav
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Maria Wagnerova
- Department of Oncology, East Slovakia Comprehensive Cancer Center, Kosice, Slovakia
| | - Marian Stresko
- Department of Oncology, Faculty Hospital, Trnava, Slovakia
| | | | - Dagmar Sutekova
- Department of Oncology, University Hospital Martin, Martin, Slovakia
| | - Sona Ciernikova
- Biomedical Research Center, Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniela Svetlovska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Lubos Drgona
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
6
|
Heston SM, Hurst JH, Kelly MS. Understanding the influence of the microbiome on childhood infections. Expert Rev Anti Infect Ther 2024; 22:529-545. [PMID: 38605646 PMCID: PMC11464204 DOI: 10.1080/14787210.2024.2340664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
INTRODUCTION The microbiome is known to have a substantial impact on human health and disease. However, the impacts of the microbiome on immune system development, susceptibility to infectious diseases, and vaccine-elicited immune responses are emerging areas of interest. AREAS COVERED In this review, we provide an overview of development of the microbiome during childhood. We highlight available data suggesting that the microbiome is critical to maturation of the immune system and modifies susceptibility to a variety of infections during childhood and adolescence, including respiratory tract infections, Clostridioides difficile infection, and sexually transmitted infections. We discuss currently available and investigational therapeutics that have the potential to modify the microbiome to prevent or treat infections among children. Finally, we review the accumulating evidence that the gut microbiome influences vaccine-elicited immune responses among children. EXPERT OPINION Recent advances in sequencing technologies have led to an explosion of studies associating the human microbiome with the risk and severity of infectious diseases. As our knowledge of the extent to which the microbiome influences childhood infections continues to grow, microbiome-based diagnostics and therapeutics will increasingly be incorporated into clinical practice to improve the prevention, diagnosis, and treatment of infectious diseases among children.
Collapse
Affiliation(s)
- Sarah M Heston
- Pediatrics, Duke University School of Medicine, Durham, NC, UK
| | - Jillian H Hurst
- Pediatrics, Duke University School of Medicine, Durham, NC, UK
| | - Matthew S Kelly
- Pediatrics, Duke University School of Medicine, Durham, NC, UK
| |
Collapse
|
7
|
Hoenders R, Ghelman R, Portella C, Simmons S, Locke A, Cramer H, Gallego-Perez D, Jong M. A review of the WHO strategy on traditional, complementary, and integrative medicine from the perspective of academic consortia for integrative medicine and health. Front Med (Lausanne) 2024; 11:1395698. [PMID: 38933107 PMCID: PMC11201178 DOI: 10.3389/fmed.2024.1395698] [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: 03/04/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Despite important progress in modern medicine, widely regarded as an indispensable foundation of healthcare in all highly advanced nations and regions, not all patients respond well to available treatments in biomedicine alone. Additionally, there are concerns about side effects of many medications and interventions, the unsustainable cost of healthcare and the low resolution of chronic non-communicable diseases and mental disorders whose incidence has risen in the last decades. Besides, the chronic stress and burnout of many healthcare professionals impairs the therapeutic relationship. These circumstances call for a change in the current paradigm and practices of biomedicine healthcare. Most of the world population (80%) uses some form of traditional, complementary, and integrative medicine (T&CM), usually alongside biomedicine. Patients seem equally satisfied with biomedicine and T&CM, but in the field of T&CM there are also many challenges, such as unsupported claims for safety and/or efficacy, contamination of herbal medicines and problems with regulation and quality standards. As biomedicine and T&CM seem to have different strengths and weaknesses, integration of both approaches may be beneficial. Indeed, WHO has repeatedly called upon member states to work on the integration of T&CM into healthcare systems. Integrative medicine (IM) is an approach that offers a paradigm for doing so. It combines the best of both worlds (biomedicine and T&CM), based on evidence for efficacy and safety, adopting a holistic personalized approach, focused on health. In the last decades academic health centers are increasingly supportive of IM, as evidenced by the foundation of national academic consortia for integrative medicine in Brazil (2017), the Netherlands (2018), and Germany (2024) besides the pioneering American consortium (1998). However, the integration process is slow and sometimes met with criticism and even hostility. The WHO T&CM strategies (2002-2005 and 2014-2023) have provided incipient guidance on the integration process, but several challenges are yet to be addressed. This policy review proposes several possible solutions, including the establishment of a global matrix of academic consortia for IM, to update and extend the WHO T&CM strategy, that is currently under review.
Collapse
Affiliation(s)
- Rogier Hoenders
- Dutch Consortium for Integrative Care and Health, Center for Integrative Psychiatry, Lentis, Groningen, The Netherlands and Faculty of Religion, Culture and Society, University of Groningen, Groningen, Netherlands
| | - Ricardo Ghelman
- Brazilian Academic Consortium for Integrative Health and Department of Medicine on Primary Care, Faculty of Medicine Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Caio Portella
- Brazilian Academic Consortium for Integrative Health and Universidade de São Paulo, Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Faculdade de Medicina FMUSP, São Paulo, Brazil
| | - Samantha Simmons
- Academic Consortium for Integrative Medicine and Health, Lake Oswego, OR, United States
| | - Amy Locke
- Academic Consortium for Integrative Medicine and Health and Department of Family and Preventive Medicine University of Utah Health, Salt Lake City, UT, United States
| | - Holger Cramer
- Academic Consortium for Traditional & Integrative Medicine and Health, Germany and Institute of General Practice and Interprofessional Care, University Hospital Tübingen, Tübingen, Germany and Robert Bosch Center for Integrative Medicine and Health, Bosch Health Campus, Stuttgart, Germany
| | - Daniel Gallego-Perez
- Physical Medicine and Rehabilitation Department University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Miek Jong
- National Research Center in Complementary and Alternative Medicine (NAFKAM), Department of Community Medicine, Faculty of Health Sciences, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
8
|
Zhao W, Ren A, Shan S, Li Z, Su R, Yang R, Zhai F, Wu L, Tang Z, Yang J, Yue L. Inhibitory Effects of Soluble Dietary Fiber from Foxtail Millet on Colorectal Cancer by the Restoration of Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12130-12145. [PMID: 38748495 DOI: 10.1021/acs.jafc.4c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Colorectal cancer (CRC) is a common malignant tumor that occurs in the colon. Gut microbiota is a complex ecosystem that plays an important role in the pathogenesis of CRC. Our previous studies showed that the soluble dietary fiber of foxtail millet (FMB-SDF) exhibited significant antitumor activity in vitro. The present study evaluated the anticancer potential of FMB-SDF in the azoxymethane (AOM)- and dextran sodium sulfate (DSS)-induced mouse CRC models. The results showed that FMB-SDF could significantly alleviate colon cancer symptoms in mice. Further, we found that FMB-SDF consumption significantly altered gut microbiota diversity and the overall structure and regulated the abundance of some microorganisms in CRC mice. Meanwhile, KEGG pathway enrichment showed that FMB-SDF can also alleviate the occurrence of colon cancer in mice by regulating certain cancer-related signaling pathways. In conclusion, our findings may provide a novel approach for the prevention and biotherapy of CRC.
Collapse
Affiliation(s)
- Wenjing Zhao
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
- Shanxi Academy of Agricultural Sciences, Taiyuan 030031, China
| | - Aiqi Ren
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Shuhua Shan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, China
| | - Ruijun Su
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Ruipeng Yang
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Feihong Zhai
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Lihua Wu
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Zhaohui Tang
- Shanxi Academy of Agricultural Sciences, Taiyuan 030031, China
| | - Jieya Yang
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| | - Linzhong Yue
- Biological Science and Technology Colledge, Taiyuan Normal University, Jinzhong 030619, China
| |
Collapse
|
9
|
Alnezary FS, Alamri AR, Alrehaili RD, Alnizari DS, Alzahrani F, Mahmoud M, Almutairi MS, Kurdi A, Godman B. Managing infectious diarrhea among young children in community pharmacies in Saudi Arabia and the implications for AMR. Front Pediatr 2024; 12:1342493. [PMID: 38562134 PMCID: PMC10982503 DOI: 10.3389/fped.2024.1342493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 03/01/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Diarrhea remains a major global health issue for children under five, contributing substantially to morbidity and mortality. Community pharmacists play a pivotal role in the management of these children; however, their competence in managing childhood diarrhea in Saudi Arabia is under-researched. This is important to ensure optimal patient care. Method Simulated patients (SPs) presenting with three pediatric diarrhea scenarios were used to evaluate pharmacists' practice in terms of their counselling, history taking, over-the-counter (OTC) prescribing, medication instructions, diet/fluid advice, and/or information provision. Pharmacists' practice was categorized into adequate, less adequate, and poor. Results 182 community pharmacists, primarily male and non-Saudi, participated in the study, of which 60% were in chain pharmacies. Only 5% showed adequate practice in currently managing pediatric diarrhea. Of the 182 simulated patient visits, 62% received medication in all three scenarios and 20% were referred to physicians, with 16% of pharmacists failing to provide any form of intervention. The main medications recommended were kaolin (34%), pectin (34%) and metronidazole (11%). While most pharmacists (86%) asked about the patient's identity and age, 15% provided incorrect management information, 16% failed to provide guidance on the prescribed medicines, and 18% dispensed antimicrobials without a valid prescription. Conclusion A high level of inadequate management of pediatric diarrhea in Saudi Arabia was observed. This highlights the need for extensive training to improve community pharmacists' practice in service delivery including providing counselling and advice on the appropriate management of childhood diarrhea. The latter is particularly important to reduce antimicrobial resistance.
Collapse
Affiliation(s)
- Faris S. Alnezary
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Amira R. Alamri
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Rafa D. Alrehaili
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Dina S. Alnizari
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Fahad Alzahrani
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mansour Mahmoud
- Department of Pharmacy Practice, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Masaad S. Almutairi
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Amanj Kurdi
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
- Department of Clinical Pharmacy, College of Pharmacy, Hawler Medical University, Erbil, Iraq
- Department of Clinical Pharmacy, College of Pharmacy, Al-Kitab University, Kirkuk, Iraq
| | - Brian Godman
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
- Department of Public Health Pharmacy and Management, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, South Africa
| |
Collapse
|
10
|
Waitzberg D, Guarner F, Hojsak I, Ianiro G, Polk DB, Sokol H. Can the Evidence-Based Use of Probiotics (Notably Saccharomyces boulardii CNCM I-745 and Lactobacillus rhamnosus GG) Mitigate the Clinical Effects of Antibiotic-Associated Dysbiosis? Adv Ther 2024; 41:901-914. [PMID: 38286962 PMCID: PMC10879266 DOI: 10.1007/s12325-024-02783-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024]
Abstract
Dysbiosis corresponds to the disruption of a formerly stable, functionally complete microbiota. In the gut, this imbalance can lead to adverse health outcomes in both the short and long terms, with a potential increase in the lifetime risks of various noncommunicable diseases and disorders such as atopy (like asthma), inflammatory bowel disease, neurological disorders, and even behavioural and psychological disorders. Although antibiotics are highly effective in reducing morbidity and mortality in infectious diseases, antibiotic-associated diarrhoea is a common, non-negligible clinical sign of gut dysbiosis (and the only visible one). Re-establishment of a normal (functional) gut microbiota is promoted by completion of the clinically indicated course of antibiotics, the removal of any other perturbing external factors, the passage of time (i.e. recovery through the microbiota's natural resilience), appropriate nutritional support, and-in selected cases-the addition of probiotics. Systematic reviews and meta-analyses of clinical trials have confirmed the strain-specific efficacy of some probiotics (notably the yeast Saccharomyces boulardii CNCM I-745 and the bacterium Lactobacillus rhamnosus GG) in the treatment and/or prevention of antibiotic-associated diarrhoea in children and in adults. Unusually for a probiotic, S. boulardii is a eukaryote and is not therefore directly affected by antibiotics-making it suitable for administration in cases of antibiotic-associated diarrhoea. A robust body of evidence from clinical trials and meta-analyses shows that the timely administration of an adequately dosed probiotic (upon initiation of antibiotic treatment or within 48 h) can help to prevent or resolve the consequences of antibiotic-associated dysbiosis (such as diarrhoea) and promote the resilience of the gut microbiota and a return to the pre-antibiotic state. A focus on the prescription of evidence-based, adequately dosed probiotics should help to limit unjustified and potentially ineffective self-medication.
Collapse
Affiliation(s)
- Dan Waitzberg
- Department of Gastroenterology, LIM-35, School of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Iva Hojsak
- Referral Centre for Pediatric Gastroenterology and Nutrition, School of Medicine, University of Zagreb, Zagreb, Croatia
- University of Zagreb Medical School, Zagreb, Croatia
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie Dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - D Brent Polk
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, San Diego, and Rady Children's Hospital, University of California, San Diego, CA, USA
| | - Harry Sokol
- Gastroenterology Department, Saint-Antoine Hospital, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, 184 Rue du Faubourg Saint-Antoine, 75571, Paris Cedex 12, France.
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| |
Collapse
|
11
|
Huang N, Murphy L, Kandasamy S, Wahi G, Pernica JM. Caregiver knowledge and attitudes relating to paediatric pneumonia and antimicrobial stewardship: a qualitative study. Arch Dis Child 2024; 109:222-226. [PMID: 38041668 DOI: 10.1136/archdischild-2023-326080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVE The objective of this study was to understand caregiver perspectives and experiences relating to the treatment of paediatric community-acquired pneumonia (CAP). DESIGN, SETTING AND PATIENTS This was a phenomenological qualitative study involving interviews with caregivers of young children in Hamilton, Ontario. Caregivers were asked open-ended questions relating to germ theory, pneumonia and the role of antibiotic treatment. The principles of conventional content analysis guided the coding and synthesis of the transcribed interviews. RESULTS Eleven caregivers were interviewed. Many knew that antibiotics were not effective against all types of infections and stated that there was an increased risk of developing resistance with frequent use. However, there were misconceptions that probiotics effectively mitigated antibiotic side effects, and few were familiar with the potential long-term consequences of antibiotic use in children.There was variability in the perceived severity of paediatric CAP. Some participants thought that antibiotic treatment would accelerate recovery and prevent caregivers from feeling helpless. However, others also thought it was inappropriate for physicians to prescribe antibiotics solely to make the caregiver feel better. Many caregivers also felt strongly that clinical follow-up and discussions on treatment risks/benefits would be desirable to counteract feelings of helplessness that result from being sent home without a prescription. CONCLUSION Recognising that parents may have misperceptions about antibiotic use for CAP (and may seek antibiotics without strong rationale) can inform clinicians' efforts to better educate and support caregivers in the emergency department. Care strategies informed by caregiver experiences can improve parent-provider communication and reduce antibiotic misuse.
Collapse
Affiliation(s)
- Nelson Huang
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lara Murphy
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Sujane Kandasamy
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Child and Youth Studies, Brock University, St Catherines, Ontario, Canada
| | - Gita Wahi
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Jeffrey M Pernica
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- Department of Pediatrics, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
12
|
Levy EI, Dinleyici M, Dinleyici E, Vandenplas Y. Clostridioides difficile Infections: Prevention and Treatment Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:175-186. [PMID: 39060738 DOI: 10.1007/978-3-031-58572-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Clostridioides difficile is the most common causative agent of antibiotic-associated diarrhea. This spore forming, obligate anaerobic, gram-positive bacillus is becoming responsible for an increasing number of infections worldwide, both in community and in hospital settings, whose severity can vary widely from an asymptomatic infection to a lethal disease. While discontinuation of antimicrobial agents and antibiotic treatment of the infection remain the cornerstone of therapy, more recent fecal microbiota transplantation has also been valid as a therapy. The use of probiotics, especially Saccharomyces boulardii CNCM I-745 have become valid forms of prevention therapy. Although there are studies in adults with microbiota-targeted new generation therapies and Clostridium difficile vaccines, there are no data in the paediatric age group yet.
Collapse
Affiliation(s)
- Elvira Ingrid Levy
- Department of Pediatrics, C.H.U. Saint-Pieter, Free University of Brussels, Brussels, Belgium
| | - Meltem Dinleyici
- Eskisehir Osmangazi University Faculty of Medicine, Department of Social Pediatrics, Eskisehir, Turkey
| | - Ener Dinleyici
- Department of Pediatrics, Eskisehir Osmangazi University School of Medicine, Eskisehir, Turkey
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, KidZ Health Castle, Brussels, Belgium.
| |
Collapse
|
13
|
Pernigoni N, Guo C, Gallagher L, Yuan W, Colucci M, Troiani M, Liu L, Maraccani L, Guccini I, Migliorini D, de Bono J, Alimonti A. The potential role of the microbiota in prostate cancer pathogenesis and treatment. Nat Rev Urol 2023; 20:706-718. [PMID: 37491512 DOI: 10.1038/s41585-023-00795-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2023] [Indexed: 07/27/2023]
Abstract
The human body hosts a complex and dynamic population of trillions of microorganisms - the microbiota - which influences the body in homeostasis and disease, including cancer. Several epidemiological studies have associated specific urinary and gut microbial species with increased risk of prostate cancer; however, causal mechanistic data remain elusive. Studies have associated bacterial generation of genotoxins with the occurrence of TMPRSS2-ERG gene fusions, a common, early oncogenic event during prostate carcinogenesis. A subsequent study demonstrated the role of the gut microbiota in prostate cancer endocrine resistance, which occurs, at least partially, through the generation of androgenic steroids fuelling oncogenic signalling via the androgen receptor. These studies present mechanistic evidence of how the host microbiota might be implicated in prostate carcinogenesis and tumour progression. Importantly, these findings also reveal potential avenues for the detection and treatment of prostate cancer through the profiling and modulation of the host microbiota. The latter could involve approaches such as the use of faecal microbiota transplantation, prebiotics, probiotics, postbiotics or antibiotics, which can be used independently or combined with existing treatments to reverse therapeutic resistance and improve clinical outcomes in patients with prostate cancer.
Collapse
Affiliation(s)
- Nicolò Pernigoni
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Christina Guo
- Institute of Cancer Research, London, UK
- Royal Marsden Hospital, London, UK
| | | | - Wei Yuan
- Institute of Cancer Research, London, UK
| | - Manuel Colucci
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Martina Troiani
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Lei Liu
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Luisa Maraccani
- Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Ilaria Guccini
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Denis Migliorini
- Department of Oncology, Geneva University Hospitals, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland
- Swiss Cancer Center Léman, Lausanne and Geneva, Geneva, Switzerland
- AGORA Cancer Research Center, Lausanne, Switzerland
| | - Johann de Bono
- Institute of Cancer Research, London, UK
- Royal Marsden Hospital, London, UK
| | - Andrea Alimonti
- Institute of Oncology Research, Bellinzona, Switzerland.
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
- Veneto Institute of Molecular Medicine, Padova, Italy.
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland.
- Department of Medicine, University of Padova, Padova, Italy.
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
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
|
15
|
Goya-Jorge E, Gonza I, Bondue P, Druart G, Al-Chihab M, Boutaleb S, Douny C, Scippo ML, Thonart P, Delcenserie V. Evaluation of Four Multispecies Probiotic Cocktails in a Human Colonic Fermentation Model. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10162-7. [PMID: 37725305 DOI: 10.1007/s12602-023-10162-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 09/21/2023]
Abstract
Bacteriotherapy represents an attractive approach for both prophylaxis and treatment of human diseases. However, combining probiotic bacteria in "cocktails" is underexplored, despite its potential as an alternative multi-target therapy. Herein, three-strain probiotic mixtures containing different combinations of Bacillus (Bc.) coagulans [ATB-BCS-042], Levilactobacillus (Lv.) brevis [THT 0303101], Lacticaseibacillus (Lc.) paracasei [THT 031901], Bacillus subtilis subsp. natto [ATB-BSN-049], Enterococcus faecium [ATB-EFM-030], and Bifidobacterium (Bf.) animalis subsp. lactis [THT 010802] were prepared. Four cocktails (PA: Bc. coagulans + Lv. brevis + Lc. paracasei, PB: Bc. subtilis subsp. natto + Lv. brevis + Lc. paracasei, PC: E. faecium + Lv. brevis + Lc. paracasei, PD: Bc. coagulans + Lv. brevis + Bf. animalis subsp. lactis) were tested using a short-term (72 h) simulation of the human colonic microbiota in a final dose of 6 × 109 CFU. All these probiotic mixtures significantly increased butyrate production compared to the parallel control experiment. PA and PB promoted a bifidogenic effect and facilitated lactobacilli colonization. Furthermore, reporter gene assays using the AhR_HT29-Lucia cell line revealed that fermentation supernatants from PA and PB notably induced AhR transactivity. Subsequent examination of the metabolic outputs of PA and PB in intestinal epithelial models using cell culture inserts suggested no significant impact on the transepithelial electrical resistance (TEER). Assessment of the expression of proinflammatory and anti-inflammatory cytokines, as well as AhR-related target genes in the Caco-2 cell monolayers indicated that PB's metabolic output upregulated most of the measured endpoints. This in vitro investigation evaluated the potential impact of four multispecies probiotic mixtures in the human colonic microbiota and identified a promising formulation comprising a combination of Bc. subtilis subsp. natto, Lv. brevis, and Lc. paracasei as a promising formulation for further study.
Collapse
Affiliation(s)
- Elizabeth Goya-Jorge
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Irma Gonza
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Pauline Bondue
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Germain Druart
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Mohamed Al-Chihab
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Samiha Boutaleb
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Caroline Douny
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Marie-Louise Scippo
- Laboratory of Food Analysis, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium
| | - Philippe Thonart
- Lacto Research Sprl, Rue Herman Méganck 21, 5032, Isnes-Gembloux, Belgium
| | - Véronique Delcenserie
- Laboratory of Food Quality Management, Department of Food Sciences, FARAH - Veterinary Public Health, University of Liège, B43b, 4000, Liège, Belgium.
| |
Collapse
|
16
|
Liang H, Tran NT, Deng T, Li J, Lei Y, Bakky MAH, Zhang M, Li R, Chen W, Zhang Y, Chen X, Li S. Identification and Characterization of a Potential Probiotic, Clostridium butyricum G13, Isolated from the Intestine of the Mud Crab (Scylla paramamosain). Microbiol Spectr 2023; 11:e0131723. [PMID: 37522814 PMCID: PMC10434012 DOI: 10.1128/spectrum.01317-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
The butyrate-producing bacterium Clostridium butyricum has been proven to be important in improving the growth and health benefits of aquatic animals. In this study, C. butyricum G13 was isolated for the first time from the gut of the mud crab (Scylla paramamosain). The results of this study showed that C. butyricum G13 could produce a high concentration of butyric acid and grow well in a wide range of pHs (4 to 9) and NaCl (1 to 2.5%) and bile salt (0.2 to 1.0%) concentrations. In vitro characterization revealed that C. butyricum G13 is a Gram-positive and gamma-hemolytic bacterium sensitive to most antibiotics and shows hydrophobicity and the capacity to degrade starch. In vitro fermentation using mud crab gut contents showed that C. butyricum G13 alone or in combination with galactooligosaccharides (GOS) and/or resistant starch (RS) significantly increased butyric acid production and beneficially affected the abundance and diversity of intestinal microbiota. In addition, C. butyricum G13 can improve the survival rate of mud crabs and effectively maintain the normal structure of gut morphology after infection with Vibrio parahaemolyticus. In conclusion, C. butyricum G13 can be considered a potential probiotic that improves the immune capacity and confers health benefits on mud crabs. IMPORTANCE With the development of society, more and more aquatic animals are demanded. Intensification in the aquaculture scale is facing problems, such as disease outbreaks, eutrophication of water bodies, and misuse of antibiotics. Among these challenges, disease outbreak is the most important factor directly affecting aquaculture production. It is crucial to explore new approaches effective for the prevention and control of diseases. Probiotics have been widely used in aquaculture and have shown beneficial effects on the host. In this study, the butyrate-producing bacterium Clostridium butyricum G13 was isolated for the first time from the intestine of the mud crab through in vitro fermentation. The bacterium has probiotic properties and changes the gut microbiota to be beneficial to hosts in vitro as well as protecting hosts from Vibrio parahaemolyticus infection in vivo. The outcomes of this study indicate that C. butyricum G13 can be used as a potential probiotic in mud crab aquaculture.
Collapse
Affiliation(s)
- Huifen Liang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Ngoc Tuan Tran
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Taoqiu Deng
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Jinkun Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Yifan Lei
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Mohammad Akibul Hasan Bakky
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Ming Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Rui Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Wenxuan Chen
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Yueling Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| | - Xiuli Chen
- Guangxi Academy of Fishery Sciences, Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Nanning, China
| | - Shengkang Li
- Guangdong Provincial Key Laboratory of Marine Biology, Shantou University, Shantou, China
- Institute of Marine Sciences, Shantou University, Shantou, China
| |
Collapse
|
17
|
Mego M, Danis R, Chovanec J, Jurisova S, Bystricky B, Porsok S, Konkolovsky P, Vaclav V, Wagnerova M, Streško M, Brezinova B, Rečková M, Sutekova D, Pazderova N, Novisedlakova M, Zomborska E, Ciernikova S, Svetlovska D, Drgona L. Randomized double-blind, placebo-controlled multicenter phase III study of prevention of irinotecan-induced diarrhea by a probiotic mixture containing Bifidobacterium BB-12 ®Lactobacillus rhamnosus LGG ® in colorectal cancer patients. Front Oncol 2023; 13:1168654. [PMID: 37601667 PMCID: PMC10438450 DOI: 10.3389/fonc.2023.1168654] [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] [Received: 02/17/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Background The incidence of irinotecan-induced diarrhea varies between 60-90%, by which the incidence of severe diarrhea is 20-40%. The objective of this phase III trial was to determine the effectiveness of the probiotic mixture containing Bifidobacterium, BB-12® and Lactobacillus rhamnosus, LGG® in the prophylaxis of irinotecan-induced diarrhea in metastatic colorectal cancer patients due to a reduction in the activity of intestinal beta-D-glucuronidase. Methods From March 2016 to May 2022, a total of 242 patients with colorectal cancer starting a new line of irinotecan-based therapy were registered to the study in 11 cancer centers in Slovakia. Patients were randomized in a ratio 1:1 to probiotic formula vs. placebo that was administered for 6 weeks. Each capsule of Probio-Tec® BG-Vcap-6.5 contained 2.7x109 colony-forming units (CFU) of 2 lyophilized probiotic strains Bifidobacterium, BB-12® (50%) and Lactobacillus rhamnosus GG, LGG® (50%). Results Administration of probiotics compared to placebo was not associated with a significant reduction of grade 3/4 diarrhea (placebo arm 11.8% vs. probiotic arm 7.9%, p=0.38). Neither the overall incidence of diarrhea (46.2% vs. 41.2%, p=0.51) nor the incidence of enterocolitis (3.4% vs. 0.9%, p=0.37) was different in the placebo vs. probiotic arm. Subgroup analysis revealed that patients with colostomy had higher incidence of any diarrhea and grade 3/4 diarrhea in the placebo arm compared to the probiotic arm (48.5% vs. 22.2%, p=0.06 and 15.2% vs. 0%, p=0.06, respectively). Moreover, patients on probiotic arm had significantly better diarrhea-free survival (HR = 0.41, 95%CI 0.18 - 0.95, p=0.05) and needed less loperamide (p=0.01) compared to patients on placebo arm. We did not observe any infection caused by probiotic strains used in this study. Conclusion This study failed to achieve its primary endpoint, and results suggest a lack of benefit of administered probiotic formula for the prevention of irinotecan-induced diarrhea. However, subgroup analysis suggests a possible benefit in patients with colostomy.
Collapse
Affiliation(s)
- Michal Mego
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Radoslav Danis
- Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Jozef Chovanec
- Department of Oncology, St. Jacob Hospital, Bardejov, Slovakia
| | - Silvia Jurisova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Stefan Porsok
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | | | - Vladimir Vaclav
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Maria Wagnerova
- Department of Oncology, East Slovakia Comprehensive Cancer Center, Kosice, Slovakia
| | - Marian Streško
- Department of Oncology, Faculty Hospital, Trnava, Trebisov, Slovakia
| | | | - Mária Rečková
- Department of Oncology, Regional Cancer Center, Poprad, Slovakia
| | - Dagmar Sutekova
- Department of Oncology, University Hospital Martin, Martin, Slovakia
| | - Natalia Pazderova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Mária Novisedlakova
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
- Department of Oncology, University Hospital Milosrdni Bratia, Bratislava, Slovakia
| | - Eva Zomborska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Sona Ciernikova
- Biomedical Center, Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Daniela Svetlovska
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| | - Lubos Drgona
- Faculty of Medicine, Comenius University and National Cancer Institute, Bratislava, Slovakia
| |
Collapse
|
18
|
Yang Q, Hu Z, Lei Y, Li X, Xu C, Zhang J, Liu H, Du X. Overview of systematic reviews of probiotics in the prevention and treatment of antibiotic-associated diarrhea in children. Front Pharmacol 2023; 14:1153070. [PMID: 37564180 PMCID: PMC10411537 DOI: 10.3389/fphar.2023.1153070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Background: Antibiotics alter the microbial balance commonly resulting in antibiotic-associated diarrhea (AAD). Probiotics may prevent and treat AAD by providing the gut barrier and restoring the gut microflora. This study will overview the Systematic Reviews (SRs) of probiotics in preventing and treating AAD in children. It will also assess the reporting, methodological, and evidence quality of the included SRs to provide evidence for their clinical practice. Methods: After searching PubMed, Embase, Cochrane Library, CNKI, CBM, VIP, and WanFang Data databases, and finally included SRs of probiotics in the prevention and treatment of AAD in children, which were published before 1 October 2022. The reporting, methodological, and evidence quality of the included SRs were assessed by PRISMA 2020 statement, AMSTAR 2 tool, and GRADE system. Results: A total of 20 SRs were included, and the results of PRISMA 2020 showed that 4 out of 20 SRs with relatively complete reporting, and the others within some reporting deficiencies, with scores ranging from 17 points to 26.5 points; the results of AMSTAR 2 showed that 3 SRs belonged to moderate quality level, 10 SRs belonged to low-quality level and 7 SRs being extremely low-quality level; the results of the GRADE system showed that a total of 47 outcomes were reported for the included SRs, three were high-level evidence quality, 16 were medium-level evidence quality, 24 were low-level evidence quality, and four were extremely low-level evidence quality; the results of the Meta-analysis showed that high doses (5-40 billion CFUs per day) of probiotics had a significant effect in the prevention of AAD, but it is too early to conclude the effectiveness and safety of other probiotic drugs for AAD in children, except for Lacticaseibacillus rhamnosus and Saccharomyces boulardii. Conclusion: Current evidence shows that probiotics effectively prevent and treat AAD in children, and the effect of probiotics on pediatric AAD may be a potential dose-response effect. However, the conclusion should be treated with caution due to deficiencies in the methodological, reporting, and evidence quality of the included SRs. Therefore, the methodological, reporting, and evidence quality of relevant SRs still need further improvement. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/, identifier CRD42022362328.
Collapse
Affiliation(s)
- Qingrui Yang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zeyu Hu
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yuyu Lei
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xinzhu Li
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chao Xu
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Jie Zhang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Haitao Liu
- Department of Gastroenterology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Xiaoquan Du
- Department of Gastroenterology, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
19
|
Vizioli C, Jaime-Lara R, Daniel SG, Franks A, Diallo AF, Bittinger K, Tan TP, Merenstein DJ, Brooks B, Joseph PV, Maki KA. Administration of Bifidobacterium animalis subsp. lactis strain BB-12 ® in healthy children: characterization, functional composition, and metabolism of the gut microbiome. Front Microbiol 2023; 14:1165771. [PMID: 37333640 PMCID: PMC10275293 DOI: 10.3389/fmicb.2023.1165771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/17/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction The consumption of probiotics may influence children's gut microbiome and metabolome, which may reflect shifts in gut microbial diversity composition and metabolism. These potential changes might have a beneficial impact on health. However, there is a lack of evidence investigating the effect of probiotics on the gut microbiome and metabolome of children. We aimed to examine the potential impact of a two (Streptococcus thermophilus and Lactobacillus delbrueckii; S2) vs. three (S2 + Bifidobacterium animalis subsp. lactis strain BB-12) strain-supplemented yogurt. Methods Included in this study were 59 participants, aged one to five years old, recruited to phase I of a double-blinded, randomized controlled trial. Fecal samples were collected at baseline, after the intervention, and at twenty days post-intervention discontinuation, and untargeted metabolomics and shotgun metagenomics were performed. Results Shotgun metagenomics and metabolomic analyses showed no global changes in either intervention group's gut microbiome alpha or beta diversity indices, except for a lower microbial diversity in the S2 + BB12 group at Day 30. The relative abundance of the two and three intervention bacteria increased in the S2 and S2 + BB12 groups, respectively, from Day 0 to Day 10. In the S2 + BB12 group, the abundance of several fecal metabolites increased at Day 10, including alanine, glycine, lysine, phenylalanine, serine, and valine. These fecal metabolite changes did not occur in the S2 group. Discussion In conclusion, there were were no significant differences in the global metagenomic or metabolomic profiles between healthy children receiving two (S2) vs. three (S2 + BB12) probiotic strains for 10 days. Nevertheless, we observed a significant increase (Day 0 to Day 10) in the relative abundance of the two and three probiotics administered in the S2 and S2 + BB12 groups, respectively, indicating the intervention had a measurable impact on the bacteria of interest in the gut microbiome. Future research using longer probiotic intervention durations and in children at risk for gastrointestinal disorders may elucidate if functional metabolite changes confer a protective gastrointestinal effect.
Collapse
Affiliation(s)
- Carlotta Vizioli
- Department of Health and Human Services, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Department of Health and Human Services, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Rosario Jaime-Lara
- Department of Health and Human Services, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
- Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
- UCLA School of Nursing, University of California, Los Angeles, Los Angeles, CA, United States
| | - Scott G. Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Alexis Franks
- Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Ana F. Diallo
- Family and Community Health Nursing, School of Nursing, Institute of Inclusion, Inquiry and Innovation (iCubed), Virginia Commonwealth University, Richmond, VA, United States
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Tina P. Tan
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Daniel J. Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Brianna Brooks
- Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Paule V. Joseph
- Department of Health and Human Services, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
- Department of Health and Human Services, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, United States
| | - Katherine A. Maki
- Translational Biobehavioral and Health Disparities Branch, National Institutes of Health, Clinical Center, Bethesda, MD, United States
| |
Collapse
|
20
|
Zeng Z, Yue W, Kined C, Wang P, Liu R, Liu J, Chen X. Bacillus licheniformis reverses the environmental ceftriaxone sodium-induced gut microbial dysbiosis and intestinal inflammation in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 257:114890. [PMID: 37084659 DOI: 10.1016/j.ecoenv.2023.114890] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/18/2023] [Accepted: 04/08/2023] [Indexed: 05/03/2023]
Abstract
Antibiotics used as a common clinical treatment have saved many lives. Widespread use of antibiotic therapy has been known to disrupt the balance of pathogenic bacteria, host-associated microorganisms and environment. However, our understanding of Bacillus licheniformis for health benefits and ability to restore the ceftriaxone sodium-induced gut microbial dysbiosis is severely limited. We used Caco-2 cell, H&E (hematoxylin-eosin staining), RT-PCR and 16S rRNA sequencing techniques to investigate the influence of Bacillus licheniformis on gut microbial dysbiosis and inflammation following ceftriaxone sodium treatment. The results showed that treatment of ceftriaxone sodium in 7 days suppressed the expression of Nf-κB pathway mRNA levels, which caused cytoplasmic vacuolization in intestinal tissues, afterward, the administration of Bacillus licheniformis could effectively restore intestinal morphology and inflammation levels. Moreover, the ceftriaxone sodium treatment entirely affected the intestinal microbial ecology, leading to a decrease in microbial abundance. Firmicutes, Proteobacteria, and Epsilonbacteraeota were the most predominant phyla in each of the four groups. Specifically, the MA group (ceftriaxone sodium treatment) resulted in a significant decrease in the relative abundance of 2 bacterial phyla and 20 bacterial genera compared to the administration of Bacillus licheniformis after ceftriaxone sodium treatment. The supplementation of Bacillus licheniformis could increase the growth of Firmicutes and Lactobacillus and encourage the construction of a more mature and stable microbiome. Furthermore, Bacillus licheniformis could restore the intestinal microbiome disorders and inflammation levels following ceftriaxone sodium treatment.
Collapse
Affiliation(s)
- Zhibo Zeng
- Institute of Animal Husbandry and Veterinary Medicine/Fujian Key Laboratory of Animal Genetics and Breeding, Fujian Academy of Agricultural Sciences, Fuzhou 350013, PR China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China; Institute of Agricultural Sciences, ETH Zurich, Universitaetstrasse 2, Zurich 8092, Switzerland
| | - Wen Yue
- Institute of Animal Husbandry and Veterinary Medicine/Fujian Key Laboratory of Animal Genetics and Breeding, Fujian Academy of Agricultural Sciences, Fuzhou 350013, PR China
| | - Cermon Kined
- Institute of Agricultural Sciences, ETH Zurich, Universitaetstrasse 2, Zurich 8092, Switzerland
| | - PengPeng Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Ran Liu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China
| | - Jing Liu
- Institute of Animal Husbandry and Veterinary Medicine/Fujian Key Laboratory of Animal Genetics and Breeding, Fujian Academy of Agricultural Sciences, Fuzhou 350013, PR China
| | - Xinzhu Chen
- Institute of Animal Husbandry and Veterinary Medicine/Fujian Key Laboratory of Animal Genetics and Breeding, Fujian Academy of Agricultural Sciences, Fuzhou 350013, PR China.
| |
Collapse
|
21
|
Choudhury J, Makkar A, Sharma V, Karamath SP, Parmar V, Kumar J P, Veligandla KC, Pinto CS, Mane A, Rathod R, Kotak BP. A Real-World Perspective of Co-amoxiclav Prescription Pattern With Probiotics for Pediatric Patients With Respiratory Tract Infections: Results of Quantitative and Qualitative Approach in Indian Outpatient Settings. Cureus 2023; 15:e36269. [PMID: 37073213 PMCID: PMC10105827 DOI: 10.7759/cureus.36269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Probiotics are co-prescribed with co-amoxiclav to prevent antibiotic-associated diarrhea (AAD). The study assesses the co-prescription pattern of probiotics with co-amoxiclav in pediatric patients with respiratory tract infections (RTIs). METHODS This was a mixed methods research study with a retrospective study and a prospective survey. The retrospective part included a multicenter, observational, real-world study utilizing patients' electronic medical records for three years (2018-2020) from seven outpatient pediatric clinics and hospitals. The qualitative evaluation was performed with a predefined questionnaire. RESULTS The patients having RTIs (N=984) were prescribed Clamp® (46.7%), CAA (23.8%), and CAM (29.5%). The mean age of the patients was 4.05 years, with 59.25% males and most patients having upper RTIs. Co-amoxiclav was prescribed twice daily for one to 15 days. A significantly lesser number of probiotic co-prescriptions were observed with Clamp® (19.57%) than with CAA (38.46%) and CAM (29.31%) at baseline (p<0.001). Similar findings were observed for follow-up visits one and two. Saccharomyces boulardii, Bacillus clausii,and lactic acid bacillus were the most commonly co-prescribed probiotics. The qualitative evaluation indicated that most clinicians were aware of the co-amoxiclav-related gastrointestinal side effects and the benefits of probiotics in preventing them. CONCLUSION The frequency of co-prescriptions of probiotics with Clamp® among pediatric patients with RTIs was significantly less, potentially indicating better gastrointestinal tolerability.
Collapse
|
22
|
Wang Y, Wen L, Tang H, Qu J, Rao B. Probiotics and Prebiotics as Dietary Supplements for the Adjunctive Treatment of Type 2 Diabetes. Pol J Microbiol 2023; 72:3-9. [PMID: 36929892 DOI: 10.33073/pjm-2023-013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/13/2023] [Indexed: 03/18/2023] Open
Abstract
In modern lifestyles, high-fat diets and prolonged inactivity lead to more people developing type 2 diabetes (T2D). Based on the modern pathogenesis of T2D, food, and its components have become one of the top concerns for patients. Recent studies have found that dysbiosis and gut-related inflammation are more common in T2D patients. Probiotics and prebiotics play complementary roles in the gut as dietary supplements. Together, they may help improve dysbiosis and intestinal inflammation in people with T2D, increase the production of blood glucose-lowering hormones such as incretin, and help reduce insulin resistance and lower blood glucose. Therefore, changing the dietary structure and increasing the intake of probiotics and prebiotics is expected to become a new strategy for the adjuvant treatment of T2D.
Collapse
Affiliation(s)
- Yuying Wang
- 1Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- 2Key Laboratory of Cancer FSMP for State Market Regulation, Beijing Shijitan Hospital, Beijing, China
| | - Lina Wen
- 1Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- 2Key Laboratory of Cancer FSMP for State Market Regulation, Beijing Shijitan Hospital, Beijing, China
| | - Huazhen Tang
- 1Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- 2Key Laboratory of Cancer FSMP for State Market Regulation, Beijing Shijitan Hospital, Beijing, China
| | - Jinxiu Qu
- 1Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- 2Key Laboratory of Cancer FSMP for State Market Regulation, Beijing Shijitan Hospital, Beijing, China
| | - Benqiang Rao
- 1Department of Gastrointestinal Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- 2Key Laboratory of Cancer FSMP for State Market Regulation, Beijing Shijitan Hospital, Beijing, China
| |
Collapse
|
23
|
Bai H, Liu T, Wang S, Shen L, Wang Z. Variations in gut microbiome and metabolites of dogs with acute diarrhea in poodles and Labrador retrievers. Arch Microbiol 2023; 205:97. [PMID: 36823480 DOI: 10.1007/s00203-023-03439-6] [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: 01/19/2023] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/25/2023]
Abstract
For different breeds of dogs with acute diarrhea, the gut microbiota and metabolome profiles are unclear. This prospective observational study analyzed the gut microbiomes of poodles with acute diarrhea and Labrador retrievers with acute diarrhea based on 16S amplicon sequencing, with respective healthy dogs as controls. Fecal non-target metabolomics and metagenomics were performed on poodles with acute diarrhea. This study found that the diversity and structure of the microbial community differed significantly between the two breeds in cohorts of healthy dogs. Two breeds of dogs with acute diarrhea demonstrated different changes in microbial communities and metabolic functions. The metabolism of starch and sucrose was significantly decreased in dogs with acute diarrhea, which may be attributed to the reduced activity of dextran dextrinase. Non-targeted metabolomics identified 21 abnormal metabolic pathways exhibited by dogs with acute diarrhea, including starch, amino acid, bile acid metabolism, etc., and were closely related to specific intestinal flora. This study provided new insights into breed specificity and the development of dietary treatment strategy in canine gastrointestinal disease.
Collapse
Affiliation(s)
- Huasong Bai
- Nourse Centre for Pet Nutrition, Wuhu, 241200, China
| | - Tong Liu
- Nourse Centre for Pet Nutrition, Wuhu, 241200, China
| | - Songjun Wang
- Nourse Centre for Pet Nutrition, Wuhu, 241200, China
| | - Liya Shen
- Nourse Centre for Pet Nutrition, Wuhu, 241200, China
| | | |
Collapse
|
24
|
Hypoglycemic Effects of Inactivated Lactobacillus brevis YM 1301 on T2DM Mice. J Food Biochem 2023. [DOI: 10.1155/2023/9363576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Lactobacillus brevis (L. brevis) is a widely used probiotic with health-promoting properties. Previous studies reported that L. brevis has beneficial impacts on T2DM mice. On the other hand, the differences in effects between live and inactivated L. brevis are still inconclusive. This study aims to evaluate the hypoglycemic effects of inactivated L. brevis YM1301 (YM1301) on T2DM mice induced by a high-fat diet (HFD) combined with streptozocin (STZ). T2DM mice were randomly stratified into four groups and administered either saline (model group), metformin (positive control group), live YM1301 (LB group), or inactivated YM1301 (ILB group). Results demonstrated that inactivated YM1301 reduced serum content of fasting blood glucose, insulin, and glycated hemoglobin, enhanced the level of glucose tolerance, and decreased insulin resistance in T2DM mice. Inactivated YM1301 also decreased fat accumulation in T2DM mice. In addition, inactivated YM1301 effectively promoted serum GLP-1 levels. These results showed that inactivated YM1301 can significantly improve symptoms of T2DM. This study provides theoretical feasibility for the development of low-cost, easy to store, safer, and more effective probiotic products related to L. brevis.
Collapse
|
25
|
Wu EH, Guo Z, Zhu WM. Postoperative diarrhea in Crohn’s disease: Pathogenesis, diagnosis, and therapy. World J Clin Cases 2023; 11:7-16. [PMID: 36687182 PMCID: PMC9846968 DOI: 10.12998/wjcc.v11.i1.7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Diarrhea is a frequent symptom in postoperative patients with Crohn’s diseases (CD), and several different mechanisms likely account for postoperative diarrhea in CD. A targeted strategy based on a comprehensive understanding of postoperative diarrhea is helpful for better postoperative recovery.
Collapse
Affiliation(s)
- En-Hao Wu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Zhen Guo
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Wei-Ming Zhu
- Department of General Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
26
|
Merenstein D, Pot B, Leyer G, Ouwehand AC, Preidis GA, Elkins CA, Hill C, Lewis ZT, Shane AL, Zmora N, Petrova MI, Collado MC, Morelli L, Montoya GA, Szajewska H, Tancredi DJ, Sanders ME. Emerging issues in probiotic safety: 2023 perspectives. Gut Microbes 2023; 15:2185034. [PMID: 36919522 PMCID: PMC10026873 DOI: 10.1080/19490976.2023.2185034] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are used for both generally healthy consumers and in clinical settings. However, theoretical and proven adverse events from probiotic consumption exist. New probiotic strains and products, as well as expanding use of probiotics into vulnerable populations, warrants concise, and actionable recommendations on how to work toward their safe and effective use. The International Scientific Association for Probiotics and Prebiotics convened a meeting to discuss and produce evidence-based recommendations on potential acute and long-term risks, risks to vulnerable populations, the importance for probiotic product quality to match the needs of vulnerable populations, and the need for adverse event reporting related to probiotic use. The importance of whole genome sequencing, which enables determination of virulence, toxin, and antibiotic resistance genes, as well as clear assignment of species and strain identity, is emphasized. We present recommendations to guide the scientific and medical community on judging probiotic safety.
Collapse
Affiliation(s)
- Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DCUSA
| | - Bruno Pot
- Yakult Europe BV, Almere, Netherlands
| | | | - Arthur C. Ouwehand
- Global Health & Nutrition Sciences, International Flavors & Fragrances, Kantvik, Finland
| | - Geoffrey A. Preidis
- Division of Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Christopher A. Elkins
- Clinical and Environmental Microbiology Branch, Division of Healthcare Quality Promotion, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Andi L. Shane
- Division of Infectious Diseases, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Emory Children’s Center, Atlanta, Georgia
| | - Niv Zmora
- Scientific consultant, Elinav Lab, Immunology Department, Weizmann Institute of Science, Department of Gastroenterology and Liver Diseases, Tel Aviv, Israel
| | | | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Lorenzo Morelli
- Department of Food Science and Technology, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Gina A. Montoya
- Department of Chemical Risk Assessment, Nestlé S.A., Lausanne, Switzerland
| | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Daniel J. Tancredi
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Centennial, CO, USA
| |
Collapse
|
27
|
Danis R, Mego M, Antonova M, Stepanova R, Svobodnik A, Hejnova R, Wawruch M. Orally Administered Probiotics in the Prevention of Chemotherapy (± Radiotherapy)-Induced Gastrointestinal Toxicity: A Systematic Review With Meta-Analysis of Randomized Trials. Integr Cancer Ther 2022; 21:15347354221144309. [PMID: 36567453 PMCID: PMC9806400 DOI: 10.1177/15347354221144309] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Chemoradiotherapy-induced gastrointestinal toxicity may lead to a significant impairment of the oncological patient's quality of life, as well as to reduced adherence to the treatment, which may have a negative impact on survival and mortality rates. OBJECTIVE The aim of this review was to investigate whether oral probiotic administration prevents chemotherapy (± radiotherapy)-induced gastrointestinal toxicity, particularly diarrhea. METHODS We searched the MEDLINE, Web of Science, and SCOPUS databases for randomized controlled trials in English published between 1990 and 2020. We conducted statistical data analyses expressing the treatment effect size as a risk ratio (RR) together with a 95% confidence interval (CI). Implications are based on trials rated as having a low risk of bias (RoB). RESULTS We included 8 trials (n = 697 participants), from which 3 studies rated as low RoB contained primary endpoint data; the risk of developing grade 3/4 diarrhea in patients receiving probiotics was reduced by 78% compared to the control group (RR = 0.22 [95% CI 0.05-1.08]; P = .06; n = 114 participants). Probiotics showed preventive effects in patients treated with chemotherapy alone (RR = 0.34 [0.12-0.94]; P = .04, n = 121 participants) and in patients with colorectal cancer (RR = 0.56 [0.34-0.92]; P = .02; n = 208 participants). The reduction in the incidence of overall diarrhea was not significant. CONCLUSIONS Probiotics failed to prove a preventive effect of statistical significance against the development of severe and overall diarrhea in cancer patients treated with chemotherapy (± radiotherapy). However, we cannot rule out that the effects of probiotics are clinically relevant, especially in certain subgroups of patients. This needs to be clarified in further well-performed studies.
Collapse
Affiliation(s)
- Radoslav Danis
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia,Radoslav Danis, Comenius University in
Bratislava Faculty of Medicine, Spitalska 24, Bratislava 813 72, Slovakia.
| | - Michal Mego
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia,National Cancer Institute, Bratislava,
Slovak Republic
| | | | | | | | - Renata Hejnova
- Masaryk University Faculty of Medicine,
Brno, Czech Republic
| | - Martin Wawruch
- Comenius University in Bratislava
Faculty of Medicine, Bratislava, Slovakia
| |
Collapse
|
28
|
Amato M, Di Spirito F, D’Ambrosio F, Boccia G, Moccia G, De Caro F. Probiotics in Periodontal and Peri-Implant Health Management: Biofilm Control, Dysbiosis Reversal, and Host Modulation. Microorganisms 2022; 10:2289. [PMID: 36422359 PMCID: PMC9694231 DOI: 10.3390/microorganisms10112289] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/10/2022] [Accepted: 11/16/2022] [Indexed: 07/30/2023] Open
Abstract
Periodontitis and peri-implantitis are microbially associated diseases of the tissues supporting the teeth and dental implants that are mediated by host inflammation and eventually lead to tooth and dental implant loss. Given the probiotics' role in biofilm control, dysbiosis reversal, and host modulation, their potential beneficial effects on the improvement of periodontitis and peri-implantitis have been recently investigated. Moreover, probiotics use has also been proposed in periodontal health management in patients undergoing fixed orthodontic therapy. Therefore, the present study aimed to review, considering the periodontal microbiome composition around teeth and dental implants in healthy and pathological conditions, the putative favorable effects of probiotics on gingivitis, periodontitis, and peri-implantitis. The secondary aim of the present narrative review was to synthesize the supporting evidence and proposed protocols for probiotics use as adjuncts in periodontitis and peri-implantitis treatment and the periodontal health management of orthodontic patients with fixed appliances. Contrasting findings from the literature may be due to the different methods, posology, and duration of probiotics prescriptions and due to the heterogeneous biological and clinical measurement methods employed. Thus, no definitive conclusions could be drawn about the effectiveness of probiotics in periodontal management, both in healthy and pathological conditions. Further studies are needed to validate probiotics for periodontal management and provide recommended protocols.
Collapse
|
29
|
Yang J, Qin S, Zhang H. Precise strategies for selecting probiotic bacteria in treatment of intestinal bacterial dysfunctional diseases. Front Immunol 2022; 13:1034727. [PMID: 36341458 PMCID: PMC9632739 DOI: 10.3389/fimmu.2022.1034727] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/07/2022] [Indexed: 11/20/2022] Open
Abstract
Abundant microbiota resides in the organs of the body, which utilize the nutrition and form a reciprocal relationship with the host. The composition of these microbiota changes under different pathological conditions, particularly in response to stress and digestive diseases, making the microbial composition and health of the hosts body interdependent. Probiotics are living microorganisms that have demonstrated beneficial effects on physical health and as such are used as supplements to ameliorate symptoms of various digestive diseases by optimizing microbial composition of the gut and restore digestive balance. However, the supplementary effect does not achieve the expected result. Therefore, a targeted screening strategy on probiotic bacteria is crucial, owing to the presence of several bacterial strains. Core bacteria work effectively in maintaining microbiological homeostasis and stabilization in the gastrointestinal tract. Some of the core bacteria can be inherited and acquired from maternal pregnancy and delivery; others can be acquired from contact with the mother, feces, and the environment. Knowing the genera and functions of the core bacteria could be vital in the isolation and selection of probiotic bacteria for supplementation. In addition, other supporting strains of probiotic bacteria are also needed. A comprehensive strategy for mining both core and supporting bacteria before its clinical use is needed. Using metagenomics or other methods of estimation to discern the typically differentiated strains of bacteria is another important strategy to treat dysbiosis. Hence, these two factors are significant to carry out targeted isolation and selection of the functional strains to compose the resulting probiotic preparation for application in both research and clinical use. In conclusion, precise probiotic supplementation, by screening abundant strains of bacteria and isolating specific probiotic strains, could rapidly establish the core microbiota needed to confer resilience, particularly in bacterial dysfunctional diseases. This approach can help identify distinct bacteria which can be used to improve supplementation therapies.
Collapse
Affiliation(s)
- Jiajun Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, China
| | - Shunyi Qin
- Key Laboratory of Agricultural Animal Breeding and Healthy Breeding of Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Hao Zhang
- College of Animal Science and Technology, Chinese Agricultural University, Beijing, China
- *Correspondence: Hao Zhang,
| |
Collapse
|
30
|
Effect of Traditional Chinese Medicine on Treating Antibiotic-Associated Diarrhea in Children: A Systematic Review and Meta-Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6108772. [PMID: 36185092 PMCID: PMC9522518 DOI: 10.1155/2022/6108772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/30/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022]
Abstract
Background Due to the limited treatment options in antibiotic-associated diarrhea (AAD) in children, more effective treatments should be explored. Traditional Chinese medicine (TCM) has a long history in China, which has produced a pretty effect in clinical practice. Many randomized clinical trials (RCTs) have explored the effect of traditional Chinese medicine on treating AAD in children. However, there has been no systematic review or meta-analysis on the impact of TCM on AAD in children. The aim of this study was to systematically review RCTs on the effect of TCM in children with AAD. Methods RCTs in the past ten years on TCM for AAD in children were included. We searched Electronic databases as much as possible. This paper was registered in PROSPERO (CRD42022301034). Results 26 studies were included in this systematic review. 25 studies reported the effects of TCM interventions on the total effective rate (RR = 1.20, CI 1.16 to 1.24; p < 0.001). 7 studies reported the effects of TCM interventions on the time to change the shape of feces (MD = −1.37, CI −1.67 to −1.07; p < 0.001). 17 studies reported the effects of TCM interventions (MD = −1.43, CI −1.71 to −1.15; p < 0.001). The pooled results showed that there were no significant differences between the two groups in CD3+, CD4+, CD8+, CD4 : CD8, time for bowel sounds to return to normal, hs-CRP, and IgM. There was a significant difference between the two groups in frequency of diarrhea on the third day after TCM intervention, vomiting improvement time, diamine oxidase, IL-8, TNF, IgA, IgG, and average hospital stay. Conclusions TCM interventions combined with conventional therapy can improve the therapeutic effect of AAD in children. However, future studies are still needed for the low methodological quality.
Collapse
|
31
|
Dahshan D, Gallagher N, Workman A, Perdue J, Aikens J, Schmicker T, Shuler FD. Targeting the Gut Microbiome for Inflammation and Pain Management in Orthopedic Conditions. Orthopedics 2022; 45:e226-e234. [PMID: 35700403 DOI: 10.3928/01477447-20220608-07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The human gut microbiome can be altered with probiotics, prebiotics, synbiotics, and anti-inflammatory foods and spices as part of an evidence-based strategy that targets inflammation and pain in common orthopedic conditions. Implementing these strategies avoids adverse effects associated with nonsteroidal anti-inflammatory drugs and minimizes the potential for opioid use. This review focuses exclusively on human trials studying the effects of gut microbiome alterations to address pain and inflammatory markers in common orthopedic conditions: osteoarthritis, rheumatoid arthritis, fractures/osteoporosis, and bone pain associated with chemotherapy. Individualized supplementation strategies can be further explored with the information in this review. [Orthopedics. 2022;45(5):e226-e234.].
Collapse
|
32
|
Chang C, Yuan X, Zhang X, Chen X, Li K. Gastrointestinal Microbiome and Multiple Health Outcomes: Umbrella Review. Nutrients 2022; 14:3726. [PMID: 36145102 PMCID: PMC9505003 DOI: 10.3390/nu14183726] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/29/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, there has been growing concern about the impact of the gastrointestinal microbiome on human health outcomes. To clarify the evidence for a link between the gastrointestinal microbiome and a variety of health outcomes in humans, we conducted an all-encompassing review of meta-analyses and systematic reviews that included 195 meta-analyses containing 950 unique health outcomes. The gastrointestinal microbiome is related to mortality, gastrointestinal disease, immune and metabolic outcomes, neurological and psychiatric outcomes, maternal and infant outcomes, and other outcomes. Existing interventions for intestinal microbiota (such as probiotics, fecal microbiota transplant, etc.) are generally safe and beneficial to a variety of human health outcomes, but the quality of evidence is not high, and more detailed and well-designed randomized controlled trials are necessary.
Collapse
Affiliation(s)
- Chengting Chang
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingzhu Yuan
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xingxia Zhang
- Department of Organization, West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Xinrong Chen
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| | - Ka Li
- West China School of Nursing, Sichuan University/West China Hospital, Sichuan University, 37 Guo Xue Rd., Chengdu 610041, China
| |
Collapse
|
33
|
Łukasik J, Dierikx T, Besseling-van der Vaart I, de Meij T, Szajewska H. Multispecies Probiotic for the Prevention of Antibiotic-Associated Diarrhea in Children: A Randomized Clinical Trial. JAMA Pediatr 2022; 176:860-866. [PMID: 35727573 PMCID: PMC9214631 DOI: 10.1001/jamapediatrics.2022.1973] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
IMPORTANCE The efficacy of multispecies probiotic formulations in the prevention of antibiotic-associated diarrhea (AAD) remains unclear. OBJECTIVE To assess the effect of a multispecies probiotic on the risk of AAD in children. DESIGN, SETTING, AND PARTICIPANTS This randomized, quadruple-blind, placebo-controlled trial was conducted from February 2018 to May 2021 in a multicenter, mixed setting (inpatients and outpatients). Patients were followed up throughout the intervention period. Eligibility criteria included age 3 months to 18 years, recruitment within 24 hours following initiation of broad-spectrum systemic antibiotics, and signed informed consent. In total, 646 eligible patients were approached and 350 patients took part in the trial. INTERVENTIONS A multispecies probiotic consisting of Bifidobacterium bifidum W23, Bifidobacterium lactis W51, Lactobacillus acidophilus W37, L acidophilus W55, Lacticaseibacillus paracasei W20, Lactiplantibacillus plantarum W62, Lacticaseibacillus rhamnosus W71, and Ligilactobacillus salivarius W24, for a total dose of 10 billion colony-forming units daily, for the duration of antibiotic treatment and for 7 days after. MAIN OUTCOMES AND MEASURES The primary outcome was AAD, defined as 3 or more loose or watery stools per day in a 24-hour period, caused either by Clostridioides difficile or of otherwise unexplained etiology, after testing for common diarrheal pathogens. The secondary outcomes included diarrhea regardless of the etiology, diarrhea duration, and predefined diarrhea complications. RESULTS A total of 350 children (192 boys and 158 girls; mean [range] age, 50 [3-212] months) were randomized and 313 were included in the intention-to-treat analysis. Compared with placebo (n = 155), the probiotic (n = 158) had no effect on risk of AAD (relative risk [RR], 0.81; 95% CI, 0.49-1.33). However, children in the probiotic group had a lower risk of diarrhea regardless of the etiology (RR, 0.65; 95% CI, 0.44-0.94). No differences were observed between the groups for most of the secondary outcomes, including adverse events. CONCLUSIONS AND RELEVANCE A multispecies probiotic did not reduce the risk of AAD in children when analyzed according to the most stringent definition. However, it reduced the overall risk of diarrhea during and for 7 days after antibiotic treatment. Our study also shows that the AAD definition has a significant effect on clinical trial results and their interpretation. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT03334604.
Collapse
Affiliation(s)
- Jan Łukasik
- Department of Pediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Thomas Dierikx
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam UMC, Academic Medical Centre, Amsterdam, the Netherlands
| | | | - Tim de Meij
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Amsterdam UMC, Academic Medical Centre, Amsterdam, the Netherlands
| | - Hania Szajewska
- Department of Pediatrics, The Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
34
|
Chen X, Yan Z, Liu L, Zhang R, Zhang X, Peng C, Geng Y, Zhou F, Han Y, Hou X. Characteristics of gut microbiota of term small gestational age infants within 1 week and their relationship with neurodevelopment at 6 months. Front Microbiol 2022; 13:912968. [PMID: 36090083 PMCID: PMC9449527 DOI: 10.3389/fmicb.2022.912968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Small for gestational age (SGA) infants are at a higher risk of neurodevelopmental delay than infants appropriate for gestational age (AGA). Previous studies have confirmed that gut microbiota in early life influences subsequent neurodevelopment. However, few studies have reported corresponding data in SGA populations. Objective We aimed to evaluate the characteristics of the gut microbiota of term SGA infants and the associations between the gut microbiota in SGA infants and neurodevelopmental outcomes at 6 months of age. Methods Fecal samples were collected on days 1, 3, 5, and 7 from term SGA and AGA infants born between June 2020 and June 2021 at the Peking University First Hospital. 16S ribosomal deoxyribonucleic acid amplicon sequencing was used to analyze the fecal microbiota. We followed up for 6 months and used the Ages and Stages Questionnaires-3 (ASQ-3) to evaluate the neurodevelopmental outcomes among SGA infants. Results A total of 162 neonates were enrolled, with 41 SGA infants (25.3%) in the study group and 121 AGA infants (74.7%) in the control group. The gut microbial diversity in the SGA group was lower than that in the AGA group on days 1, 3, 5, and 7. Non-metric multidimensional scaling and analysis of similarities showed significant differences between the two groups. The SGA group had increased relative abundances of Ralstonia (3, 5, and 7 days) and Clostridium (3 and 7 days). The dominant microorganisms of the SGA group were Ralstonia on day 1, Escherichia_Shigella on days 3 and 7, and Clostridia on day 5. We found that the gut microbial diversity of SGA infants with poor communication scores was higher than that of SGA infants with good communication scores on day 3. Fine motor scores were negatively correlated with the relative abundance of Bacteroides_fragilis on day 1. A negative correlation was observed between gross motor scores and relative abundance of Clostridium_saccharobutylicum on day 7. Bacteroidota, Bacteroidia, Bacteroides, and Bacteroides_fragilis were the dominant microorganisms in the good communication score group on day 7. Communication scores were positively correlated with the relative abundance of Bacteroidota, Bacteroides, and Bacteroides_fragilis on day 7. Conclusion The gut microbial diversity of term SGA infants was significantly lower in the first week of life than that of term AGA infants. Certain pathogenic and conditional pathogenic bacteria, such as Escherichia_Shigella, Ralstonia and Clostridium increased or formed the dominant microbiota in SGA infants. Alpha diversity, Bacteroidota, Bacteroides, Bacteroides_fragilis, and Clostridium_saccharobutylicum found in SGA infants may be associated with neurodevelopmental outcomes at 6 months of age, indicating possible therapeutic targets for clinical intervention.
Collapse
|
35
|
Abstract
BACKGROUND Probiotics are live micro-organisms that may give a beneficial physiological effect when administered in adequate amounts. Some trials show that probiotic strains can prevent respiratory infections. Even though our previously published review showed the benefits of probiotics for acute upper respiratory tract infections (URTIs), several new studies have been published. This is an update of a review first published in 2011 and updated in 2015. OBJECTIVES To assess the effectiveness and safety of probiotics (any specified strain or dose), compared with placebo or no treatment, in the prevention of acute URTIs in people of all ages, at risk of acute URTIs. SEARCH METHODS We searched CENTRAL (2022, Issue 6), MEDLINE (1950 to May week 2, 2022), Embase (1974 to 10 May 2022), Web of Science (1900 to 10 May 2022), the Chinese Biomedical Literature Database, which includes the China Biological Medicine Database (from 1978 to 10 May 2022), the Chinese Medicine Popular Science Literature Database (from 2000 to 10 May 2022), and the Master's Degree Dissertation of Beijing Union Medical College Database (from 1981 to 10 May 2022). We searched the World Health Organization International Clinical Trials Registry Platform and ClinicalTrials.gov for completed and ongoing trials on 10 May 2022. SELECTION CRITERIA We included individual randomised controlled trials (RCTs) and cluster-RCTs comparing probiotics with placebo or no treatment to prevent acute URTIs. The participants were children, adults, or the elderly in the community, care facilities, schools, or hospitals. Our main outcomes were the number of participants diagnosed with URTIs (at least one event and at least three events), the incidence rate (number of cases/person year) of acute URTIs, and the mean duration of an episode of URTIs. Our secondary outcomes were the number of participants who were absent from childcare centre, school, or work due to acute URTIs; the number of participants who used prescribed antibiotics for acute URTIs; and the number of participants who experienced at least one adverse event from probiotics. We excluded studies if they did not specify acute respiratory infections as 'upper'; studies with more than 50% of participants vaccinated against influenza or other acute URTIs within the last 12 months; and studies with significantly different proportions of vaccinated participants between the probiotics arm and the placebo or no treatment arm. DATA COLLECTION AND ANALYSIS Two review authors independently assessed the eligibility of trials and extracted data using standard Cochrane methodological procedures. We analysed both intention-to-treat and per-protocol data and used a random-effects model. We expressed results as risk ratios (RRs) for dichotomous outcomes and mean differences (MDs) for continuous outcomes, both with 95% confidence intervals (CIs). We assessed the certainty of the evidence using the GRADE approach. MAIN RESULTS We included 23 individual RCTs and one cluster-RCT. As one of the individual RCTs did not report outcomes in a usable way, we could only meta-analyse data from 23 trials, involving a total of 6950 participants including children (aged from one month to 11 years old), adults (mean age 37.3), and older people (mean age 84.6 years). One trial reported 22.5% flu-vaccine participants within the last 12 months, and 25.4% flu-vaccine participants during the intervention. Probiotics were more likely to be given with milk-based food in children; administered in powder form in adults; and given with milk-based food or in capsules in the elderly. Most of the studies used one or two strains (e.g. Lactobacillus plantarum HEAL9, Lactobacillus paracasei (8700:2 or N1115)) and 109 or 1011 colony-forming units (CFU)/day of probiotics for more than three months. We found that probiotics may reduce the number of participants diagnosed with URTIs (at least one event) (RR 0.76, 95% CI 0.67 to 0.87; P < 0.001; 16 studies, 4798 participants; low-certainty evidence); likely reduce the number of participants diagnosed with URTIs (at least three events) (RR 0.59, 95% CI 0.38 to 0.91; P = 0.02; 4 studies, 763 participants; moderate-certainty evidence); may reduce the incidence rate (number of cases/person year) of URTIs (rate ratio 0.82, 95% CI 0.73 to 0.92, P = 0.001; 12 studies, 4364 participants; low-certainty evidence); may reduce the mean duration of an episode of acute URTIs (MD -1.22 days, 95% CI -2.12 to -0.33; P = 0.007; 6 studies, 2406 participants; low-certainty evidence); likely reduce the number of participants who used prescribed antibiotics for acute URTIs (RR 0.58, 95% CI 0.42 to 0.81; P = 0.001; 6 studies, 1548 participants; moderate-certainty evidence); and may not increase the number of participants who experienced at least one adverse event (RR 1.02, 95% CI 0.90 to 1.15; P = 0.79; 8 studies, 2456 participants; low-certainty evidence). Evidence showing a decrease in the number of people absent from childcare centre, school, or work due to acute URTIs with probiotics is very uncertain (RR 0.14, 95% CI 0.03 to 0.59; 1 study, 80 participants; very low-certainty evidence). Adverse events from probiotics were minor, and most commonly gastrointestinal symptoms, such as vomiting, flatulence, diarrhoea, and bowel pain. AUTHORS' CONCLUSIONS: Overall, we found that probiotics were better than placebo or no treatment in preventing acute URTIs.
Collapse
Affiliation(s)
- Yunli Zhao
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Bi Rong Dong
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiukui Hao
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Study on the Relationship between AAD and Clinical Features in Emergency Ward Patients and the Application Effect of Probiotics. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6164843. [PMID: 35919499 PMCID: PMC9293535 DOI: 10.1155/2022/6164843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
In order to investigate the influencing factors of AAD in the emergency ward and the clinical effect of probiotics, 100 AAD patients admitted to the emergency ward of our hospital from January 2021 to January 2022 and 100 healthy physical examination subjects are selected for conducting clinical trials. The subjects are, respectively, included in the AAD group and the healthy group. By using the random number table method, 100 patients in the AAD group are randomly divided into the probiotic group and traditional group, with 50 patients in each group. The traditional group receives conventional treatment, and the probiotic group is treated with Clostridium caseinate bifidobacterial probiotics additionally. Multivariate logistic analysis is performed on the risk factors for AAD, and the comparison of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and the intestinal microflora of the different groups before and after treatment is conducted. The experimental result reveals that probiotics treatment for AAD patients can reduce inflammatory response and promote intestinal colonization of beneficial bacteria such as bifidobacteria.
Collapse
|
37
|
Infection prevention requirements for the medical care of immunosuppressed patients: recommendations of the Commission for Hospital Hygiene and Infection Prevention (KRINKO) at the Robert Koch Institute. GMS HYGIENE AND INFECTION CONTROL 2022; 17:Doc07. [PMID: 35707229 PMCID: PMC9174886 DOI: 10.3205/dgkh000410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In Germany, guidelines for hygiene in hospitals are given in form of recommendations by the Commission for Hospital Hygiene and Infection Prevention (Kommission für Krankenhaushygiene und Infektionsprävention, "KRINKO"). The KRINKO and its voluntary work are legitimized by the mandate according to § 23 of the Infection Protection Act (Infektionsschutzgesetz, "IfSG"). The original German version of this document was published in February 2021 and has now been made available to the international professional public in English. The guideline provides recommendations on infection prevention and control for immunocompromised individuals in health care facilities. This recommendation addresses not only measures related to direct medical care of immunocompromised patients, but also management aspects such as surveillance, screening, antibiotic stewardship, and technical/structural aspects such as patient rooms, air quality, and special measures during renovations.
Collapse
|
38
|
Effects of Lacidophilin Tablets, Yogurt, and Bifid Triple Viable Capsules on the Gut Microbiota of Mice with Antibiotic-Associated Diarrhea. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2022; 2022:6521793. [PMID: 35360462 PMCID: PMC8964159 DOI: 10.1155/2022/6521793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022]
Abstract
Antibiotic-associated diarrhea (AAD) is a common morbidity caused by antibiotic use and is characterized by the dysbiosis of the gut microbiota. Several clinical trials have shown that probiotics can prevent AAD. This study aimed at investigating the effects of Lacidophilin tablets (LB), yogurt (YG), and bifid triple viable capsules (BT) on the gut microbiota of mice with AAD. Mice with diarrhea were randomly allocated to treatment groups or the control group and were treated with either LB, YG, BT, or vehicle control. The body weight, diarrhea scores, cecum index, and cecal length were determined. Fecal samples of all mice were analyzed using 16S rRNA high-throughput sequencing. The results showed that LB, YG, and BT significantly decreased the diarrhea scores and inhibited increases in the cecum index and cecal length induced by AAD. In addition, they significantly changed the composition and richness of the gut microbiota. Specifically, they increased the abundance of the phylum Firmicutes and decreased the abundance of the phyla Bacteroidetes and the family Bacteroidaceae. Treatment with LB and YG also decreased the abundance of the phylum Proteobacteria and only LB could mediate the reduced levels of Lactobacillaceae in AAD mice. At the genus level, YG and BT treatment decreased the abundance of Bacteroides or Parasutterella. To our surprise, only LB treatment dramatically increased the abundance of Lactobacillus and decreased that of potential pathogens, such as Bacteroides, Parabacteroides, and Parasutterella, to almost normal values. Our findings indicate that LB, YG, and BT ameliorated diarrhea by regulating the composition and structure of the gut microbiota and that LB plays an important role in regulating the gut microbiota.
Collapse
|
39
|
Murugaiyan J, Kumar PA, Rao GS, Iskandar K, Hawser S, Hays JP, Mohsen Y, Adukkadukkam S, Awuah WA, Jose RAM, Sylvia N, Nansubuga EP, Tilocca B, Roncada P, Roson-Calero N, Moreno-Morales J, Amin R, Kumar BK, Kumar A, Toufik AR, Zaw TN, Akinwotu OO, Satyaseela MP, van Dongen MBM. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics (Basel) 2022; 11:200. [PMID: 35203804 PMCID: PMC8868457 DOI: 10.3390/antibiotics11020200] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 11/24/2022] Open
Abstract
Antibiotic resistance, and, in a broader perspective, antimicrobial resistance (AMR), continues to evolve and spread beyond all boundaries. As a result, infectious diseases have become more challenging or even impossible to treat, leading to an increase in morbidity and mortality. Despite the failure of conventional, traditional antimicrobial therapy, in the past two decades, no novel class of antibiotics has been introduced. Consequently, several novel alternative strategies to combat these (multi-) drug-resistant infectious microorganisms have been identified. The purpose of this review is to gather and consider the strategies that are being applied or proposed as potential alternatives to traditional antibiotics. These strategies include combination therapy, techniques that target the enzymes or proteins responsible for antimicrobial resistance, resistant bacteria, drug delivery systems, physicochemical methods, and unconventional techniques, including the CRISPR-Cas system. These alternative strategies may have the potential to change the treatment of multi-drug-resistant pathogens in human clinical settings.
Collapse
Affiliation(s)
- Jayaseelan Murugaiyan
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - P. Anand Kumar
- Department of Veterinary Microbiology, NTR College of Veterinary Science, Sri Venkateswara Veterinary University, Gannavaram 521102, India;
| | - G. Srinivasa Rao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Science, Sri Venkateswara Veterinary University, Tirupati 517502, India;
| | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1295, 31000 Toulouse, France;
- INSPECT-LB: Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573, Lebanon
- Faculty of Pharmacy, Lebanese University, Beirut 6573, Lebanon
| | | | - John P. Hays
- Department of Medical Microbiology, Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), 3015 GD Rotterdam, The Netherlands;
| | - Yara Mohsen
- Department of Epidemiology, High Institute of Public Health, Alexandria University, Alexandria 21544, Egypt;
- Infectious Disease Clinical Pharmacist, Antimicrobial Stewardship Department, International Medical Center Hospital, Cairo 11511, Egypt
| | - Saranya Adukkadukkam
- Department of Biological Sciences, SRM University-AP, Guntur District, Amaravati 522240, India;
| | - Wireko Andrew Awuah
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Ruiz Alvarez Maria Jose
- Research Coordination and Support Service, National Institute of Health (ISS) Viale Regina -Elena, 299, 00161 Rome, Italy;
| | - Nanono Sylvia
- Infectious Diseases Institute (IDI), College of Health Sciences, Makerere University, Kampala 7072, Uganda;
| | | | - Bruno Tilocca
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Paola Roncada
- Department of Health Science, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (B.T.); (P.R.)
| | - Natalia Roson-Calero
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Javier Moreno-Morales
- ISGlobal, Hospital Clínic-Universitat de Barcelona, 08036 Barcelona, Spain; (N.R.-C.); (J.M.-M.)
| | - Rohul Amin
- James P Grant School of Public Health, BRAC University, Dhaka 1212, Bangladesh;
| | - Ballamoole Krishna Kumar
- Nitte (Deemed to be University), Division of Infectious Diseases, Nitte University Centre for Science Education and Research, Deralakatte, Mangalore 575018, India;
| | - Abishek Kumar
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, India;
| | - Abdul-Rahman Toufik
- Faculty of Medicine, Sumy State University, 40007 Sumy, Ukraine; (W.A.A.); (A.-R.T.)
| | - Thaint Nadi Zaw
- Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK;
| | - Oluwatosin O. Akinwotu
- Department of Microbiology and Biotechnology Centre, Maharaja Sayajirao University of Baroda, Vadodara 390002, India;
- Environmental and Biotechnology Unit, Department of Microbiology, University of Ibadan, 200132 Ibadan, Nigeria
| | | | | |
Collapse
|
40
|
Supplementation of Bacillus sp. DU-106 Alleviates Antibiotic-Associated Diarrhea in Association with the Regulation of Intestinal Microbiota in Mice. Probiotics Antimicrob Proteins 2022; 14:372-383. [DOI: 10.1007/s12602-022-09906-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2022] [Indexed: 02/08/2023]
|
41
|
New Wild-Type Lacticaseibacillus rhamnosus Strains as Candidates to Manage Type 1 Diabetes. Microorganisms 2022; 10:microorganisms10020272. [PMID: 35208726 PMCID: PMC8875344 DOI: 10.3390/microorganisms10020272] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/16/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
The incidence of type 1 diabetes (T1D) has been dramatically increased in developed countries, and beyond the genetic impact, environmental factors, including diet, seem to play an important role in the onset and development of the disease. In this vein, five Lacticaseibacillus rhamnosus, isolated from traditional fermented Greek products, were screened for potential probiotic properties, aiming at maintaining gut homeostasis and antidiabetic capability to alleviate T1D symptoms. L. rhamnosus cell-free supernatants induced strong growth inhibitory activity against common food spoilage and foodborne pathogenic microorganisms, associated with several diseases, including T1D, and were also able to inhibit α-glucosidase activity (up to 44.87%), a promising property for alternatives to the antidiabetic drugs. In addition, survival rates up to 36.76% were recorded during the application of the static in vitro digestion model. The strains had no hemolytic activity and were sensitive to common antibiotics suggested by the European Food and Safety Association, apart from chloramphenicol. However, it is highly unlikely that the resistance has been acquired. In conclusion, our results suggest a great health-promoting potential of the newly isolated wild-type L. rhamnosus strains, but further confirmation of their efficiency in experimental animal models is considered an essential next research step.
Collapse
|
42
|
Duboux S, Van Wijchen M, Kleerebezem M. The Possible Link Between Manufacturing and Probiotic Efficacy; a Molecular Point of View on Bifidobacterium. Front Microbiol 2022; 12:812536. [PMID: 35003044 PMCID: PMC8741271 DOI: 10.3389/fmicb.2021.812536] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Probiotics for food or supplement use have been studied in numerous clinical trials, addressing a broad variety of diseases, and conditions. However, discrepancies were observed in the clinical outcomes stemming from the use of lactobacillaceae and bifidobacteria strains. These differences are often attributed to variations in the clinical trial protocol like trial design, included target population, probiotic dosage, or outcome parameters measured. However, a contribution of the methods used to produce the live bioactive ingredients should not be neglected as a possible additional factor in the observed clinical outcome variations. It is well established that manufacturing conditions play a role in determining the survival and viability of probiotics, but much less is known about their influence on the probiotic molecular composition and functionality. In this review, we briefly summarize the evidence obtained for Lacticaseibacillus rhamnosus GG and Lactiplantibacillus plantarum WCFS1, highlighting that expression and presence of probiotic niche factor (NF) and/or effector molecules (EM) may be altered during production of those two well-characterized lactobacillaceae probiotic strains. Subsequently, we summarize in more depth what is the present state of knowledge about bifidobacterial probiotic NF and EM; how their expression may be modified by manufacturing related environmental factors and how that may affect their biological activity in the host. This review highlights the importance of gathering knowledge on probiotic NF and EM, to validate them as surrogate markers of probiotic functionality. We further propose that monitoring of validated NF and/or EM during production and/or in the final preparation could complement viable count assessments that are currently applied in industry. Overall, we suggest that implementation of molecular level quality controls (i.e., based on validated NF and EM), could provide mode of action based in vitro tests contributing to better control the health-promoting reliability of probiotic products.
Collapse
Affiliation(s)
- Stéphane Duboux
- Nestlé Research, Lausanne, Switzerland.,Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Myrthe Van Wijchen
- Nestlé Research, Lausanne, Switzerland.,Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| | - Michiel Kleerebezem
- Host-Microbe Interactomics Group, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
43
|
Hay W, Steinke L, Foster L. Complementary and Alternative Medicine. Fam Med 2022. [DOI: 10.1007/978-3-030-54441-6_143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
44
|
Dudnyk VM, Andrikevych II, Khromykh K, Mantak HI, Rudenko HM. ANTIBIOTIC ASSOCIATIVE DISORDERS OF THE MICROBIOCENOSIS OF THE COLON IN INFANTS WITH ACUTE RESPIRATORY DISEASES. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2022; 75:1323-1327. [PMID: 35758452 DOI: 10.36740/wlek202205217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
OBJECTIVE The aim: Study the effect of antibiotics of different groups on the condition of the colon microflora in infants with acute respiratory pathology. PATIENTS AND METHODS Materials and methods: 140 infants with acute respiratory pathology were examined. Clinical, laboratory and instrumental examination, assessment of the functional state of all organs and systems, chest x ray, clinical signs of the colon microbiocenosis violation, analysis of bacteriological examination and immunological studies of local colon immunity (immunoglobulin concentration (sIgA, IgA, IgG, IgM) in coprofiltrates) were done. RESULTS Results: The negative effect of antibiotics of different pharmacological groups on the colon microflora state in infants with acute respiratory diseases has been established. The indigenous microflora of the colon is most inhibited by drugs from the group of 3rd generation cephalosporins, aminoglycosides and their combination. While cephalosporins 1-2nd generations, penicillins and macrolides to a lesser extent affect the state of the microbiocenosis of the colon. The use of two courses of antibacterial therapy to a greater extent disrupts the microbiocenosis of the colon in the examined children, compared with one course of therapy. In commune acquired pneumonia and acute complicated bronchiolitis in infants on the background of antibiotic therapy there is a probable decrease in secretory immunoglobulin in coprofiltrate (sIgA), compared with healthy children (p <0.05). CONCLUSION Conclusions: The analysis of the obtained results showed that antibiotic therapy negatively affects not only the condition of the colon microflora in the examined children, but also suppresses humoral factors of local immunity of the colonic mucosa. Key words: digestive tract microbiocenosis, antibiotic therapy, children.
Collapse
Affiliation(s)
| | | | | | - Halyna I Mantak
- NATIONAL PIROGOV MEMORIAL MEDICAL UNIVERSITY, VINNYTSIA, UKRAINE
| | | |
Collapse
|
45
|
Turner RB, Lehtoranta L, Hibberd A, Männikkö S, Zabel B, Yeung N, Huttunen T, Burns FR, Lehtinen MJ. Effect of Bifidobacterium animalis spp. lactis Bl-04 on Rhinovirus-Induced Colds: A Randomized, Placebo-Controlled, Single-Center, Phase II Trial in Healthy Volunteers. EClinicalMedicine 2022; 43:101224. [PMID: 34927036 PMCID: PMC8649651 DOI: 10.1016/j.eclinm.2021.101224] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/11/2021] [Accepted: 11/17/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND This study was designed to assess the efficacy of Bifidobacterium animalis ssp. lactis (Bl-04) for prevention of rhinovirus colds and to explore the interactions between the probiotic, the viral infection, the host response and the host microbiome. METHODS The effect of ingestion of the probiotic Bl-04 was evaluated in a randomized, double-blinded rhinovirus (RV) challenge study. Healthy volunteers recruited from a university community in USA were randomized 1:1 using a computer generated code to ingest either Bl-04 (n=165) or placebo (n=169) for 28 days and were then challenged with RV-A39, and followed for 14 days. All study interactions and sample collection occurred in dedicated clinical research space. The primary analysis was the effect of the probiotic on the incidence of RV-associated illness. (Trial registration: NCT02679807, study complete). FINDINGS The first cohort of volunteers was randomized on March 14, 2016 and the last (5th) cohort was randomized on March 12, 2018. Sixty-three (56%, 95% CI [47%; 66%]) of the 112 subjects in the active group and 60 (50%,95% CI [41%; 59%]) of the 120 subjects in the placebo group had a protocol-defined rhinovirus-associated illness (χ2=0·91, p=0·34). The point estimate of the difference in illness (active-placebo) is 6.3% (95% CI -6.7;19.1). There were no adverse events that were judged as definitely or probably related to the study product. INTERPRETATION In this study there was no effect of orally administered Bl-04 on the occurrence of RV-associated illness. FUNDING Danisco Sweeteners Oy (now IFF Health & Biosciences).
Collapse
Affiliation(s)
- Ronald B. Turner
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA
- Corresponding To: 512 Rosemont Drive, Charlottesville, VA 22903
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sun N, Xue Y, Wei S, Wu B, Wang H, Zeng D, Zhao Y, Khalique A, Pan K, Zeng Y, Shu G, Jing B, Ni X. Compound Probiotics Improve Body Growth Performance by Enhancing Intestinal Development of Broilers with Subclinical Necrotic Enteritis. Probiotics Antimicrob Proteins 2021; 15:558-572. [PMID: 34735679 DOI: 10.1007/s12602-021-09867-4] [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] [Accepted: 10/18/2021] [Indexed: 11/25/2022]
Abstract
The aim of this study is to explore whether or not the combined application of BS15 and H2 is capable to have a more effective control effect on SNE in broilers. A total of 240 1-day-old female chickens were randomly divided into 5 groups: (a) basal diet in negative control group (NC group); (b) basal diet + SNE infection (coccidiosis vaccine + CP) (PC group); (c) basal diet + SNE infection + H2 pre-treatment (BT group); (d) basal diet + SNE infection + BS15 pre-treatment (LT group); and (e) basal diet + SNE infection + H2 pre-treatment + BS15 pre-treatment (MT group). The results showed the MT group had the most positive effect on inhibiting the negative effect of growth performance at 42 days of age. In the detection of the NC, PC, and MT group indicators at 28 days of age, we found that MT group significantly promoted ileum tissue development of broilers, and the ileum of broilers in the MT group formed a flora structure different from NC and PC, although it was found that the MT group had no effect on the butyrate level in the cecum, but it could affect the serum immune level, such as significantly reducing the level of pro-inflammatory cytokine IL-8 and increasing the content of immunoglobulin IgM and IgG. In conclusion, the composite preparation of Lactobacillus johnsonii BS15 and Bacillus licheniformis H2 could effectively improve the growth performance against SNE broilers, which is possibly caused by the improvement of the immune levels, the reduction of inflammation levels, and the promotion of the intestinal development.
Collapse
Affiliation(s)
- Ning Sun
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Xue
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Siyi Wei
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bangyuan Wu
- College of Life Sciences, China West Normal University, Nanchong, Sichuan, China
| | - Hesong Wang
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.,Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Ying Zhao
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Yan Zeng
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Shu
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
47
|
D’Agostin M, Squillaci D, Lazzerini M, Barbi E, Wijers L, Da Lozzo P. Invasive Infections Associated with the Use of Probiotics in Children: A Systematic Review. CHILDREN-BASEL 2021; 8:children8100924. [PMID: 34682189 PMCID: PMC8534463 DOI: 10.3390/children8100924] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022]
Abstract
Although the effectiveness of probiotics has only been proven in specific conditions, their use in children is massively widespread because of their perception as harmless products. Recent evidence raises concerns about probiotics' safety, especially but not only in the paediatric population due to severe opportunistic infections after their use. This review aimed at summarising available case reports on invasive infections related to probiotics' use in children. For this purpose, we assessed three electronic databases to identify papers describing paediatric patients with documented probiotic-derived invasive infections, with no language restrictions. A total of 49 case reports from 1995 to June 2021 were identified. The infections were caused by Lactobacillus spp. (35%), Saccharomyces spp. (29%), Bifidobacterium spp. (31%), Bacillus clausii (4%), and Escherichia coli (2%). Most (80%) patients were younger than 2 years old and sepsis was the most observed condition (69.4%). All the patients except one had at least one condition facilitating the development of invasive infection, with prematurity (55%) and intravenous catheter use (51%) being the most frequent. Three (6%) children died. Given the large use of probiotics, further studies aiming at evaluating the real incidence of probiotic-associated systemic infections are warranted.
Collapse
Affiliation(s)
- Martina D’Agostin
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
| | - Domenica Squillaci
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
- Correspondence:
| | - Marzia Lazzerini
- Institute of Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65/1, 34137 Trieste, Italy;
| | - Egidio Barbi
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
- Institute of Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria 65/1, 34137 Trieste, Italy;
| | - Lotte Wijers
- Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Prisca Da Lozzo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127 Trieste, Italy; (M.D.); (E.B.); (P.D.L.)
| |
Collapse
|
48
|
Genetically encoded probiotic EcN 1917 alleviates alcohol-induced acute liver injury and restore gut microbiota homeostasis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104661] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
49
|
Maity C, Gupta AK. Therapeutic efficacy of probiotic Alkalihalobacillus clausii 088AE in antibiotic-associated diarrhea: A randomized controlled trial. Heliyon 2021; 7:e07993. [PMID: 34585011 PMCID: PMC8453216 DOI: 10.1016/j.heliyon.2021.e07993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/26/2021] [Accepted: 09/10/2021] [Indexed: 01/22/2023] Open
Abstract
The use of probiotics in gastrointestinal ailments has shown therapeutic effects. The imbalance of the microbiota caused by antibiotic treatment or others has been shown to be restored to normal with probiotic treatment. In this study, a genomically and phenotypically safe probiotic Alkalihalobacillus clausii 088AE has been evaluated for ameliorating antibiotic-associated diarrhea (AAD) in pediatrics (PE, n = 60, 2-10 years), adolescent and adults (AA, n = 60, 11-65 years) through a randomized controlled clinical trial. A. clausii 088AE was administered for seven days (PE, 4 and AA, 6 billion/day) and primary and secondary endpoints were evaluated on different visits. Compared to the respective placebo arms, A. clausii 088AE improved the diarrheal (time to last unformed stool and diarrheal frequency) conditions in children, adolescents and adults. A. clausii 088AE treatment decreased AAD-severity score on visit 5 in both pediatric (0.12 ± 0.33, 12.39 folds), adult and adolescent (0.54 ± 0.36, 2.34 folds) groups compared to those respective placebo arm (p < 0.05). A. clausii 088AE was well tolerated, did not cause significant changes in vital and clinical safety parameters and subjects reported no adverse effects or serious adverse reactions. A. clausii 088AE is safe and therapeutically effective against AAD, reducing onset of diarrhea and related severity symptoms including abdominal discomfort and pain, bloating and flatulence. A. clausii 088AE may be recommended as a live bio-therapeutic agent for improving clinical pathophysiology of gastrointestinal ailments, in particular antibiotic-associated diarrhea and related symptoms.
Collapse
Affiliation(s)
- Chiranjit Maity
- Probiotics Laboratory, Advanced Enzyme Technologies Ltd., 5Th Floor, A-Wing, Sun Magnetica, LIC Service Road, Louiswadi, Thane (W), 400 604, Maharashtra, India
| | - Anil Kumar Gupta
- Probiotics Laboratory, Advanced Enzyme Technologies Ltd., 5Th Floor, A-Wing, Sun Magnetica, LIC Service Road, Louiswadi, Thane (W), 400 604, Maharashtra, India
| |
Collapse
|
50
|
Is Recent Exposure to Antibiotics a Risk Factor for Hospitalisation in Korean Children with Acute Non-Bacterial Gastroenteritis? A Nationwide Population-Based Study. CHILDREN 2021; 8:children8090809. [PMID: 34572241 PMCID: PMC8467275 DOI: 10.3390/children8090809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to evaluate the effect of recent antibiotic therapy and probiotics on hospitalisation in children with acute gastroenteritis. Using a retrospective study design, data from the population aged up to 18 years were collected from the Korean National Health Insurance Service-National Sample Cohort. The duration of antibiotic therapy within 14 days of the index visit, prescription of probiotics at initial presentation, the effect size of antibiotic exposure on hospitalisation, and its modification by probiotics were assessed. Of 275,395 patients with acute gastroenteritis, 51,008 (18.5%) had prior exposure to antibiotics. Hospitalisation within 7 days of the index visit was positively associated with exposure to antibiotics (p-trend < 0.001). The prescription of probiotics (as a main effect; odds ratio, 0.80; 95% confidence interval 0.72–0.87) was associated with a decreased risk of hospitalisation. Prior exposure to antibiotics might be a significant risk factor for hospitalisation in children presenting with acute gastroenteritis. This may be favourably modified by administering probiotics at the initial presentation.
Collapse
|