1
|
Malan-Müller S, Martín-Hernández D, Caso JR, Matthijnssens J, Rodríguez-Urrutia A, Lowry CA, Leza JC. Metagenomic symphony of the intestinal ecosystem: How the composition affects the mind. Brain Behav Immun 2024; 123:510-523. [PMID: 39368785 DOI: 10.1016/j.bbi.2024.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024] Open
Abstract
Mental health disorders and neurodegenerative diseases place a heavy burden on patients and societies, and, although great strides have been made to understand the pathophysiology of these conditions, advancement in drug development is lagging. The importance of gastrointestinal health in maintaining overall health and preventing disease is not a new concept. Hundreds of years ago, healers from various cultures and civilizations recognized the crucial role of the gut in sustaining health. More than a century ago, scientists began exploring the restorative effects of probiotics, marking the early recognition of the importance of gut microbes. The omics era brought more enlightenment and enabled researchers to identify the complexity of the microbial ecosystems we harbour, encompassing bacteria, eukaryotes (including fungi), archaea, viruses, and other microorganisms. The extensive genetic capacity of the microbiota is dynamic and influenced by the environment. The microbiota therefore serves as a significant entity within us, with evolutionarily preserved functions in host metabolism, immunity, development, and behavior. The significant role of the bacterial gut microbiome in mental health and neurodegenerative disorders has been realized and described within the framework of the microbiota-gut-brain axis. However, the bacterial members do not function unaccompanied, but rather in concert, and there is a substantial knowledge gap regarding the involvement of non-bacterial microbiome members in these disorders. In this review, we will explore the current literature that implicates a role for the entire metagenomic ensemble, and how their complex interkingdom relationships could influence CNS functioning in mental health disorders and neurodegenerative diseases.
Collapse
Affiliation(s)
- Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain.
| | - David Martín-Hernández
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Javier R Caso
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Jelle Matthijnssens
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Division of Clinical and Epidemiological Virology, Laboratory of Viral Metagenomics, Leuven, Belgium
| | - Amanda Rodríguez-Urrutia
- Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain; Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Catalonia, Spain; Group of Psychiatry, Mental Health and Addictions, Vall d'Hebron Research Institute (VHIR), Barcelona, Catalonia, Spain; Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain; Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| |
Collapse
|
2
|
Kim S, Cho J, Keum GB, Kwak J, Doo H, Choi Y, Kang J, Kim H, Chae Y, Kim ES, Song M, Kim HB. Investigation of the impact of multi-strain probiotics containing Saccharomyces cerevisiae on porcine production. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:876-890. [PMID: 39398307 PMCID: PMC11466735 DOI: 10.5187/jast.2024.e79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 10/15/2024]
Abstract
A balanced intestinal microbiome controls intestinal bacterial diseases, helps regulate immunity, and digests and utilizes nutrients, ultimately having a positive effect on the productivity of industrial animals. Yeasts help in the digestion process by breaking down indigestible fibers and producing organic acids, vitamins, and minerals. In particular, polysaccharides such as beta-glucan and mannan-oligosaccharides, which are present in the cell wall of yeast, inhibit the adhesion of pathogens to the surface of the gastrointestinal tract and increase resistance to disease to help maintain and improve intestinal health. Among the yeast additives used in animal feed, Saccharomyces cerevisiae is one of the most commonly used probiotics. However, it does not naturally reside in the intestine, so if it is supplied in combination with other species of probiotics that can compensate for it, many benefits and synergies can be expected for pigs in terms of maintaining intestinal health such as supplementing the immune system and improving digestion. A number of previous studies have demonstrated that dietary complex probiotic supplementation has growth-promoting effects in pigs, suggesting that multiple strains of probiotics may be more effective than single strain probiotics due to their additive and synergistic effects. In practice, however, the effects of complex probiotics are not always consistent, and can be influenced by a variety of factors. Therefore, this review comprehensively examines and discusses the literature related to the effects of complex probiotics using Saccharomyces cerevisiae in pig production.
Collapse
Affiliation(s)
- Sheena Kim
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Jinho Cho
- Division of Food and Animal Science,
Chungbuk National University, Cheongju 28644, Korea
| | - Gi Beom Keum
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Jinok Kwak
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Hyunok Doo
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Yejin Choi
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Juyoun Kang
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Haram Kim
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Yeongjae Chae
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Eun Sol Kim
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| | - Minho Song
- Division of Animal and Dairy Science,
Chungnam National University, Daejeon 31434, Korea
| | - Hyeun Bum Kim
- Department of Animal Biotechnology,
Dankook University, Cheonan 31116, Korea
| |
Collapse
|
3
|
Wang L, Cao Y, Lou E, Zhao X, Chen X. The role of gut fungi in Clostridioides difficile infection. Biomed J 2024; 47:100686. [PMID: 38086471 PMCID: PMC11220531 DOI: 10.1016/j.bj.2023.100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 06/10/2024] Open
Abstract
Clostridioides difficile, the etiological agent of C. difficile infection (CDI), elicits a spectrum of diarrheal symptoms with varying severity and the potential to result in severe complications such as colonic perforation, pseudomembranous colitis, and toxic megacolon. The perturbation of gut microbiome, often triggered by antibiotic usage, represents the primary factor augmenting the risk of CDI. This underscores the significance of interactions between C. difficile and the microbiome in determining pathogen adaptability. In recent years, researchers have increasingly recognized the pivotal role played by intestinal microbiota in host health and its therapeutic potential as a target for medical interventions. While extensive evidence has been established regarding the involvement of gut bacteria in CDI, our understanding of symbiotic interactions between hosts and fungi within intestinal microbiota remains limited. Herein, we aim to comprehensively elucidate both composition and key characteristics of gut fungal communities that significantly contribute to CDI, thereby enhancing our comprehension from pharmacological and biomarker perspectives while exploring their prospective therapeutic applications for CDI.
Collapse
Affiliation(s)
- Lamei Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Eddie Lou
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xuanyin Zhao
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Wang M, Gao C, Lessing DJ, Chu W. Saccharomyces cerevisiae SC-2201 Attenuates AOM/DSS-Induced Colorectal Cancer by Modulating the Gut Microbiome and Blocking Proinflammatory Mediators. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10228-0. [PMID: 38329696 DOI: 10.1007/s12602-024-10228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Colorectal cancer is the third most common cancer in the world today, and studies have shown that the ratio of Candida to Saccharomyces cerevisiae increased, and the abundance of S. cerevisiae in the intestines of patients with colorectal cancer decreased, which suggests that there is an imbalance in the proportion of fungi in the intestines of patients with colorectal cancer. The objective of this study was to screen S. cerevisiae isolate from traditional Chinese fermentation starters and assess its ability to ameliorate dysbiosis and to alleviate the carcinogenic process of azoxymethane/dextran sodium sulfate-induced colorectal cancer in mice model. S. cerevisiae strain SC-2201 was isolated and exhibited probiotic properties, including the ability to survive in an acidic pH environment and in the presence of bile salts in the gastrointestinal tract, as well as antioxidant activities. Oral administration of S. cerevisiae SC-2201 not only alleviated weight loss but also reduced colonic shortening and histological damage in azoxymethane/dextran sodium sulfate-induced colorectal cancer in mice. Furthermore, the administration of S. cerevisiae SC-2201 suppressed the expression of proinflammatory mediators, such as interleukin-1β, interleukin-6, cyclooxygenase-2, vascular endothelial growth factor, nucleotide-binding domain, leucine-rich repeat, and pyrin domain-containing protein 3. Specifically, the analysis of gut bacteriome showed a significant decrease in Bacteroidota and Campylobacterota levels, as well as an increase in Proteobacteria level in the colorectal cancer group, which was alleviated by supplementation with S. cerevisiae SC-2201. The analysis of the mycobiome revealed a significant increase in the levels of Basidiomycota, Apiosordaria, Naganishia, and Taphrina genera in the colorectal cancer group, which were alleviated after supplementation with S. cerevisiae SC-2201. However, the levels of Xenoramularia, Entoloma, and Keissleriella were significantly increased after administration with S. cerevisiae SC-2201. Overall, the findings of this study demonstrate that S. cerevisiae SC-2201 possesses potential probiotic properties and can effectively attenuate the development of colorectal cancer, highlighting its cancer-preventive potential. This is the first report of a S. cerevisiae strain isolated from traditional Chinese fermentation starters which showed good probiotic properties, and mitigated azoxymethane/dextran sodium sulfate-induced colorectal cancer by modulating the gut microbiome and blocking proinflammatory mediators in mice.
Collapse
Affiliation(s)
- Minyu Wang
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Jiangsu Province, Nanjing, 210009, People's Republic of China
| | - Chongzheng Gao
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Jiangsu Province, Nanjing, 210009, People's Republic of China
| | - Duncan James Lessing
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Jiangsu Province, Nanjing, 210009, People's Republic of China
| | - Weihua Chu
- School of Life Science and Technology, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Jiangsu Province, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
5
|
Krishnamurthy HK, Pereira M, Bosco J, George J, Jayaraman V, Krishna K, Wang T, Bei K, Rajasekaran JJ. Gut commensals and their metabolites in health and disease. Front Microbiol 2023; 14:1244293. [PMID: 38029089 PMCID: PMC10666787 DOI: 10.3389/fmicb.2023.1244293] [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: 06/22/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose of review This review comprehensively discusses the role of the gut microbiome and its metabolites in health and disease and sheds light on the importance of a holistic approach in assessing the gut. Recent findings The gut microbiome consisting of the bacteriome, mycobiome, archaeome, and virome has a profound effect on human health. Gut dysbiosis which is characterized by perturbations in the microbial population not only results in gastrointestinal (GI) symptoms or conditions but can also give rise to extra-GI manifestations. Gut microorganisms also produce metabolites (short-chain fatty acids, trimethylamine, hydrogen sulfide, methane, and so on) that are important for several interkingdom microbial interactions and functions. They also participate in various host metabolic processes. An alteration in the microbial species can affect their respective metabolite concentrations which can have serious health implications. Effective assessment of the gut microbiome and its metabolites is crucial as it can provide insights into one's overall health. Summary Emerging evidence highlights the role of the gut microbiome and its metabolites in health and disease. As it is implicated in GI as well as extra-GI symptoms, the gut microbiome plays a crucial role in the overall well-being of the host. Effective assessment of the gut microbiome may provide insights into one's health status leading to more holistic care.
Collapse
Affiliation(s)
| | | | - Jophi Bosco
- Vibrant America LLC., San Carlos, CA, United States
| | | | | | | | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States
| | | |
Collapse
|
6
|
Maturana M, Castillejos L, Martin-Orue SM, Minel A, Chetty O, Felix AP, Adib Lesaux A. Potential benefits of yeast Saccharomyces and their derivatives in dogs and cats: a review. Front Vet Sci 2023; 10:1279506. [PMID: 37954670 PMCID: PMC10634211 DOI: 10.3389/fvets.2023.1279506] [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: 08/18/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Yeast Saccharomyces and its derivatives have been largely used in livestock and poultry nutrition for their potential positive impact on growth, performance, and general health. Originally included in animal diets as a source of protein, yeasts can also offer a wide range of by-products with interesting bioactive compounds that would confer uses beyond nutrition. Although its supplementation in livestock, poultry and even in humans is well documented, the available body of literature on the use of yeast and its derivatives in companion animals' food, mainly dogs and cats' diets, is still developing. Despite this, gut microbiota modulation, immune system enhancement or decreasing of potentially pathogenic microorganisms have been reported in pets when using these products, highlighting their possible role as probiotics, prebiotics, and postbiotics. This review attempts to provide the reader with a comprehensive on the effects of Saccharomyces and its derivatives in pets and the possible mechanisms that confer their functional properties.
Collapse
Affiliation(s)
- Marta Maturana
- Animal Nutrition and Welfare Service (SNiBA), Department of Animal and Food Science, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Lorena Castillejos
- Animal Nutrition and Welfare Service (SNiBA), Department of Animal and Food Science, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Susana M. Martin-Orue
- Animal Nutrition and Welfare Service (SNiBA), Department of Animal and Food Science, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Anaelle Minel
- Department of Research & Development, Phileo by Lesaffre, Marcq-en-Baroeul, France
| | - Olivia Chetty
- Department of Research & Development, Phileo by Lesaffre, Marcq-en-Baroeul, France
| | - Ananda P. Felix
- Department of Animal Science, Federal University of Paraná, Curitiba, Brazil
| | - Achraf Adib Lesaux
- Department of Research & Development, Phileo by Lesaffre, Marcq-en-Baroeul, France
| |
Collapse
|
7
|
Sampath V, Sureshkumar S, Kim IH. The Efficacy of Yeast Supplementation on Monogastric Animal Performance-A Short Review. Life (Basel) 2023; 13:2037. [PMID: 37895419 PMCID: PMC10608604 DOI: 10.3390/life13102037] [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: 09/18/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Due to a continual growth in the world's population and the prohibition of antibiotics in animal production, the livestock industry faces significant challenges in the global demand for meat, eggs, and dairy products. The growing demand for organic products and the prohibition on antibiotic growth promoters (AGPs) have compelled animal nutrition experts to search for natural substitutes that include medical plants and beneficial microorganisms. Natural feed additives like probiotics are found to be more effective than AGPs in reducing the load of harmful intestinal pathogens. One of the probiotics that has generated considerable interest since ancient times is yeast. Yeast is used as a supplement in animal feeds due to its relatively high protein, amino acid, energy, and micronutrient content. Yeast byproducts such as yeast cells and cell walls contain nutraceutical compounds (i.e., β-glucans, mannooligosaccharides, and nucleotides) and have been shown to improve animal growth performance and health. Though the application of yeast supplements has been reviewed to date, only a scarce amount of information exists on the yeast-derived products in non-ruminant nutrition. Additionally, it is difficult for nutritionists to differentiate the characteristics, composition, and optimal feeding among the diverse number of yeast-containing products. Due to the increasing popularity of using yeast-based products in animal feeds, the development of analytical approaches to estimate yeast and its components in these products is greatly needed. Thus, in this review, we intend to provide current knowledge of different categories of commercially available yeast and yeast-derived additives, along with their role in improving animal growth performance and health, their proposed mechanisms of action, and the challenges of quantifying yeast content and biologically active components.
Collapse
Affiliation(s)
- Vetriselvi Sampath
- Department of Animal Resource and Science, Dankook University, Cheonan 330-714, Republic of Korea; (V.S.); (S.S.)
- Smart Animal Bio Institute, Dankook University, Cheonan 330-714, Republic of Korea
| | - Shanmugam Sureshkumar
- Department of Animal Resource and Science, Dankook University, Cheonan 330-714, Republic of Korea; (V.S.); (S.S.)
- Smart Animal Bio Institute, Dankook University, Cheonan 330-714, Republic of Korea
| | - In Ho Kim
- Department of Animal Resource and Science, Dankook University, Cheonan 330-714, Republic of Korea; (V.S.); (S.S.)
- Smart Animal Bio Institute, Dankook University, Cheonan 330-714, Republic of Korea
| |
Collapse
|
8
|
Rostoll Cangiano L, Villot C, Amorin-Hegedus R, Malmuthuge N, Gruninger R, Guan LL, Steele M. Saccharomyces cerevisiae boulardii accelerates intestinal microbiota maturation and is correlated with increased secretory IgA production in neonatal dairy calves. Front Microbiol 2023; 14:1129250. [PMID: 37795296 PMCID: PMC10546063 DOI: 10.3389/fmicb.2023.1129250] [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: 12/21/2022] [Accepted: 08/30/2023] [Indexed: 10/06/2023] Open
Abstract
Neonatal calves have a limited capacity to initiate immune responses due to a relatively immature adaptive immune system, which renders them susceptible to many on-farm diseases. At birth, the mucosal surfaces of the intestine are rapidly colonized by microbes in a process that promotes mucosal immunity and primes the development of the adaptive immune system. In a companion study, our group demonstrated that supplementation of a live yeast probiotic, Saccharomyces cerevisiae boulardii (SCB) CNCM I-1079, to calves from birth to 1 week of age stimulates secretory IgA (sIgA) production in the intestine. The objective of the study was to evaluate how SCB supplementation impacts the intestinal microbiota of one-week-old male calves, and how changes in the bacterial community in the intestine relate to the increase in secretory IgA. A total of 20 calves were randomly allocated to one of two treatments at birth: Control (CON, n = 10) fed at 5 g/d of carrier with no live yeast; and SCB (n = 10) fed at 5 g of live SCB per day (10 × 109 CFU/d). Our study revealed that supplementing calves with SCB from birth to 1 week of age had its most marked effects in the ileum, increasing species richness and phylogenetic diversity in addition to expediting the transition to a more interconnected bacterial community. Furthermore, LEfSe analysis revealed that there were several differentially abundant taxa between treatments and that SCB increased the relative abundance the family Eubacteriaceae, Corynebacteriaceae, Eggerthellaceae, Bacillaceae, and Ruminococcaceae. Furthermore, network analysis suggests that SCB promoted a more stable bacterial community and appears to reduce colonization with Shigella. Lastly, we observed that the probiotic-driven increase in microbial diversity was highly correlated with the enhanced secretory IgA capacity of the ileum, suggesting that the calf's gut mucosal immune system relies on the development of a stable and highly diverse microbial community to provide the necessary cues to train and promote its proper function. In summary, this data shows that supplementation of SCB promoted establishment of a diverse and interconnected microbiota, prevented colonization of Escherichia Shigella and indicates a possible role in stimulating humoral mucosal immunity.
Collapse
Affiliation(s)
| | - Clothilde Villot
- Lallemand Animal Nutrition, Blagnac, France
- Lallemand Animal Nutrition, Milwaukee, WI, United States
| | | | - Nilusha Malmuthuge
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Robert Gruninger
- Lethbridge Research and Development Centre, Agriculture and Agri-Food Canada, Lethbridge, AB, Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Michael Steele
- Department of Animal Biosciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
9
|
Wilson A, Bogie B, Chaaban H, Burge K. The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease. Microorganisms 2023; 11:909. [PMID: 37110332 PMCID: PMC10144239 DOI: 10.3390/microorganisms11040909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The intestinal microbiome is frequently implicated in necrotizing enterocolitis (NEC) pathogenesis. While no particular organism has been associated with NEC development, a general reduction in bacterial diversity and increase in pathobiont abundance has been noted preceding disease onset. However, nearly all evaluations of the preterm infant microbiome focus exclusively on the bacterial constituents, completely ignoring any fungi, protozoa, archaea, and viruses present. The abundance, diversity, and function of these nonbacterial microbes within the preterm intestinal ecosystem are largely unknown. Here, we review findings on the role of fungi and viruses, including bacteriophages, in preterm intestinal development and neonatal intestinal inflammation, with potential roles in NEC pathogenesis yet to be determined. In addition, we highlight the importance of host and environmental influences, interkingdom interactions, and the role of human milk in shaping fungal and viral abundance, diversity, and function within the preterm intestinal ecosystem.
Collapse
Affiliation(s)
| | | | - Hala Chaaban
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathryn Burge
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
10
|
Wombwell E. Saccharomyces boulardii prophylaxis for targeted antibiotics and infectious indications to reduce healthcare facility-onset Clostridioides difficile infection. Microbes Infect 2023; 25:105041. [PMID: 36058513 DOI: 10.1016/j.micinf.2022.105041] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND Probiotic prophylaxis for Clostridioides difficile infection (CDI) is controversial stemming from deficits in strain and disease specificity considerations and concern for adverse effects. Here risk for healthcare facility-onset CDI (HO-CDI) dependent on concomitant antibiotic and infectious indication is assessed to identify opportunities for targeted prophylaxis. METHODS Retrospective matched-cohort study from January 2016 through March 2019. Patient-admissions administered high risk antibiotics were categorized by Saccharomyces boulardii administration and matched 1:1 to non-recipients. Unadjusted and adjusted HO-CDI risk estimated using Cox proportional hazards regression. RESULTS S. boulardii administration was associated with 48% risk reduction for HO-CDI compared to non-recipients (aHR 0.52, 95% CI: 0.31-0.87). Patient-admissions administered antibiotics and S. boulardii for a pneumonia indication exhibited a 57% reduction in risk for HO-CDI (aHR 0.43, 95% CI: 0.19-0.95). Administration of S. boulardii with ceftriaxone was associated with a 76% reduced risk of HO-CDI (aHR 0.24, 95% CI: 0.11-0.53) compared to ceftriaxone without S. boulardii, number needed to treat of 100. CONCLUSIONS S. boulardii administration is associated with a significant HO-CDI risk reduction for inpatients receiving antibiotics associated with CDI. Institutions interested in targeted use of S. boulardii to limit potential adverse effects may consider prophylaxis for inpatients with pneumonia or receiving ceftriaxone.
Collapse
Affiliation(s)
- Eric Wombwell
- Division of Pharmacy Practice and Administration, University of Missouri-Kansas City School of Pharmacy, Kansas City, Missouri, USA; Department of Pharmacy, Centerpoint Medical Center, Independence, Missouri, USA.
| |
Collapse
|
11
|
Zhang D, Liu H, Wang S, Liu Y, Ji H. Wheat bran fermented by Lactobacillus regulated the bacteria-fungi composition and reduced fecal heavy metals concentrations in growing pigs. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 858:159828. [PMID: 36368386 DOI: 10.1016/j.scitotenv.2022.159828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
Lactobacillus fermentation can increase the value of wheat bran, but the benefits of fermented wheat bran for pig production are poorly understood. We evaluated the phenolic acid content of wheat bran fermented with Lactobacillus. The bacterial and fungal compositions, short-chain fatty acids, and heavy metals concentrations in the feces of growing pigs were determined, and the correlations between the bacterial and fungal compositions and short-chain fatty acid and heavy metals concentrations were also assessed. The concentrations of phenolic acids (caffeic acid, catechinic acid, and gallic acid) were higher in fermented bran than in control wheat bran. The diversity of feces bacterial species was significantly higher, whereas the diversity of fungi was lower in fermented wheat bran treatment than those in the control group, and pigs consuming fermented and control wheat bran with different bacterial and fungal compositions had different growth rates. The abundance of genera in fungi that were less abundant in the fermented group samples than in the control samples (including Wallemia, Trichosporon, Candida, Aspergillus, and unclassified_f__Microascaceae) was positively correlated with heavy metals concentrations in pig feces, and the abundances of these fungi were negatively correlated with caffeic acid, catechinic acid, and gallic acid concentrations. Metagenomic function predictions indicated that larger amounts of secondary metabolites were synthesized in the fermented group than in the control group. The results provide new insights into the roles of bacterial-fungal interactions in the growth and decreasing environmental pollution of pigs consuming fermented wheat bran.
Collapse
Affiliation(s)
- Dongyan Zhang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Hui Liu
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Sixin Wang
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Yajuan Liu
- Mountainous Area Research Institute of Hebei Province, Hebei Agricultural University, China
| | - Haifeng Ji
- Institute of Animal Science and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China.
| |
Collapse
|
12
|
Lacotte PA, Simons A, Bouttier S, Malet-Villemagne J, Nicolas V, Janoir C. Inhibition of In Vitro Clostridioides difficile Biofilm Formation by the Probiotic Yeast Saccharomyces boulardii CNCM I-745 through Modification of the Extracellular Matrix Composition. Microorganisms 2022; 10:microorganisms10061082. [PMID: 35744599 PMCID: PMC9227484 DOI: 10.3390/microorganisms10061082] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/21/2022] [Accepted: 05/21/2022] [Indexed: 12/14/2022] Open
Abstract
Clostridioides difficile is responsible for post-antibiotic diarrhea and most of the pseudomembranous colitis cases. Multiple recurrences, one of the major challenges faced in C. difficile infection (CDI) management, can be considered as chronic infections, and the role of biofilm formation in CDI recurrences is now widely considered. Therefore, we explored if the probiotic yeast Saccharomyces boulardii CNCM I-745 could impact the in vitro formation of C. difficile biofilm. Biomass staining and viable bacterial cell quantification showed that live S. boulardii exerts an antagonistic effect on the biofilm formation for the three C. difficile strains tested. Confocal laser scanning microscopy observation revealed a weakening and an average thickness reduction of the biofilm structure when C. difficile is co-incubated with S. boulardii, compared to the single-species bacterial biofilm structure. These effects, that were not detected with another genetically close yeast, S. cerevisiae, seemed to require direct contact between the probiotic yeast and the bacterium. Quantification of the extrapolymeric matrix components, as well as results obtained after DNase treatment, revealed a significant decrease of eDNA, an essential structural component of the C. difficile biofilm matrix, in the dual-species biofilm. This modification could explain the reduced cohesion and robustness of C. difficile biofilms formed in the presence of S. boulardii CNCM I-745 and be involved in S. boulardii clinical preventive effect against CDI recurrences.
Collapse
Affiliation(s)
- Pierre-Alexandre Lacotte
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, 92290 Châtenay-Malabry, France; (P.-A.L.); (A.S.); (S.B.); (J.M.-V.)
| | - Alexis Simons
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, 92290 Châtenay-Malabry, France; (P.-A.L.); (A.S.); (S.B.); (J.M.-V.)
- Laboratoire Eau, Environnement et Systèmes Urbains (Leesu), Université Paris-Est Créteil, École des Ponts ParisTech, 94010 Créteil, France
| | - Sylvie Bouttier
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, 92290 Châtenay-Malabry, France; (P.-A.L.); (A.S.); (S.B.); (J.M.-V.)
| | - Jeanne Malet-Villemagne
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, 92290 Châtenay-Malabry, France; (P.-A.L.); (A.S.); (S.B.); (J.M.-V.)
| | - Valérie Nicolas
- Ingénierie et Plateformes au Service de l’Innovation (IPSIT), UMS IPSIT Université Paris-Saclay-US 31 INSERM-UAR 3679 CNRS, Plateforme d’Imagerie Cellulaire MIPSIT, 92290 Châtenay-Malabry, France;
| | - Claire Janoir
- INRAE, Université Paris-Saclay, AgroParisTech, Micalis Institute, 92290 Châtenay-Malabry, France; (P.-A.L.); (A.S.); (S.B.); (J.M.-V.)
- Correspondence:
| |
Collapse
|
13
|
Abid R, Waseem H, Ali J, Ghazanfar S, Muhammad Ali G, Elasbali AM, Alharethi SH. Probiotic Yeast Saccharomyces: Back to Nature to Improve Human Health. J Fungi (Basel) 2022; 8:jof8050444. [PMID: 35628700 PMCID: PMC9147304 DOI: 10.3390/jof8050444] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 02/02/2023] Open
Abstract
Saccharomyces cerevisiae var. boulardii is best known for its treatment efficacy against different gastrointestinal diseases. This probiotic yeast can significantly protect the normal microbiota of the human gut and inhibit the pathogenicity of different diarrheal infections. Several clinical investigations have declared S. cerevisiae var. boulardii a biotherapeutic agent due to its antibacterial, antiviral, anti-carcinogenic, antioxidant, anti-inflammatory and immune-modulatory properties. Oral or intramuscular administration of S. cerevisiae var. boulardii can remarkably induce health-promoting effects in the host body. Different intrinsic and extrinsic factors are responsible for its efficacy against acute and chronic gut-associated diseases. This review will discuss the clinical and beneficial effects of S. cerevisiae var. boulardii in the treatment and prevention of different metabolic diseases and highlight some of its health-promising properties. This review article will provide fundamental insights for new avenues in the fields of biotherapeutics, antimicrobial resistance and one health.
Collapse
Affiliation(s)
- Rameesha Abid
- Department of Biotechnology, University of Sialkot, Sialkot 51310, Pakistan;
- National Agriculture Research Center, National Institute of Genomics and Agriculture Biotechnology (NIGAB), Islamabad 44100, Pakistan;
- Correspondence: (A.M.E.); (R.A.)
| | - Hassan Waseem
- Department of Biological Sciences, Muslim Youth University, Islamabad 44100, Pakistan;
| | - Jafar Ali
- Department of Biotechnology, University of Sialkot, Sialkot 51310, Pakistan;
- Department of Biological Sciences, Muslim Youth University, Islamabad 44100, Pakistan;
| | - Shakira Ghazanfar
- National Agriculture Research Center, National Institute of Genomics and Agriculture Biotechnology (NIGAB), Islamabad 44100, Pakistan;
| | - Ghulam Muhammad Ali
- Pakistan Agricultural Research Council (PARC) 20, Ataturk Avenue, G-5/1, Islamabad 44000, Pakistan;
| | - Abdelbaset Mohamed Elasbali
- Department of Clinical Laboratory Science, College of Applied Sciences-Qurayyat, Jouf University, Al-Jouf P.O. Box 2014, Saudi Arabia
- Correspondence: (A.M.E.); (R.A.)
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran 66262, Saudi Arabia;
| |
Collapse
|
14
|
Wang H, Li J, Wu G, Zhang F, Yin J, He Y. The effect of intrinsic factors and mechanisms in shaping human gut microbiota. MEDICINE IN MICROECOLOGY 2022. [DOI: 10.1016/j.medmic.2022.100054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
15
|
Dissection and enhancement of prebiotic properties of yeast cell wall oligosaccharides through metabolic engineering. Biomaterials 2022; 282:121379. [DOI: 10.1016/j.biomaterials.2022.121379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/09/2021] [Accepted: 01/16/2022] [Indexed: 02/06/2023]
|
16
|
Gaisawat MB, Lopez-Escalera S, MacPherson CW, Iskandar MM, Tompkins TA, Kubow S. Probiotics Exhibit Strain-Specific Protective Effects in T84 Cells Challenged With Clostridioides difficile-Infected Fecal Water. Front Microbiol 2022; 12:698638. [PMID: 35154018 PMCID: PMC8826048 DOI: 10.3389/fmicb.2021.698638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
Clostridioides difficile infection (CDI) is frequently associated with intestinal injury and mucosal barrier dysfunction, leading to an inflammatory response involving neutrophil localization and upregulation of pro-inflammatory cytokines. The severity of clinical manifestations is associated with the extent of the immune response, which requires mitigation for better clinical management. Probiotics could play a protective role in this disorder due to their immunomodulatory ability in gastrointestinal disorders. We assessed five single-strain and three multi-strain probiotics for their ability to modulate CDI fecal water (FW)-induced effects on T84 cells. The CDI-FW significantly (p < 0.05) decreased T84 cell viability. The CDI-FW-exposed cells also exhibited increased pro-inflammatory cytokine production as characterized by interleukin (IL)-8, C-X-C motif chemokine 5, macrophage inhibitory factor (MIF), IL-32, and tumor necrosis factor (TNF) ligand superfamily member 8. Probiotics were associated with strain-specific attenuation of the CDI-FW mediated effects, whereby Saccharomyces boulardii CNCM I-1079 and Lacticaseibacillus rhamnosus R0011 were most effective in reducing pro-inflammatory cytokine production and in increasing T84 cell viability. ProtecFlor™, Lactobacillus helveticus R0052, and Bifidobacterium longum R0175 showed moderate effectiveness, and L. rhamnosus GG R0343 along with the two other multi-strain combinations were the least effective. Overall, the findings showed that probiotic strains possess the capability to modulate the CDI-mediated inflammatory response in the gut lumen.
Collapse
Affiliation(s)
| | | | - Chad W MacPherson
- Rosell Institute for Microbiome and Probiotics, Montréal, QC, Canada
| | | | - Thomas A Tompkins
- Rosell Institute for Microbiome and Probiotics, Montréal, QC, Canada
| | - Stan Kubow
- School of Human Nutrition, McGill University, Montréal, QC, Canada
| |
Collapse
|
17
|
Coker OO. Non-bacteria microbiome (virus, fungi, and archaea) in gastrointestinal cancer. J Gastroenterol Hepatol 2022; 37:256-262. [PMID: 34825404 DOI: 10.1111/jgh.15738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022]
Abstract
The gastrointestinal tract houses millions of microbes collectively referred to as the gut microbiome. The gut microbes comprise of bacteria, viruses, fungi, archaea, and microscopic eukaryotes, which co-evolved or colonize the gut forming complex symbiotic and mutualistic relationships. A state of homeostasis is required between host and gut microbiome relationship to maintain several host beneficial processes. Alterations in the taxonomic and functional composition of the gut microbes are associated with several human diseases including gastrointestinal cancers. Owed to their overwhelming abundance and ease of characterization, several studies focus on the role of bacteria in gastrointestinal cancers. There is however growing evidence that non-bacteria gut microbes are associated with the pathogenesis of gastrointestinal cancers. This review details the association of non-bacteria gut microbes including fungi, viruses, and archaea and their potential manipulation in the prevention and treatment of human gastrointestinal cancers.
Collapse
Affiliation(s)
- Olabisi Oluwabukola Coker
- Institute of Digestive Disease and The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
18
|
Effects of Saccharomyces cerevisiae var. boulardii CNCM I-1079 on performance, colostrum and milk composition, and litter performance of mixed-parity sows in a tropical humid climate. Trop Anim Health Prod 2022; 54:41. [PMID: 35013815 DOI: 10.1007/s11250-022-03051-8] [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: 07/07/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
This study aimed to evaluate the effects of Saccharomyces cerevisiae var. boulardii (SB) for sows on their productive performance, colostrum and milk composition, and litter performance, in tropical humid climatic conditions. A total of 105 sows (Topigs Norsvin®) were allotted to a 5 × 3 completely randomized factorial design, with five diets (control diet; SBGL4 and SBGL8: 0.04 and 0.08% SB supplementation from the 90th day of gestation until 24th day of lactation; SBL4 and SBL8: 0.04 and 0.08% SB supplementation during lactation) and three parity order groups (PO: 1st and 2nd; 3rd and 4th; 5th to 7th), considering each sow and their litter as experimental unit. Sows above the 5th PO that fed control diet had a lower daily milk production (DMP), number of weaned piglets (NWP), and daily weight gain of litter (DWGL) than sows from 1st to 4th PO that fed the same diet (P < 0.05). Dietary supply of SBGL4 and SBGL8 to older sows provided a higher DMP, NWP, and DWGL when compared to sows of same PO that fed the control diet (P < 0.05). Dietary supply of SBGL4, SBGL8, SBL4, and SBL8 provided a higher dry matter and protein contents in sows' milk of 1st and 2nd PO when compared to sows from same PO that fed control diet (P < 0.05). Dietary supply of SB enhances milk yield of older sows and their litter performance, as well as the dry matter and protein content of younger sows' milk in tropical humid climatic conditions.
Collapse
|
19
|
Diarrheal disease and gut microbiome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:149-177. [DOI: 10.1016/bs.pmbts.2022.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
20
|
Guli M, Winarsih S, Barlianto W, Illiandri O, Sumarno SP. Mechanism of Lactobacillus reuteri Probiotic in Increasing Intestinal Mucosal Immune System. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Probiotics are defined as live microorganisms which, when consumed in adequate quantities as food ingredients, provide health benefits to the host. Lactobacillus, Bifidobacterium, and Saccharomyces, are three probiotics that are intensively used as probiotics in humans and animals. Probiotics have beneficial effects on health when given adequate amounts. The concept of probiotics on human health, namely modulating the gut microbiota and its effect on the host. Probiotics play an important role in maintaining intestinal integrity through a number of different interactions, including changes in cytokine expression in the mucosa. Probiotics compete with intestinal pathogens for mucosal receptors, thereby increasing interepithelial resistance. Probiotics such as Lactobacillus casei sp GG strain was used as a prophylaxis that could increase the expression of epithelial mucin, thereby reducing the translocation of pathogenic bacteria. Abnormal local immune response is characterized by decreased secretion of IgA, thus allowing enterocyte attachment and local translocation of bacterial antigens, which are the main stimulation of pathological events. Colonic stasis can promote the growth of pathogenic bacteria which allows malignant porin bacterial strains to thrive. The gut microbiota has a major influence on human health. The microbial population has an important role in the host, such as the metabolic activity of probiotics producing energy and nutrient absorption, developing the host immune system, and preventing colonization and infection of pathogens. Lactobacillus reuteri is a hetero-fermentative bacterium that lives in the digestive tract of humans. L. reuteri has been used to treat infant necrotizing pseudomembrane. In this paper, the mechanism of L reuteri to increase host immunological response will be reviewed.
Collapse
|
21
|
Goodman C, Keating G, Georgousopoulou E, Hespe C, Levett K. Probiotics for the prevention of antibiotic-associated diarrhoea: a systematic review and meta-analysis. BMJ Open 2021; 11:e043054. [PMID: 34385227 PMCID: PMC8362734 DOI: 10.1136/bmjopen-2020-043054] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE To evaluate existing evidence for the use of probiotics in preventing antibiotic-associated diarrhoea (AAD) in adults. DESIGN Systematic review and meta-analysis of randomised controlled trials (RCTs). DATA SOURCES We performed a literature search of the electronic databases CINAHL Plus, EMBASE, MEDLINE (Ovid) and Web of Science from database inception to May 2021 as well as hand searching of trial registries and reference lists of related reviews. STUDY SELECTION Two reviewers identified whether RCTs met the following inclusion criteria: adult population to whom antibiotics were administered; a probiotic intervention; a placebo, alternative dose, alternative probiotic strain or no treatment control; and incidence of AAD as the outcome. DATA EXTRACTION AND SYNTHESIS Two reviewers independently collected data and assessed risk of bias using preconstructed data extraction forms. We used a random effects model for all analyses. Subgroup analyses were performed to evaluate species-specific and dose-specific response. RESULTS Forty-two studies (11,305 participants) were included in this review. The pooled analysis suggests that co-administration of probiotics with antibiotics reduces the risk of AAD in adults by 37% (risk ratio (RR)=0.63 (95% CI 0.54 to 0.73), p<0.00001). The overall quality of the evidence using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) criteria was found to be moderate. In subgroup analyses, high dose compared with low dose of the same probiotic demonstrated a positive protective effect (RR 0.54 (95% CI 0.38 to 0.76), p<0.01), and only certain species, mainly of the lactobacillus and bifidobacteria genera, were found to be effective. Studies with a low baseline AAD risk did not show any difference in risk but studies with moderate or high baseline AAD risk demonstrated a significant risk reduction. CONCLUSIONS Probiotics are effective for preventing AAD. Secondary analyses of higher dosages and certain species have shown increased effectiveness. Our results may not be applicable in clinical scenarios of lower baseline AAD risk.
Collapse
Affiliation(s)
- Clare Goodman
- National School of Medicine, The University of Notre Dame Australia, Sydney, New South Wales, Australia
| | - Georgia Keating
- Department of Pharmacy, Alice Springs Hospital, Alice Springs, Northern Territory, Australia
| | - Ekavi Georgousopoulou
- National School of Medicine, The University of Notre Dame Australia, Sydney, New South Wales, Australia
| | - Charlotte Hespe
- National School of Medicine, The University of Notre Dame Australia, Sydney, New South Wales, Australia
| | - Kate Levett
- National School of Medicine, The University of Notre Dame Australia, Sydney, New South Wales, Australia
| |
Collapse
|
22
|
Wu Y, Wang L, Luo R, Chen H, Nie C, Niu J, Chen C, Xu Y, Li X, Zhang W. Effect of a Multispecies Probiotic Mixture on the Growth and Incidence of Diarrhea, Immune Function, and Fecal Microbiota of Pre-weaning Dairy Calves. Front Microbiol 2021; 12:681014. [PMID: 34335503 PMCID: PMC8318002 DOI: 10.3389/fmicb.2021.681014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/20/2021] [Indexed: 12/29/2022] Open
Abstract
The effects of different doses of a multispecies probiotic (MSP) mixture on growth performance, the incidence of diarrhea rate and immune function, and fecal microbial diversity and structure were evaluated in pre-weaning Holstein dairy calves at WK2, WK4, WK6, and WK8. Forty Chinese Holstein female newborn calves were randomly assigned to four treatments with 10 calves in each group, C (control group), T1 (0.5 g MSP/calf/day, T2 (1 g MSP/calf/day), and T3 (2 g MSP/calf/day) groups. The experimental period was 56 days. Feed intake and health scoring were recorded every day until the end of the experiment. Fecal contents and blood samples were sampled at WK2, WK4, WK6, and WK8. Growth performance, incidence of diarrhea, and total serum concentrations (IgA, IgG, and IgM) were analyzed. Bacterial 16S rRNA and fungal ITS genes were high-throughput sequenced for fecal microbiota. The relationships among the populations of the principal fecal microbiota at WK2 and the growth performance or serum immunoglobulin concentrations were analyzed using Pearson's rank correlation coefficients. The MSP supplementation reduced the incidence of diarrhea in the first 4 weeks of life, and serum IgA, IgG, and IgM concentrations increased between WK2 and WK8 in the T3 group. There was an increase in growth performance and reduction in the incidence of diarrhea until WK4 after birth in T3 group, compared with the control, T1, and T2 groups. The results of fecal microbiota analysis showed that Firmicutes and Bacteroides were the predominant phyla, with Blautia, Ruminococcaceae_UCG-005, norank_f__Muribaculaceae, Bacteroides, Subdoligranulum, and Bifidobacterium being the dominant genera in calf feces. Aspergillus, Thermomyces, and Saccharomyces were the predominant fungal phyla. Compared with the control, in T1 and T2 groups, the MSP supplementation reduced the relative abundance of Bacteroidetes and increased the relative abundance of Bifidobacterium, Lactobacillus, Collinsella, and Saccharomyces at WK2 in group T3. Thus, the fecal microbial composition and diversity was significantly affected by the MSP mixture during the first 2 weeks of the calves' life. MSP mixtures reduced the incidence of diarrhea in pre-weaning calves (during the first 4 weeks of life). There was a significant improvement in growth performance, reduction in calf diarrhea, balance in the fecal microbiota, and an overall improvement in serum immunity, compared with the control group. We, therefore, recommend adding 2 g/day of multispecies probiotic mixture supplementation in diets of dairy calves during their first 4 weeks of life before weaning.
Collapse
Affiliation(s)
- Yanyan Wu
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Lili Wang
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Ruiqing Luo
- Xinjiang Tianshan Junken Animal Husbandry Co., Ltd., Shihezi, China
| | - Hongli Chen
- Xinjiang Tianshan Junken Animal Husbandry Co., Ltd., Shihezi, China
| | - Cunxi Nie
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Junli Niu
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Cheng Chen
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| | - Yongping Xu
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Xiaoyu Li
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Wenjun Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, China
| |
Collapse
|
23
|
Ansari F, Alian Samakkhah S, Bahadori A, Jafari SM, Ziaee M, Khodayari MT, Pourjafar H. Health-promoting properties of Saccharomyces cerevisiae var. boulardii as a probiotic; characteristics, isolation, and applications in dairy products. Crit Rev Food Sci Nutr 2021; 63:457-485. [PMID: 34254862 DOI: 10.1080/10408398.2021.1949577] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Saccharomyces cerevisiae var. boulardii (S. boulardii) has been isolated from lychee (Litchi chinensis), mangosteen fruit, kombucha, and dairy products like kefir. Dairy products containing S. boulardii have been revealed to possess potential probiotic activities owing to their ability to produce organic acids, essential enzymes, vitamins, and other important metabolites such as vanillic acid, phenyl ethyl alcohol, and erythromycin. S. boulardii has a wide spectrum of anti-carcinogenic, antibacterial antiviral, and antioxidant activity, and is known to reduce serum cholesterol levels. However, this yeast has mainly been prescribed for prophylaxis treatment of gastrointestinal infectious diseases, and stimulating the immune system in a number of commercially available products. The present comprehensive review article reviews the properties of S. boulardii related to their use in fermented dairy foods as a probiotic microorganism or starter culture. Technical aspects regarding the integration of this yeast into the dairy foods matrix its health advantages, therapeutic functions, microencapsulation, and viability in harsh conditions, and safety aspects are highlighted.
Collapse
Affiliation(s)
- Fereshteh Ansari
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran.,Research Center for Evidence-Based Medicine, Health Management and Safety Promotion Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Iranian EBM Centre: A Joanna Briggs Institute Affiliated Group
| | - Shohre Alian Samakkhah
- Department of Food Hygiene and Quality Control, Faculty of Veterinary of Medicine, Amol University of Special Modern Technology, Amol, Iran
| | - Ali Bahadori
- Department of Medical Microbiology, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Seyedeh Maedeh Jafari
- Department of Comparative Bioscience, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| | - Mojtaba Ziaee
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | | | - Hadi Pourjafar
- Alborz University of Medical Sciences, Dietary Supplements and Probiotic Research Center, Karaj, Iran.,Department of Food Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
24
|
Chen K, Zhu Y, Zhang Y, Hamza T, Yu H, Saint Fleur A, Galen J, Yang Z, Feng H. A probiotic yeast-based immunotherapy against Clostridioides difficile infection. Sci Transl Med 2021; 12:12/567/eaax4905. [PMID: 33115949 DOI: 10.1126/scitranslmed.aax4905] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 02/12/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Antibiotic-resistant Clostridioides difficile is an anaerobic Gram-positive bacterium that colonizes the colon and is responsible for more than 29,000 deaths in the United States each year. Hence, C. difficile infection (CDI) poses an urgent threat to public health. Antibody-mediated neutralization of TcdA and TcdB toxins, the major virulence factors of CDI, represents an effective strategy to combat the disease without invoking antibiotic resistance. However, current antitoxin approaches are mostly based on parenteral infusion of monoclonal antibodies that are costly, narrow spectrum, and not optimized against the intestinal disease. Here, we engineered probiotic Saccharomyces boulardii to constitutively secrete a single tetra-specific antibody that potently and broadly neutralized both toxins and demonstrated protection against primary and recurrent CDI in both prophylactic and therapeutic mouse models of disease. This yeast immunotherapy is orally administered, can be used concurrently with antibiotics, and may have potential as a prophylactic against CDI risk and as a therapeutic for patients with CDI.
Collapse
Affiliation(s)
- Kevin Chen
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Yixuan Zhu
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yongrong Zhang
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Therwa Hamza
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Hua Yu
- FZata Inc., Halethorpe, MD 21227, USA
| | - Ashley Saint Fleur
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - James Galen
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | - Hanping Feng
- Department of Microbial Pathogenesis, University of Maryland School of Dentistry, Baltimore, MD 21201, USA.
| |
Collapse
|
25
|
Staniszewski A, Kordowska-Wiater M. Probiotic and Potentially Probiotic Yeasts-Characteristics and Food Application. Foods 2021; 10:1306. [PMID: 34200217 PMCID: PMC8228341 DOI: 10.3390/foods10061306] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Probiotics are live microorganisms which when administered in adequate amounts confer a health benefit on the host. Besides the well-known and tested lactic acid bacteria, yeasts may also be probiotics. The subject of probiotic and potentially probiotic yeasts has been developing and arising potential for new probiotic products with novel properties, which are not offered by bacteria-based probiotics available on the current market. The paper reviews the first probiotic yeast Saccharomyces cerevisiae var. boulardii, its characteristics, pro-healthy activities and application in functional food production. This species offers such abilities as improving digestion of certain food ingredients, antimicrobial activities and even therapeutic properties. Besides Saccharomyces cerevisiae var. boulardii, on this background, novel yeasts with potentially probiotic features are presented. They have been intensively investigated for the last decade and some species have been observed to possess probiotic characteristics and abilities. There are yeasts from the genera Debaryomyces, Hanseniaspora, Pichia, Meyerozyma, Torulaspora, etc. isolated from food and environmental habitats. These potentially probiotic yeasts can be used for production of various fermented foods, enhancing its nutritional and sensory properties. Because of the intensively developing research on probiotic yeasts in the coming years, we can expect many discoveries and possibly even evolution in the segment of probiotics available on the market.
Collapse
Affiliation(s)
| | - Monika Kordowska-Wiater
- Department of Biotechnology, Microbiology and Human Nutrition, University of Life Sciences in Lublin, Skromna 8, 20-704 Lublin, Poland;
| |
Collapse
|
26
|
Sommermeyer H, Pituch HM, Wultanska D, Wojtyla-Buciora P, Piatek J, Bernatek M. Inhibition of Quinolone- and Multi-Drug-Resistant Clostridioides Difficile Strains by Multi Strain Synbiotics-An Option for Diarrhea Management in Nursing Facilities. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:5871. [PMID: 34070727 PMCID: PMC8198539 DOI: 10.3390/ijerph18115871] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/18/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Diarrhea is a common problem in nursing homes. A survey among nursing facilities in Poland was used to characterize diarrhea outbreaks, the burden caused for residents and caregivers and the employed measures. Survey results confirmed that diarrhea is a common problem in nursing homes and in most cases affects groups of residents. The related burden is high or very high for 27% of residents and 40% of caregivers. In 80% of nursing facilities pro or synbiotics are part of the measures used to manage diarrhea. Administration of these kinds of products has been suggested for the management of diarrhea, especially in cases caused by Clostridioides (C.) difficile. C. difficile is one of many potential causes for diarrhea, but is of particular concern for nursing homes because it is responsible for a large proportion of diarrhea outbreaks and is often caused by multi-drug resistant strains. In vitro inhibition of a quinolone-resistant and a multi-drug resistant C. difficile strain was used to evaluate the growth inhibitory effects of commonly used products containing probiotic microorganisms. Growth of both strains was best inhibited by multi-strain synbiotic preparations. These findings suggest that multi-strain synbiotics can be considered as an interventional option for diarrhea caused by C. difficile.
Collapse
Affiliation(s)
- Henning Sommermeyer
- Department of Health Sciences, Calisia University-Kalisz, Nowy Swiat 4, 62-800 Kalisz, Poland; (H.S.); (P.W.-B.); (M.B.)
| | - Hanna M. Pituch
- Department of Medical Microbiology, Medical University of Warsaw, ul. Żwirki i Wigury 61, 02-091 Warsaw, Poland; (H.M.P.); (D.W.)
| | - Dorota Wultanska
- Department of Medical Microbiology, Medical University of Warsaw, ul. Żwirki i Wigury 61, 02-091 Warsaw, Poland; (H.M.P.); (D.W.)
| | - Paulina Wojtyla-Buciora
- Department of Health Sciences, Calisia University-Kalisz, Nowy Swiat 4, 62-800 Kalisz, Poland; (H.S.); (P.W.-B.); (M.B.)
| | - Jacek Piatek
- Department of Health Sciences, Calisia University-Kalisz, Nowy Swiat 4, 62-800 Kalisz, Poland; (H.S.); (P.W.-B.); (M.B.)
| | - Malgorzata Bernatek
- Department of Health Sciences, Calisia University-Kalisz, Nowy Swiat 4, 62-800 Kalisz, Poland; (H.S.); (P.W.-B.); (M.B.)
| |
Collapse
|
27
|
Yang Z, Wang Y, He T, Ziema Bumbie G, Wu L, Sun Z, Sun W, Tang Z. Effects of Dietary Yucca Schidigera Extract and Oral Candida utilis on Growth Performance and Intestinal Health of Weaned Piglets. Front Nutr 2021; 8:685540. [PMID: 34124128 PMCID: PMC8187599 DOI: 10.3389/fnut.2021.685540] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/22/2021] [Indexed: 12/21/2022] Open
Abstract
Weaning piglets experienced the transformation from breast milk to solid feed and present the proliferation of pathogens, the presence of diarrhea, poor growth performance and even death. Plant extracts and probiotics have certain potential in improving animal growth performance, antioxidant capacity and immune function. The purpose of this study was to explore the effects of dietary yucca schidigera extract (YSE) and oral Candida utilis (CU) on growth performance and intestinal health weaned piglets. According to a 2 × 2 factorial design with the main factors being CU (orally administered 1 mL of 0.85% saline with or without CU; fed basal diet with or without 120 mg/kg YSE), forty 28 d healthy weaned piglets were randomly allocated into four groups of 10 barrows each: (1) piglets fed basal diet and orally administered 1 mL of 0.85% saline (CON); (2) piglets fed basal diet and orally administered 1 mL 1 × 109 cfu/mL C. utilis in 0.85% saline (CU); (3) piglets fed the basal diet containing YSE (120 mg/kg) and orally administered 1 mL of 0.85% saline (YSE); (4) Piglets fed the basal diet containing 120 mg/kg YSE and 1 mL 1 × 109 cfu/mL C. utilis in 0.85% saline (YSE+CU). This study lasted 28 days and evaluated the effects of dietary YSE and oral CU on growth performance, immunity, antioxidant function, ileal morphology, and intestinal microflora in weaned piglets. Dietary YSE increased ADG, the spleen and lymph node indexes, serum GLU, BUN, T-SOD, T-AOC, CAT concentrations, ileal villus height and villus height/crypt depth, jejunal occludin, and β-definsin-2 concentrations and ileal occludin concentration in weaned piglets (P < 0.05); decreased the diarrhea rate and mortality, rectal pH and urine pH, the BUN and MDA concentrations, crypt depth (P < 0.05); improved the diversity of cecal microflora. Orally CU increased ADG, and ADFI, the T-SOD, T-AOC, and CAT activity, ileal villus height, villus height/crypt depth, jejunum occludin, and β-definsin-2 concentrations (P < 0.05); reduced the diarrhea rate and mortality, urine pH, the BUN and MDA concentrations, crypt depth (P < 0.05); improved the diversity of cecal microflora. Dietary YSE and orally CU increased the T-SOD, T-AOC, and CAT activity, villus height/crypt depth, jejunal occludin concentration; reduced the diarrhea rate of weaned piglets by 28%, gastric pH, ileal pH, cecal pH and urine pH, MDA, crypt depth; improved the diversity of cecal microflora. YSE and CU could improve the growth performance, reduce the diarrhea rate, improve intestinal health, and increase the diversity and abundance of cecal microflora in weaned piglets and expected to be used as antibiotics alternative feed additives in the production of weaned piglets.
Collapse
Affiliation(s)
- Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yao Wang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Gifty Ziema Bumbie
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Liuting Wu
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhihong Sun
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Weizhong Sun
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhiru Tang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
28
|
Raheem A, Liang L, Zhang G, Cui S. Modulatory Effects of Probiotics During Pathogenic Infections With Emphasis on Immune Regulation. Front Immunol 2021; 12:616713. [PMID: 33897683 PMCID: PMC8060567 DOI: 10.3389/fimmu.2021.616713] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In order to inhibit pathogenic complications and to enhance animal and poultry growth, antibiotics have been extensively used for many years. Antibiotics applications not only affect target pathogens but also intestinal beneficially microbes, inducing long-lasting changes in intestinal microbiota associated with diseases. The application of antibiotics also has many other side effects like, intestinal barrier dysfunction, antibiotics residues in foodstuffs, nephropathy, allergy, bone marrow toxicity, mutagenicity, reproductive disorders, hepatotoxicity carcinogenicity, and antibiotic-resistant bacteria, which greatly compromise the efficacy of antibiotics. Thus, the development of new antibiotics is necessary, while the search for antibiotic alternatives continues. Probiotics are considered the ideal antibiotic substitute; in recent years, probiotic research concerning their application during pathogenic infections in humans, aquaculture, poultry, and livestock industry, with emphasis on modulating the immune system of the host, has been attracting considerable interest. Hence, the adverse effects of antibiotics and remedial effects of probiotics during infectious diseases have become central points of focus among researchers. Probiotics are live microorganisms, and when given in adequate quantities, confer good health effects to the host through different mechanisms. Among them, the regulation of host immune response during pathogenic infections is one of the most important mechanisms. A number of studies have investigated different aspects of probiotics. In this review, we mainly summarize recent discoveries and discuss two important aspects: (1) the application of probiotics during pathogenic infections; and (2) their modulatory effects on the immune response of the host during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Abdul Raheem
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Lin Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Guangzhi Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Shangjin Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| |
Collapse
|
29
|
Guo J, Ren C, Han X, Huang W, You Y, Zhan J. Role of IgA in the early-life establishment of the gut microbiota and immunity: Implications for constructing a healthy start. Gut Microbes 2021; 13:1-21. [PMID: 33870860 PMCID: PMC8078773 DOI: 10.1080/19490976.2021.1908101] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Colonization and maturation of the gut microbiota (GM) during early life is a landmark event that fundamentally influences the (early) immunity and later-life health of various mammals. This is a delicate, systematic process that is biologically actively regulated by infants and their mothers, where (secretory) IgA, an important regulator of microbes found in breast milk and generated actively by infants, may play a key role. By binding to microbes, IgA can inhibit or enhance their colonization, influence their gene expression, and regulate immune responses. IgA dysfunction during early life is associated with disrupted GM maturation and various microbe-related diseases, such as necrotizing enterocolitis and diarrhea, which can also have a lasting effect on GM and host health. This review discusses the process of early GM maturation and its interaction with immunity and the role of IgA (focusing on milk secretory IgA) in regulating this process. The possible application of this knowledge in promoting normal GM maturation processes and immune education has also been highlighted.
Collapse
Affiliation(s)
- Jielong Guo
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Chenglong Ren
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Xue Han
- Peking University School of Basic Medical Science, Peking University Health Science Centre
| | - Weidong Huang
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Yilin You
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Jicheng Zhan
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| |
Collapse
|
30
|
Herrera G, Paredes-Sabja D, Patarroyo MA, Ramírez JD, Muñoz M. Updating changes in human gut microbial communities associated with Clostridioides difficile infection. Gut Microbes 2021; 13:1966277. [PMID: 34486488 PMCID: PMC8425690 DOI: 10.1080/19490976.2021.1966277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 02/04/2023] Open
Abstract
Clostridioides difficile is the causative agent of antibiotic-associated diarrhea, a worldwide public health problem. Different factors can promote the progression of C. difficile infection (CDI), mainly altered intestinal microbiota composition. Microbial species belonging to different domains (i.e., bacteria, archaea, eukaryotes, and even viruses) are synergistically and antagonistically associated with CDI. This review was aimed at updating changes regarding CDI-related human microbiota composition using recent data and an integral approach that included the different microorganism domains. The three domains of life contribute to intestinal microbiota homeostasis at different levels in which relationships among microorganisms could explain the wide range of clinical manifestations. A holistic understanding of intestinal ecosystem functioning will facilitate identifying new predictive factors for infection and developing better treatment and new diagnostic tools, thereby reducing this disease's morbidity and mortality.
Collapse
Affiliation(s)
- Giovanny Herrera
- Centro de Investigaciones en Microbiología y Biotecnología – UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad Del Rosario, Bogotá, Colombia
| | - Daniel Paredes-Sabja
- ANID – Millennium Science Initiative Program – Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá D.C. 111321, Colombia
- Health Sciences Division, Main Campus, Universidad Santo Tomás, Bogotá D.C. 110231, Colombia
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología – UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad Del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología – UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad Del Rosario, Bogotá, Colombia
- ANID – Millennium Science Initiative Program – Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| |
Collapse
|
31
|
Gil-Rodríguez AM, Garcia-Gutierrez E. Antimicrobial mechanisms and applications of yeasts. ADVANCES IN APPLIED MICROBIOLOGY 2020; 114:37-72. [PMID: 33934852 DOI: 10.1016/bs.aambs.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Yeasts and humans have had a close relationship for millenia. Yeast have been used for food production since the first human societies. Since then, alternative uses have been discovered. Nowadays, antibiotic resistance constitutes a pressing need worldwide. In order to overcome this threat, one of the most important strategies is the search for new antimicrobials in natural sources. Moreover, biopreservation based on natural sources has emerged as an alternative to more common chemical preservatives. Yeasts constitute an underexploited source of antagonistic activity against other microorganisms. Here, we compile a summary of the antagonistic activity of yeast origin against other yeast and other microorganisms, such as bacteria or parasites. We present the mechanisms of action used by yeasts to display these activities. We also provide applications of these antagonistic activities in food industry and agriculture, medicine and veterinary, where yeast promise to play a pivotal role in the near future.
Collapse
|
32
|
Kunyeit L, K A AA, Rao RP. Application of Probiotic Yeasts on Candida Species Associated Infection. J Fungi (Basel) 2020; 6:jof6040189. [PMID: 32992993 PMCID: PMC7711718 DOI: 10.3390/jof6040189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023] Open
Abstract
Superficial and life-threatening invasive Candida infections are a major clinical challenge in hospitalized and immuno-compromised patients. Emerging drug-resistance among Candida species is exacerbated by the limited availability of antifungals and their associated side-effects. In the current review, we discuss the application of probiotic yeasts as a potential alternative/ combination therapy against Candida infections. Preclinical studies have identified several probiotic yeasts that effectively inhibit virulence of Candida species, including Candida albicans, Candida tropicalis, Candida glabrata, Candida parapsilosis, Candida krusei and Candida auris. However, Saccharomyces cerevisiae var. boulardii is the only probiotic yeast commercially available. In addition, clinical studies have further confirmed the in vitro and in vivo activity of the probiotic yeasts against Candida species. Probiotics use a variety of protective mechanisms, including posing a physical barrier, the ability to aggregate pathogens and render them avirulent. Secreted metabolites such as short-chain fatty acids effectively inhibit the adhesion and morphological transition of Candida species. Overall, the probiotic yeasts could be a promising effective alternative or combination therapy for Candida infections. Additional studies would bolster the application of probiotic yeasts.
Collapse
Affiliation(s)
- Lohith Kunyeit
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Anu-Appaiah K A
- Department of Microbiology and Fermentation Technology, CSIR- Central Food Technological Research Institute (CFTRI), Mysuru 570020, India; (L.K.); (A.K.A.)
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Reeta P. Rao
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA
- Correspondence: ; Tel.: +1-508-831-5000
| |
Collapse
|
33
|
Villot C, Chen Y, Pedgerachny K, Chaucheyras-Durand F, Chevaux E, Skidmore A, Guan LL, Steele MA. Early supplementation of Saccharomyces cerevisiae boulardii CNCM I-1079 in newborn dairy calves increases IgA production in the intestine at 1 week of age. J Dairy Sci 2020; 103:8615-8628. [PMID: 32684462 DOI: 10.3168/jds.2020-18274] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 04/22/2020] [Indexed: 01/22/2023]
Abstract
The early development of immunity and microbiota in the gut of newborn calves can have life-long consequences. Gut microbiota and the intestinal barrier interplay after birth, establishing a homeostatic state whereby mucosal cells cohabit with microorganisms to develop a healthy gut. We hypothesized that postnatal codevelopment of gut immunity and microbiota could be influenced by early-life supplementation with live yeast. Starting from birth, calves either received a daily supplementation of Saccharomyces cerevisiae boulardii CNCM I-1079 (SCB, 10 × 109 cfu/d, n = 10) in the morning meal for 7 d or no supplementation (n = 10). Each animal received 2 adequate colostrum replacer meals at 2 and 12 h of life (expected total IgG fed = 300 g) before being fed milk replacer twice a day. Passive transfer of immunity (total protein, IgG, and IgA) through colostrum was evaluated and endogenous production of IgA was investigated by measuring IgA-producing plasma cells, IgA relative gene expression (PIGR and CD79A), and secretory IgA concentration in the gut. The concentration of targeted microbial groups was evaluated with quantitative PCR in the gut digesta collected at d 7 of life. Early SCB supplementation did not impair immunoglobulin absorption and all calves had successful passive transfer of immunity (serum IgG concentration >15 mg/mL at d 1 and d 7 of age). Although the expression of IgA relative gene expression (PIGR and CD79A) was not different, SCB calves had higher secretory IgA concentrations in the ileum (1.98 ± 0.12 mg/g of dry matter; DM) and colon (1.45 ± 0.12 mg/g of DM) digesta compared with control animals (1.18 and 0.59 ± 0.12 mg/g of DM, respectively). In addition, the number of IgA-producing plasma cells were greater in both ileum (2.55 ± 0.40 cells/mm2) and colon (3.03 ± 0.40 cells/mm2) tissues for SCB calves compared with control (respectively 1.00 ± 0.40 and 0.60 ± 0.42 cells/mm2). Endogenous IgA production in the gut of SCB calves was enhanced, which could make them less prone to pathogen intrusion. In addition, SCB calves had higher Lactobacillus and tended to have higher Faecalibacterium prausnitzii in the jejunum compared with control calves, which suggests that SCB supplementation during early-life gut colonization may have a positive effect in newborn calves. Direct SCB supplementation or the cross-talk between SCB and bacteria may be responsible for stimulating IgA production and may play a key role in shaping early colonization in the gut of newborn calves.
Collapse
Affiliation(s)
- C Villot
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada; Lallemand SAS, F-31702 Blagnac, France
| | - Y Chen
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada; Lethbridge Research Center, Agriculture and Agri-Food Canada, Lethbridge, AB T1J 4B1, Canada
| | - K Pedgerachny
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| | | | - E Chevaux
- Lallemand SAS, F-31702 Blagnac, France
| | - A Skidmore
- Lallemand Specialties Inc, Milwaukee, WI 53218
| | - L L Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada
| | - M A Steele
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton T6G 2P5, Canada; Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada.
| |
Collapse
|
34
|
Prado GKS, Torrinha KC, Cruz RE, Gonçalves ABB, Silva CAV, Oliveira FMS, Nunes AC, Gomes MA, Caliari MV. Weissella paramesenteroides WpK4 ameliorate the experimental amoebic colitis by increasing the expression of MUC-2 and the intestinal epithelial regeneration. J Appl Microbiol 2020; 129:1706-1719. [PMID: 32320114 DOI: 10.1111/jam.14671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 01/03/2023]
Abstract
AIMS This study evaluates the action of Weissella paramesenteroides WpK4 on amoebic colitis. METHODS AND RESULTS Weissella paramesenteroides WpK4 was administered in Entamoeba dispar infected and noninfected mice and clinical parameters were evaluated. Following 7 days, the caeca were collected for histopathology, morphometry and immunohistochemical staining of MUC-2, CDC-47 and IgA. The treatment reduced diarrhoea and the presence of blood in the faeces and diminished the area of necrosis, also causing weight gain. Also, the addition of this bacterium enhanced the expression of the mucin (MUC-2). The reduction in necrosis and increased CDC-47 expression indicates significant epithelial regeneration. The negative correlation between CDC-47 and the necrosis area reveals that the bacterium favoured the recovery of the necrotic regions and the positive correlation found between the expression of MUC-2 and CDC-47 indicates that the epithelial regeneration also supports the synthesis of MUC-2. CONCLUSIONS Weissella paramesenteroides WpK4 was able to increase the protection of the intestinal mucosa against experimental amoebic colitis through the increase of MUC-2 and epithelial regeneration. SIGNIFICANCE AND IMPACT OF THE STUDY Weissella paramesenteroides WpK4 presents the potential to become a complementary tool in the treatment of amoebic colitis.
Collapse
Affiliation(s)
- G K S Prado
- Postgraduate Program in Pathology, Institute of Biological Sciences of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - K C Torrinha
- Postgraduate Program in Pathology, Institute of Biological Sciences of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - R E Cruz
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - A B B Gonçalves
- Postgraduate Program in Pathology, Institute of Biological Sciences of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - C A V Silva
- Postgraduate Program in Pathology, Institute of Biological Sciences of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - F M S Oliveira
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - A C Nunes
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M A Gomes
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - M V Caliari
- Postgraduate Program in Pathology, Institute of Biological Sciences of the Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
35
|
Wombwell E, Patterson ME, Bransteitter B, Gillen LR. The Effect of Saccharomyces boulardii Primary Prevention on Risk of Hospital-onset Clostridioides difficile Infection in Hospitalized Patients Administered Antibiotics Frequently Associated With C. difficile Infection. Clin Infect Dis 2020; 73:e2512-e2518. [DOI: 10.1093/cid/ciaa808] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Background
Hospital-onset Clostridioides difficile infection (HO-CDI) is a costly problem leading to readmissions, morbidity, and mortality. We evaluated the effect of a single probiotic strain, Saccharomyces boulardii, at a standardized dose on the risk of HO-CDI within hospitalized patients administered antibiotics frequently associated with HO-CDI.
Methods
This retrospective cohort study merged hospital prescribing data with HO-CDI case data. The study assessed patients hospitalized from January 2016 through March 2017 who were administered at least 1 dose of an antibiotic frequently associated with HO-CDI during hospitalization. Associations between S. boulardii administration, including timing, and HO-CDI incidence were evaluated by multivariable logistic regression.
Results
The study included 8763 patients. HO-CDI incidence was 0.66% in the overall cohort. HO-CDI incidence was 0.56% and 0.82% among patients coadministered S. boulardii with antibiotics and not coadministered S. boulardii, respectively. In adjusted analysis, patients coadministered S. boulardii had a reduced risk of HO-CDI (odds ratio [OR], 0.57 [95% confidence interval {CI}, .33–.96]; P = .04) compared to patients not coadministered S. boulardii. Patients coadministered S. boulardii within 24 hours of antibiotic start demonstrated a reduced risk of HO-CDI (OR, 0.47 [95% CI, .23–.97]; P = .04) compared to those coadministered S. boulardii after 24 hours of antibiotic start.
Conclusions
Saccharomyces boulardii administered to hospitalized patients prescribed antibiotics frequently linked with HO-CDI was associated with a reduced risk of HO-CDI.
Collapse
Affiliation(s)
- Eric Wombwell
- Division of Pharmacy Practice and Administration, University of Missouri–Kansas City School of Pharmacy, Kansas City, Missouri, USA
- Department of Pharmacy, Centerpoint Medical Center, Independence, Missouri, USA
| | - Mark E Patterson
- Division of Pharmacy Practice and Administration, University of Missouri–Kansas City School of Pharmacy, Kansas City, Missouri, USA
| | | | - Lisa R Gillen
- Department of Pharmacy, Centerpoint Medical Center, Independence, Missouri, USA
| |
Collapse
|
36
|
Saccharomyces boulardii: What Makes It Tick as Successful Probiotic? J Fungi (Basel) 2020; 6:jof6020078. [PMID: 32512834 PMCID: PMC7344949 DOI: 10.3390/jof6020078] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Saccharomyces boulardii is a probiotic yeast often used for the treatment of GI tract disorders such as diarrhea symptoms. It is genetically close to the model yeast Saccharomyces cerevisiae and its classification as a distinct species or a S. cerevisiae variant has long been discussed. Here, we review the main genetic divergencies between S. boulardii and S. cerevisiae as a strategy to uncover the ability to adapt to the host physiological conditions by the probiotic. S. boulardii does possess discernible phenotypic traits and physiological properties that underlie its success as probiotic, such as optimal growth temperature, resistance to the gastric environment and viability at low pH. Its probiotic activity has been elucidated as a conjunction of multiple pathways, ranging from improvement of gut barrier function, pathogen competitive exclusion, production of antimicrobial peptides, immune modulation, and trophic effects. This review summarizes the participation of S. boulardii in these mechanisms and the multifactorial nature by which this yeast modulates the host microbiome and intestinal function.
Collapse
|
37
|
Saccharomyces boulardii CNCM I-745: A Non-bacterial Microorganism Used as Probiotic Agent in Supporting Treatment of Selected Diseases. Curr Microbiol 2020; 77:1987-1996. [PMID: 32472262 PMCID: PMC7415030 DOI: 10.1007/s00284-020-02053-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023]
Abstract
The yeast Saccharomyces boulardii CNCM I-745 is a unique, non-bacterial microorganism classified as a probiotic agent. In this review article, at first, we briefly summarized the mechanisms responsible for its probiotic properties, e.g. adhesion to and elimination of enteropathogenic microorganisms and their toxins; extracellular cleavage of pathogens’ virulent factors; trophic and anti-inflammatory effects on the intestinal mucosa. The efficacy of S. boulardii administration was tested in variety of human diseases. We discussed the results of S. boulardii CNCM I-745 use in the treatment or prevention of Helicobacter pylori infections, diarrhoea (Clostridium difficile infections, antibiotic-associated diarrhoea, and traveller’s diarrhoea), inflammatory bowel diseases, irritable bowel syndrome, candidiasis, dyslipidemia, and small intestine bacterial overgrowth in patients with multiple sclerosis. In case of limited number of studies regarding this strain, we also presented studies demonstrating properties and efficacy of other strains of S. boulardii. Administration of S. boulardii CNCMI I-745 during antibiotic therapy has certain advantage over bacterial probiotics, because—due to its fungal natural properties—it is intrinsically resistant to the antibiotics and cannot promote the spread of antimicrobial resistance. Even though cases of fungemia following S. boulardii CNCM I-745 administration were reported, it should be treated as a widely available and safe probiotic strain.
Collapse
|
38
|
Duan J, Meng X, Liu S, Zhou P, Zeng C, Fu C, Dou Q, Wu A, Li C. Gut Microbiota Composition Associated With Clostridium difficile-Positive Diarrhea and C. difficile Type in ICU Patients. Front Cell Infect Microbiol 2020; 10:190. [PMID: 32477962 PMCID: PMC7233261 DOI: 10.3389/fcimb.2020.00190] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/09/2020] [Indexed: 01/05/2023] Open
Abstract
The gut microbiota composition of intensive care unit (ICU) patients suffering from Clostridium difficile-positive diarrhea (CDpD) is poorly understood. This prospective study aims to use 16S rDNA (and metagenome) sequencing to compare the microbiota composition of 58 (and 5) ICU patients with CDpD (CDpD group), 33 (and 4) ICU patients with C. difficile-negative diarrhea (CDnD group), and 21 (and 5) healthy control subjects (control group), as well as CDpD patients in the A+B+ (N = 34; A/B: C. difficile TcdA/B), A−B+ (N = 7), and A−B− (N = 17) subgroups. For 16S rDNA data, OTU clustering (tool: UPARSE), taxonomic assignment (tool: RDP classifier), α-diversity, and β-diversity analyses (tool: QIIME) were conducted. For metagenome data, metagenome assembly (tool: SOAPdenovo), gene calling (tools: MetaGeneMark, CD-HIT, and SoapAligner), unigene alignment (tool: DIAMOND), taxon difference analysis (tool: Metastats), and gene annotation (tool: DIAMOND) were performed. The microbial diversity of the CDpD group was lower than that of the CDnD and control groups. The abundances of 10 taxa (e.g., Deferribacteres, Cryptomycota, Acetothermia) were significantly higher in the CDpD group than in the CDnD group. The abundances of Saccharomycetes and Clostridia were significantly lower in CDpD in comparison with control. Some taxa were significantly different between the A+B+ and A−B− subgroups. CDpD might relate to a decrease in beneficial taxa (i.e., Saccharomycetes and Clostridia) and an increase in harmful taxa (e.g., Deferribacteres, Cryptomycota, Acetothermia) in gut microbiota of ICU patients. C. difficile toxin type might be slightly associated with gut microbiota composition.
Collapse
Affiliation(s)
- Juping Duan
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China.,Changsha Hospital of Traditional Chinese Medicine, Changsha, China
| | - Xiujuan Meng
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Sidi Liu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Pengcheng Zhou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Cui Zeng
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Chenchao Fu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Qingya Dou
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Anhua Wu
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| | - Chunhui Li
- Infection Control Center, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
39
|
Zhang K, Zhang X, Lv A, Fan S, Zhang J. Saccharomyces boulardii modulates necrotizing enterocolitis in neonatal mice by regulating the sirtuin 1/NF‑κB pathway and the intestinal microbiota. Mol Med Rep 2020; 22:671-680. [PMID: 32626966 PMCID: PMC7339617 DOI: 10.3892/mmr.2020.11138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 03/17/2020] [Indexed: 12/19/2022] Open
Abstract
Exaggerated inflammatory response and gut microbial dysbiosis play a crucial role in necrotizing enterocolitis (NEC). The probiotic Saccharomyces boulardii (SB) is a yeast that has a beneficial effect on NEC; however, the association between its protective effects and the regulation of the inflammation-related sirtuin 1 (SIRT1)/nuclear factor-κB (NF-κB) signaling pathway and gut microbiota in NEC is unknown. In the present study, the NEC model was established by artificial feeding and lipopolysaccharide (LPS), hypoxia and hypothermia stimulation. Mice were divided into normal, control (artificial feeding), NEC and NEC + SB groups. Hematoxylin and eosin staining demonstrated that SB improved the pathological damage of the intestine caused by NEC in neonatal mice. Furthermore, downregulation of SIRT1 and upregulation of NF-κB expression were confirmed by immunofluorescence staining, western blotting and reverse transcription-quantitative PCR (RT-qPCR) in NEC mice. SB treatment concurrently inhibited the NEC roles on the SIRT1 and NF-κB pathway at both the protein and mRNA levels. Deletion of SIRT1 [SIRT1 knockout (KO)] in the intestine abolished all the effects of SB in NEC mice, including protection of pathological damage and inhibition of the SIRT1/NF-κB pathway activation. The abundance of gut microbial composition, as determined by RT-qPCR, was significantly decreased in the control group compared with the normal group. A further decrease in microbiota abundance was observed in the NEC group, and SB administration significantly improved the enrichment of gut microbiota in neonatal mice with NEC. As anticipated, the increased abundance of gut microbiota modulated by SB was markedly reduced in SIRT1KO NEC mice. The present study revealed that the protective role of SB on NEC was associated with the SIRT1/NF-κB pathway and gut microbiota regulation.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Xi Zhang
- Department of Obstetrics, Changning Maternity and Infant Health Hospital, Shanghai 200050, P.R. China
| | - Anping Lv
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Sainan Fan
- Department of Pediatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Jinping Zhang
- Department of Pediatrics, East Campus of Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 201306, P.R. China
| |
Collapse
|
40
|
Matijašić M, Meštrović T, Paljetak HČ, Perić M, Barešić A, Verbanac D. Gut Microbiota beyond Bacteria-Mycobiome, Virome, Archaeome, and Eukaryotic Parasites in IBD. Int J Mol Sci 2020; 21:E2668. [PMID: 32290414 PMCID: PMC7215374 DOI: 10.3390/ijms21082668] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/02/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The human microbiota is a diverse microbial ecosystem associated with many beneficial physiological functions as well as numerous disease etiologies. Dominated by bacteria, the microbiota also includes commensal populations of fungi, viruses, archaea, and protists. Unlike bacterial microbiota, which was extensively studied in the past two decades, these non-bacterial microorganisms, their functional roles, and their interaction with one another or with host immune system have not been as widely explored. This review covers the recent findings on the non-bacterial communities of the human gastrointestinal microbiota and their involvement in health and disease, with particular focus on the pathophysiology of inflammatory bowel disease.
Collapse
Affiliation(s)
- Mario Matijašić
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | | | - Hana Čipčić Paljetak
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Mihaela Perić
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Anja Barešić
- Division of Electronics, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Donatella Verbanac
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
41
|
Mycobiome in the Gut: A Multiperspective Review. Mediators Inflamm 2020; 2020:9560684. [PMID: 32322167 PMCID: PMC7160717 DOI: 10.1155/2020/9560684] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 02/23/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Human gut is home to a diverse and complex microbial ecosystem encompassing bacteria, viruses, parasites, fungi, and other microorganisms that have an undisputable role in maintaining good health for the host. Studies on the interplay between microbiota in the gut and various human diseases remain the key focus among many researchers. Nevertheless, advances in sequencing technologies and computational biology have helped us to identify a diversity of fungal community that reside in the gut known as the mycobiome. Although studies on gut mycobiome are still in its infancy, numerous sources have reported its potential role in host homeostasis and disease development. Nonetheless, the actual mechanism of its involvement remains largely unknown and underexplored. Thus, in this review, we attempt to discuss the recent advances in gut mycobiome research from multiple perspectives. This includes understanding the composition of fungal communities in the gut and the involvement of gut mycobiome in host immunity and gut-brain axis. Further, we also discuss on multibiome interactions in the gut with emphasis on fungi-bacteria interaction and the influence of diet in shaping gut mycobiome composition. This review also highlights the relation between fungal metabolites and gut mycobiota in human homeostasis and the role of gut mycobiome in various human diseases. This multiperspective review on gut mycobiome could perhaps shed new light for future studies in the mycobiome research area.
Collapse
|
42
|
Unique genetic basis of the distinct antibiotic potency of high acetic acid production in the probiotic yeast Saccharomyces cerevisiae var. boulardii. Genome Res 2020; 29:1478-1494. [PMID: 31467028 PMCID: PMC6724677 DOI: 10.1101/gr.243147.118] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 06/20/2019] [Indexed: 12/14/2022]
Abstract
The yeast Saccharomyces boulardii has been used worldwide as a popular, commercial probiotic, but the basis of its probiotic action remains obscure. It is considered conspecific with budding yeast Saccharomyces cerevisiae, which is generally used in classical food applications. They have an almost identical genome sequence, making the genetic basis of probiotic potency in S. boulardii puzzling. We now show that S. boulardii produces at 37°C unusually high levels of acetic acid, which is strongly inhibitory to bacterial growth in agar-well diffusion assays and could be vital for its unique application as a probiotic among yeasts. Using pooled-segregant whole-genome sequence analysis with S. boulardii and S. cerevisiae parent strains, we succeeded in mapping the underlying QTLs and identified mutant alleles of SDH1 and WHI2 as the causative alleles. Both genes contain a SNP unique to S. boulardii (sdh1F317Y and whi2S287*) and are fully responsible for its high acetic acid production. S. boulardii strains show different levels of acetic acid production, depending on the copy number of the whi2S287* allele. Our results offer the first molecular explanation as to why S. boulardii could exert probiotic action as opposed to S. cerevisiae. They reveal for the first time the molecular-genetic basis of a probiotic action-related trait in S. boulardii and show that antibacterial potency of a probiotic microorganism can be due to strain-specific mutations within the same species. We suggest that acquisition of antibacterial activity through medium acidification offered a selective advantage to S. boulardii in its ecological niche and for its application as a probiotic.
Collapse
|
43
|
Shamekhi S, Lotfi H, Abdolalizadeh J, Bonabi E, Zarghami N. An overview of yeast probiotics as cancer biotherapeutics: possible clinical application in colorectal cancer. Clin Transl Oncol 2020; 22:1227-1239. [PMID: 31919760 DOI: 10.1007/s12094-019-02270-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/08/2019] [Indexed: 02/07/2023]
Abstract
The previous reports have established a strong link between diet, lifestyle, and gut microbiota population with the onset of the colorectal cancer (CRC). Administration of probiotics has become a particular interest in prevention and treatment of CRC. As potential dietary complements, probiotics might be able to lower the risk of CRC and manage the safety of traditional cancer therapies such as surgery, radiation therapy, and chemotherapy. This review investigates the promising effects of probiotics as biotherapeutics, with due attention to possible clinical application of yeast probiotics in prevention and treatment of CRC. In addition, various underlying anti-cancer mechanisms are covered here based on scientific evidence and findings from numerous experimental studies. Application of probiotics as biotherapeutics in CRC, however, needs to be approved by human clinical trials. It is of prime concern, to find potential probiotic strains, effective doses for administrations and regimes, and molecular mechanisms involved in prevention and treatment.
Collapse
Affiliation(s)
- S Shamekhi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - H Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - J Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - E Bonabi
- Department of Medical Microbiology, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| | - N Zarghami
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Clinical Biochemistry and Laboratory Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
44
|
Probiotic Supplementation in a Clostridium difficile-Infected Gastrointestinal Model Is Associated with Restoring Metabolic Function of Microbiota. Microorganisms 2019; 8:microorganisms8010060. [PMID: 31905795 PMCID: PMC7023328 DOI: 10.3390/microorganisms8010060] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/20/2019] [Accepted: 12/27/2019] [Indexed: 01/19/2023] Open
Abstract
Clostridium (C.) difficile-infection (CDI), a nosocomial gastrointestinal disorder, is of growing concern due to its rapid rise in recent years. Antibiotic therapy of CDI is associated with disrupted metabolic function and altered gut microbiota. The use of probiotics as an adjunct is being studied extensively due to their potential to modulate metabolic functions and the gut microbiota. In the present study, we assessed the ability of several single strain probiotics and a probiotic mixture to change the metabolic functions of normal and C. difficile-infected fecal samples. The production of short-chain fatty acids (SCFAs), hydrogen sulfide (H2S), and ammonia was measured, and changes in microbial composition were assessed by 16S rRNA gene amplicon sequencing. The C. difficile-infection in fecal samples resulted in a significant decrease (p < 0.05) in SCFA and H2S production, with a lower microbial alpha diversity. All probiotic treatments were associated with significantly increased (p < 0.05) levels of SCFAs and restored H2S levels. Probiotics showed no effect on microbial composition of either normal or C. difficile-infected fecal samples. These findings indicate that probiotics may be useful to improve the metabolic dysregulation associated with C. difficile infection.
Collapse
|
45
|
Elghandour M, Tan Z, Abu Hafsa S, Adegbeye M, Greiner R, Ugbogu E, Cedillo Monroy J, Salem A. Saccharomyces cerevisiaeas a probiotic feed additive to non and pseudo‐ruminant feeding: a review. J Appl Microbiol 2019; 128:658-674. [DOI: 10.1111/jam.14416] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 07/06/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022]
Affiliation(s)
- M.M.Y. Elghandour
- Facultad de Medicina Veterinaria y Zootecnia Universidad Autónoma del Estado de MéxicoEstado de México México
| | - Z.L. Tan
- CAS Key Laboratory for Agro‐Ecological Processes in Subtropical Region The Chinese Academy of Sciences Changsha China
| | - S.H. Abu Hafsa
- Department of Livestock Research Arid Lands Cultivation Research InstituteCity of Scientific Research and Technological Applications Alexandria Egypt
| | - M.J. Adegbeye
- Department of Animal Science, College of Agriculture Joseph Ayo Babalola University Ilesha Osun State Nigeria
| | - R. Greiner
- Department of Food Technology and Bioprocess Engineering Max Rubner‐InstitutFederal Research Institute of Nutrition and Food Karlsruhe Germany
| | - E.A. Ugbogu
- Department of Biochemistry Abia State University Uturu Abia State Nigeria
| | - J. Cedillo Monroy
- Centro Universitario UAEM‐Temascaltepec Universidad Autónoma del Estado de México Toluca México
| | - A.Z.M. Salem
- Facultad de Medicina Veterinaria y Zootecnia Universidad Autónoma del Estado de MéxicoEstado de México México
| |
Collapse
|
46
|
Wang G, Feng D. Therapeutic effect of Saccharomyces boulardii combined with Bifidobacterium and on cellular immune function in children with acute diarrhea. Exp Ther Med 2019; 18:2653-2659. [PMID: 31572514 PMCID: PMC6755444 DOI: 10.3892/etm.2019.7836] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 07/23/2019] [Indexed: 12/30/2022] Open
Abstract
Clinical effect of Saccharomyces boulardii combined with bifidobacterium and its effect on cellular immune function in children with acute diarrhea were studied. In total 116 cases of children with acute diarrhea admitted to Xuzhou Children's Hospital from March 2015 to March 2017 were collected and analyzed retrospectively. There were 59 children treated with Saccharomyces boulardii as control group and 57 children treated with Saccharomyces boulardii combined with bifidobacterium as experimental group. The clinical effect, stool frequency in different time periods, mean antidiarrheal time, mean antipyretic time and length of stay, and immune function of children in the two groups after treatment were analyzed. The cure rate (73.68%) and the total effective rate (87.72%) in the experimental group were significantly higher than those in the control group (47.46 and 71.19%) (P<0.05). The stool frequency in the experimental group was significantly lower than that in the control group 3 days after treatment (P<0.05). The mean antidiarrheal time in the experimental group was significantly shorter than that in the control group (P<0.05). The length of stay in the control group was significantly longer than that in the experimental group (P<0.05). CD3+, CD4+ and CD4+/CD8+ increased significantly in the experimental group after treatment while CD8+ decreased significantly (P<0.05). After treatment, the ratio of Th1 and Th2 in the two groups decreased significantly compared with before treatment (P<0.05), and the experimental group was significantly lower than the control group (P<0.05). After treatment, Th1/Th2 ratio was significantly higher than that before treatment (P<0.05), and the experimental group was significantly higher than the control group (P<0.05). In conclusion, treatment of acute diarrhea in children with Saccharomyces boulardii combined with bifidobacterium can effectively shorten the duration of diarrhea and hospital stay, reduce the number of diarrhea and enhance the cellular immune function.
Collapse
Affiliation(s)
- Guangmeng Wang
- Department of Gastroenterology, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Dongjin Feng
- Department of Gastroenterology, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
47
|
Czerucka D, Rampal P. Diversity of Saccharomyces boulardii CNCM I-745 mechanisms of action against intestinal infections. World J Gastroenterol 2019; 25:2188-2203. [PMID: 31143070 PMCID: PMC6526157 DOI: 10.3748/wjg.v25.i18.2188] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/21/2019] [Accepted: 03/30/2019] [Indexed: 02/06/2023] Open
Abstract
The yeast Saccharomyces boulardii CNCM I-745 is one of the probiotics recommended for the prevention of antibiotic-associated diarrhea. Studies conducted in vivo and in vitro demonstrated that in the case of infectious diseases there are two potential sites of action of Saccharomyces boulardii CNCM I-745: (1) An action on enteropathogenic microorganisms (adhesion of bacteria and their elimination or an effect on their virulence factors: Toxins, lipopolysaccharide, etc.); and (2) a direct action on the intestinal mucosa (trophic effects, effects on epithelial reconstitution, anti-secretory effects, anti-inflammatory, immunomodulators). Oral administration of Saccharomyces boulardii CNCM I-745 to healthy subjects does not alter their microbiota. However, in the case of diseases associated with the use of antibiotics or chronic diarrhea, Saccharomyces boulardii CNCM I-745 can restore the intestinal microbiota faster. The interaction of Saccharomyces boulardii CNCM I-745 with the innate immune system have been recently demonstrated thus opening up a new therapeutic potential of this yeast in the case of diseases associated with intestinal infections but also other pathologies associated with dysbiosis such as inflammatory diseases.
Collapse
Affiliation(s)
- Dorota Czerucka
- Department of Human Health, Division of Ecosystems and Immunity, Center Scientific of Monaco, Monaco MC98000, Monaco
| | | |
Collapse
|
48
|
Rowan-Nash AD, Korry BJ, Mylonakis E, Belenky P. Cross-Domain and Viral Interactions in the Microbiome. Microbiol Mol Biol Rev 2019; 83:e00044-18. [PMID: 30626617 PMCID: PMC6383444 DOI: 10.1128/mmbr.00044-18] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The importance of the microbiome to human health is increasingly recognized and has become a major focus of recent research. However, much of the work has focused on a few aspects, particularly the bacterial component of the microbiome, most frequently in the gastrointestinal tract. Yet humans and other animals can be colonized by a wide array of organisms spanning all domains of life, including bacteria and archaea, unicellular eukaryotes such as fungi, multicellular eukaryotes such as helminths, and viruses. As they share the same host niches, they can compete with, synergize with, and antagonize each other, with potential impacts on their host. Here, we discuss these major groups making up the human microbiome, with a focus on how they interact with each other and their multicellular host.
Collapse
Affiliation(s)
- Aislinn D Rowan-Nash
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
49
|
Feng P, Ye Z, Kakade A, Virk AK, Li X, Liu P. A Review on Gut Remediation of Selected Environmental Contaminants: Possible Roles of Probiotics and Gut Microbiota. Nutrients 2018; 11:nu11010022. [PMID: 30577661 PMCID: PMC6357009 DOI: 10.3390/nu11010022] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Various environmental contaminants including heavy metals, pesticides and antibiotics can contaminate food and water, leading to adverse effects on human health, such as inflammation, oxidative stress and intestinal disorder. Therefore, remediation of the toxicity of foodborne contaminants in human has become a primary concern. Some probiotic bacteria, mainly Lactobacilli, have received a great attention due to their ability to reduce the toxicity of several contaminants. For instance, Lactobacilli can reduce the accumulation and toxicity of selective heavy metals and pesticides in animal tissues by inhibiting intestinal absorption of contaminants and enhancing intestinal barrier function. Probiotics have also shown to decrease the risk of antibiotic-associated diarrhea possibly via competing and producing antagonistic compounds against pathogenic bacteria. Furthermore, probiotics can improve immune function by enhancing the gut microbiota mediated anti-inflammation. Thus, these probiotic bacteria are promising candidates for protecting body against foodborne contaminants-induced toxicity. Study on the mechanism of these beneficial bacterial strains during remediation processes and particularly their interaction with host gut microbiota is an active field of research. This review summarizes the current understanding of the remediation mechanisms of some probiotics and the combined effects of probiotics and gut microbiota on remediation of foodborne contaminants in vivo.
Collapse
Affiliation(s)
- Pengya Feng
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Ze Ye
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Apurva Kakade
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Amanpreet Kaur Virk
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Xiangkai Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Pu Liu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| |
Collapse
|
50
|
Clostridium difficile, the Difficult "Kloster" Fuelled by Antibiotics. Curr Microbiol 2018; 76:774-782. [PMID: 30084095 DOI: 10.1007/s00284-018-1543-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Clostridium difficile is normally present in low numbers in a healthy adult gastro-intestinal tract (GIT). Drastic changes in the microbial population, e.g., dysbiosis caused by extensive treatment with antibiotics, stimulates the growth of resistant strains and the onset of C. difficile infection (CDI). Symptoms of infection varies from mild diarrhea to colitis (associated with dehydration and bleeding), pseudomembranous colitis with yellow ulcerations in the mucosa of the colon, to fulminant colitis (perforation of the gut membrane), and multiple organ failure. Inflamed epithelial cells and damaged mucosal tissue predisposes the colon to other opportunistic pathogens such as Clostridium perfringens, Staphylococcus aureus, Klebsiella oxytoca, Candida spp., and Salmonella spp. This may lead to small intestinal bacterial overgrowth (SIBO), sepsis, toxic megacolon, and even colorectal cancer. Many stains of C. difficile are resistant to metronidazole and vancomycin. Vaccination may be an answer to CDI, but requires more research. Success in treatment with probiotics depends on the strains used. Oral or rectal fecal transplants are partly effective, as spores in the small intestine may germinate and colonize the colon. The effect of antibiotics on C. difficile and commensal gut microbiota is summarized and changes in gut physiology are discussed. The need to search for non-antibiotic methods in the treatment of CDI and C. difficile-associated disease (CDAD) is emphasized.
Collapse
|