1
|
Woelfel S, Silva MS, Stecher B. Intestinal colonization resistance in the context of environmental, host, and microbial determinants. Cell Host Microbe 2024; 32:820-836. [PMID: 38870899 DOI: 10.1016/j.chom.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024]
Abstract
Microbial communities that colonize the human gastrointestinal (GI) tract defend against pathogens through a mechanism known as colonization resistance (CR). Advances in technologies such as next-generation sequencing, gnotobiotic mouse models, and bacterial cultivation have enhanced our understanding of the underlying mechanisms and the intricate microbial interactions involved in CR. Rather than being attributed to specific microbial clades, CR is now understood to arise from a dynamic interplay between microbes and the host and is shaped by metabolic, immune, and environmental factors. This evolving perspective underscores the significance of contextual factors, encompassing microbiome composition and host conditions, in determining CR. This review highlights recent research that has shifted its focus toward elucidating how these factors interact to either promote or impede enteric infections. It further discusses future research directions to unravel the complex relationship between host, microbiota, and environmental determinants in safeguarding against GI infections to promote human health.
Collapse
Affiliation(s)
- Simon Woelfel
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Marta Salvado Silva
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer-Institute for Hygiene and Clinical Microbiology, Ludwig Maximilian University of Munich, 80336 Munich, Germany; German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany.
| |
Collapse
|
2
|
Zych-Krekora K, Sylwestrzak O, Krekora M, Oszukowski P, Grzesiak M. Potential Impact of a Pregnant Woman's Microbiota on the Development of Fetal Heart Defects: A Review of the Literature. Biomedicines 2024; 12:654. [PMID: 38540267 PMCID: PMC10968164 DOI: 10.3390/biomedicines12030654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 11/11/2024] Open
Abstract
Developments in medicine and biology in recent decades have led to a significant increase in our knowledge of the complex interactions between the microbiota and human health. In the context of perinatal medicine and neonatology, particular attention is being paid to the potential impact of the maternal microbiota on fetal development. Among the many aspects of this relationship, the question of the impact of dysbiosis on the development of fetal heart defects is an important one. In this article, we present an analysis of recent research and scientific evidence on the relationship between a pregnant woman's microbiota and the development of fetal heart defects. We also discuss potential intervention strategies, including the role of probiotics and diet in optimising the maternal microbiota.
Collapse
Affiliation(s)
- Katarzyna Zych-Krekora
- Department of Perinatology, Obstetrics and Gynaecology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
| | - Oskar Sylwestrzak
- Department of Obstetrics and Gynaecology, Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland; (O.S.); (M.K.)
- Department of Prenatal Cardiology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland
| | - Michał Krekora
- Department of Obstetrics and Gynaecology, Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland; (O.S.); (M.K.)
- Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-419 Lodz, Poland
| | - Przemysław Oszukowski
- Obstetrics Ward of the Polish Mother’s Memorial Health Institute, 93-338 Lodz, Poland;
- Department of Obstetrics, Gynaecology and Gynaecology Oncology, Medical University of Lodz, 93-113 Lodz, Poland
| | - Mariusz Grzesiak
- Department of Perinatology, Obstetrics and Gynaecology, Polish Mother’s Memorial Hospital Research Institute, 93-338 Lodz, Poland;
- Department of Gynaecology and Obstetrics, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
3
|
Deng L, Wang S. Colonization resistance: the role of gut microbiota in preventing Salmonella invasion and infection. Gut Microbes 2024; 16:2424914. [PMID: 39514544 PMCID: PMC11552263 DOI: 10.1080/19490976.2024.2424914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The human gastrointestinal tract is colonized by a complex microbial ecosystem, the gut microbiota, which is pivotal in maintaining host health and mediating resistance to diseases. This review delineates colonization resistance (CR), a critical defensive mechanism employed by the gut microbiota to safeguard against pathogenic bacterial invasions, notably by Salmonella. We detail the mechanisms through which the gut microbiota impedes Salmonella colonization, including nutrient competition, production of antimicrobial peptides, synthesis of microbial-derived metabolites, and modulation of the host immune response. Additionally, we examine how dietary interventions can influence these mechanisms, thereby augmenting the protective role of the gut microbiota. The review also discusses the sophisticated strategies utilized by Salmonella to overcome these microbial defenses. A thorough understanding of these complex interactions between microbial symbionts and pathogens is crucial for the development of innovative therapeutic strategies that enhance CR, aiming to prevent or treat microbial infections effectively.
Collapse
Affiliation(s)
- Lei Deng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Shaohui Wang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
4
|
Kaewarsar E, Chaiyasut C, Lailerd N, Makhamrueang N, Peerajan S, Sirilun S. Optimization of Mixed Inulin, Fructooligosaccharides, and Galactooligosaccharides as Prebiotics for Stimulation of Probiotics Growth and Function. Foods 2023; 12:foods12081591. [PMID: 37107386 PMCID: PMC10137966 DOI: 10.3390/foods12081591] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Prebiotics have become an important functional food because of their potential for modulating the gut microbiota and metabolic activities. However, different prebiotics can stimulate the growth of different probiotics. The optimization of prebiotics was focused on in this study in order to stimulate the representative probiotics' growth (Lacticaseibacillus rhamnosus (previously Lactobacillus rhamnosus) and Bifidobacterium animalis subsp. lactis) and their function. The culture medium was supplemented with three prebiotics, including inulin (INU), fructooligosaccharides (FOS), and galactooligosaccharides (GOS). All prebiotics can clearly stimulate the growth of probiotic strains in both monoculture and co-culture. The specific growth rates of L. rhamnosus and B. animalis subsp. lactis were shown in GOS (0.019 h-1) and FOS (0.023 h-1), respectively. The prebiotic index (PI) scores of INU (1.03), FOS (0.86), and GOS (0.84) in co-culture at 48 h were significantly higher than the control (glucose). The mixture of prebiotics to achieve high quality was optimized using the Box-Behnken design. The optimum prebiotic ratios of INU, FOS, and GOS were 1.33, 2.00, and 2.67% w/v, respectively, with the highest stimulated growth of probiotic strains occurring with the highest PI score (1.03) and total short chain fatty acid concentration (85.55 µmol/mL). The suitable ratio of mixed prebiotics will function as a potential ingredient for functional foods or colonic foods.
Collapse
Affiliation(s)
- Ekkachai Kaewarsar
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyavat Chaiyasut
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Narissara Lailerd
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Netnapa Makhamrueang
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Sasithorn Sirilun
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
5
|
Tang HJ, Chen CC, Lu YC, Huang HL, Chen HJ, Chuang YC, Lai CC, Chao CM. The effect of Lactobacillus with prebiotics on KPC-2-producing Klebsiella pneumoniae. Front Microbiol 2022; 13:1050247. [PMID: 36569071 PMCID: PMC9767986 DOI: 10.3389/fmicb.2022.1050247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives This study investigated the inhibitory effect of Lactobacillus spp. with prebiotics against Klebsiella pneumoniae carbapenemase-2 (KPC-2)-producing Klebsiella pneumoniae using both in vitro experiments and animal models. Methods Thirty-three Lactobacillus spp. strains were confirmed by 16S rDNA sequencing, and four different PFGE genotyped KPC-2-producing K. pneumoniae strains were selected for investigation. In vitro studies, including broth microdilution assays, changes in pH values in lactobacilli cultures with different prebiotics, time-kill tests of Lactobacillus spp. against KPC-2-producing K. pneumoniae and further in vivo Lactobacillus alone or in combination with prebiotics against KPC-2-producing K. pneumoniae in an animal model, were performed. Results The lower pH value of the cell-free supernatant was associated with a lower minimal inhibitory percentage of the Lactobacillus strain against KPC-2-producing K. pneumoniae. Furthermore, lactulose/isomalto-oligosaccharide/inulin and fructo-oligosaccharide can enhance the inhibitory effect of all 107 CFU/ml Lactobacillus strains against KPC001. Three Lactobacillus strains (LYC1154, LYC1322, and LYC1511) that could be persistently detected in the stool were tested for their ability to reduce the amount of KPC001 in the feces individually or in combination. A significantly better effect in reducing the amount of KPC001 was observed for the combination of three different Lactobacillus species than for each of them alone. Furthermore, their inhibitory effect was enhanced after adding lactulose or isomalto-oligosaccharide (both p < 0.05). Conclusion This study demonstrates the inhibitory effect of probiotic Lactobacillus, including LYC1154, LYC1322, and LYC1511, with prebiotics such as lactulose or isomalto-oligosaccharide against the colonization of KPC-2-producing K. pneumoniae.
Collapse
Affiliation(s)
- Hung-Jen Tang
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan,Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chi-Chung Chen
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Ying-Chen Lu
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Hui-Ling Huang
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Hung-Jui Chen
- Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yin-Ching Chuang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chih-Cheng Lai
- Division of Hospital Medicine, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Chien-Ming Chao
- Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan,*Correspondence: Chien-Ming Chao,
| |
Collapse
|
6
|
Han D, Zulewska J, Xiong K, Yang Z. Synergy between oligosaccharides and probiotics: From metabolic properties to beneficial effects. Crit Rev Food Sci Nutr 2022; 64:4078-4100. [PMID: 36315042 DOI: 10.1080/10408398.2022.2139218] [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] [Indexed: 11/03/2022]
Abstract
Synbiotic is defined as the dietary mixture that comprises both probiotic microorganisms and prebiotic substrates. The concept has been steadily gaining attention owing to the rising recognition of probiotic, prebiotics, and gut health. Among prebiotic substances, oligosaccharides demonstrated considerable health beneficial effects in varieties of food products and their combination with probiotics have been subjected to full range of evaluations. This review delineated the landscape of studies using microbial cultures, cell lines, animal model, and human subjects to explore the functional properties and host impacts of these combinations. Overall, the results suggested that these combinations possess respective metabolic properties that could facilitate beneficial activities therefore could be employed as dietary interventions for human health improvement and therapeutic purposes. However, uncertainties, such as applicational practicalities, underutilized analytical tools, contradictory results in studies, unclear mechanisms, and legislation hurdles, still challenges the broad utilization of these combinations. Future studies to address these issues may not only advance current knowledge on probiotic-prebiotic-host interrelationship but also promote respective applications in food and nutrition.
Collapse
Affiliation(s)
- Dong Han
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
- Key Laboratory of Food Bioengineering (China National Light Industry), College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Justyna Zulewska
- Department of Dairy Science and Quality Management, Faculty of Food Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Ke Xiong
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Zhennai Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing, China
| |
Collapse
|
7
|
Effects of Taro (Colocasia esculenta) Water-Soluble Non-Starch Polysaccharide, Lactobacillus acidophilus, Bifidobacterium breve, Bifidobacterium infantis, and Their Synbiotic Mixtures on Pro-Inflammatory Cytokine Interleukin-8 Production. Nutrients 2022; 14:nu14102128. [PMID: 35631269 PMCID: PMC9147535 DOI: 10.3390/nu14102128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/25/2022] Open
Abstract
In the past decades, the regulation of pro-inflammatory cytokine production, including interleukin-8 (IL-8), has been the goal of many targeted therapeutic interventions for Necrotising enterocolitis (NEC), a gastrointestinal disease commonly associated with a very low birth weight in preterm infants. In this study, the ability to regulate the production of IL-8 of the water-soluble non-starch polysaccharide (WS-NSP) from taro corm (Tc-WS-NSP) extracted using a conventional (CE) or improved conventional (ICE) extraction method, of the probiotics Lactobacillus acidophilus, Bifidobacterium breve, and Bifidobacterium infantis, and their synbiotic mixtures were evaluated. The TNF-α stimulated HT-29 cells were incubated with undigested or digested Tc-WS-NSPs (CE or ICE), probiotics, and their synbiotic mixtures with Klebsiella oxytoca, an NEC-positive-associated pathogen. Overall, the synbiotic mixtures of digested Tc-WS-NSP-ICE and high bacterial concentrations of L. acidophilus (5.57 × 109), B. breve (2.7 × 108 CFU/mL), and B. infantis (1.53 × 108) demonstrated higher (42.0%, 45.0%, 43.1%, respectively) ability to downregulate IL-8 compared to the sole use of Tc-WS-NSPs (24.5%), or the probiotics L. acidophilus (32.3%), B. breve (37.8%), or B. infantis (33.1%). The ability demonstrated by the Tc-WS-NSPs, the probiotics, and their synbiotics mixtures to downregulate IL-8 production in the presence of an NEC-positive-associated pathogen may be useful in the development of novel prophylactic agents against NEC.
Collapse
|
8
|
Sauvaitre T, Etienne-Mesmin L, Sivignon A, Mosoni P, Courtin CM, Van de Wiele T, Blanquet-Diot S. Tripartite relationship between gut microbiota, intestinal mucus and dietary fibers: towards preventive strategies against enteric infections. FEMS Microbiol Rev 2021; 45:5918835. [PMID: 33026073 DOI: 10.1093/femsre/fuaa052] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The human gut is inhabited by a large variety of microorganims involved in many physiological processes and collectively referred as to gut microbiota. Disrupted microbiome has been associated with negative health outcomes and especially could promote the onset of enteric infections. To sustain their growth and persistence within the human digestive tract, gut microbes and enteric pathogens rely on two main polysaccharide compartments, namely dietary fibers and mucus carbohydrates. Several evidences suggest that the three-way relationship between gut microbiota, dietary fibers and mucus layer could unravel the capacity of enteric pathogens to colonise the human digestive tract and ultimately lead to infection. The review starts by shedding light on similarities and differences between dietary fibers and mucus carbohydrates structures and functions. Next, we provide an overview of the interactions of these two components with the third partner, namely, the gut microbiota, under health and disease situations. The review will then provide insights into the relevance of using dietary fibers interventions to prevent enteric infections with a focus on gut microbial imbalance and impaired-mucus integrity. Facing the numerous challenges in studying microbiota-pathogen-dietary fiber-mucus interactions, we lastly describe the characteristics and potentialities of currently available in vitro models of the human gut.
Collapse
Affiliation(s)
- Thomas Sauvaitre
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France.,Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Adeline Sivignon
- Université Clermont Auvergne, UMR 1071 Inserm, USC-INRAe 2018, Microbes, Intestin, Inflammation et Susceptibilité de l'Hôte (M2iSH), Clermont-Ferrand, France
| | - Pascale Mosoni
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| | - Christophe M Courtin
- KU Leuven, Faculty of Bioscience Engineering, Laboratory of Food Chemistry and Biochemistry & Leuven Food Science and Nutrition Research Centre (LFoRCe), Leuven, Belgium
| | - Tom Van de Wiele
- Ghent University, Faculty of Bioscience Engineering, Center for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, UMR 454 INRAe, Microbiology, Digestive Environment and Health (MEDIS), Clermont-Ferrand, France
| |
Collapse
|
9
|
Structure and Function of Bovine Whey Derived Oligosaccharides Showing Synbiotic Epithelial Barrier Protective Properties. Nutrients 2020; 12:nu12072007. [PMID: 32640639 PMCID: PMC7400958 DOI: 10.3390/nu12072007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/15/2023] Open
Abstract
Commensal gut microbiota and probiotics have numerous effects on the host’s metabolic and protective systems, which occur primarily through the intestinal epithelial cell interface. Prebiotics, like galacto-oligosaccharides (GOS) are widely used to modulate their function and abundance. However, important structure–function relations may exist, requiring a detailed structural characterization. Here, we detailed the structural characterization of bovine whey derived oligosaccharide preparations enriched with GOS or not, dubbed GOS-enriched milk oligosaccharides (GMOS) or MOS, respectively. We explore GMOS’s and MOS’s potential to improve intestinal epithelial barrier function, assessed in a model based on barrier disruptive effects of the Clostridioides difficile toxin A. GMOS and MOS contain mainly GOS species composed of β1-6- and β1-3-linked galactoses, and 3′- and 6′-sialyllactose. Both GMOS and MOS, combined with lactobacilli, like Lactobacillus rhamnosus (LPR, NCC4007), gave synergistic epithelial barrier protection, while no such effect was observed with Bifidobacterium longum (BL NCC3001), Escherichia coli (Nissle) or fructo-oligosaccharides. Mechanistically, for barrier protection with MOS, (i) viable LPR was required, (ii) acidification of growth medium was not enough, (iii) LPR did not directly neutralize toxin A, and (iv) physical proximity of LPR with the intestinal epithelial cells was necessary. This is the first study, highlighting the importance of structure–function specificity and the necessity of the simultaneous presence of prebiotic, probiotic and host cell interactions required for a biological effect.
Collapse
|
10
|
Effect of Lactobacillus rhamnosus NCDC 298 with FOS in Combination on Viability and Toxin Production of Enterotoxigenic Escherichia coli. Probiotics Antimicrob Proteins 2019; 11:23-29. [PMID: 28948579 DOI: 10.1007/s12602-017-9327-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The present study was to investigate the utilization of prebiotics by Lactobacillus rhamnosus NCDC 298 and its synergistic adversary effect on both population and production of heat-labile (LT) toxin in enterotoxigenic Escherichia coli (ETEC). To select suitable prebiotic in order to enhance functionality, its utilization and the prebiotic activity score was examined. Antivirulence effect on ETEC was inspected by its inactivation rate and heat-labile toxin production in presence of different synbiotic combination. L. rhamnosus NCDC 298 strain grown well on media supplemented with fructooligosaccharides (FOS) and galactooligosaccharides (GOS), whereas significant inactivation of ETEC was observed when FOS was added to the co-culture medium. Significant decrease in LT enterotoxin was seen through GM1 ganglioside enzyme linked immunoassay (GM1 ELISA), when ETEC has grown with L. rhamnosus NCDC 298 and FOS. Short-chain FOS proved to be the most effective substrate, improving antagonistic activity for L. rhamnosus NCDC 298. Both L. rhamnosus NCDC 298 with FOS can be used as an effective synbiotic combination for secretory antidiarrheal fermented dairy formulations.
Collapse
|
11
|
Tarhani F, Nezami A. Role of probiotics in treatment of congenital heart disease and necrotizing enterocolitis. PHARMANUTRITION 2019. [DOI: 10.1016/j.phanu.2019.100144] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
12
|
Lyu Y, Wu L, Wang F, Shen X, Lin D. Carotenoid supplementation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis. Exp Biol Med (Maywood) 2018. [PMID: 29534601 DOI: 10.1177/1535370218763760] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis, a broad spectrum of imbalance of the gut microbiota, may progress to microbiota dysfunction. Dysbiosis is linked to some human diseases, such as inflammation-related disorders and metabolic syndromes. However, the underlying mechanisms of the pathogenesis of dysbiosis remain elusive. Recent findings suggest that the microbiome and gut immune responses, like immunoglobulin A production, play critical roles in the gut homeostasis and function, and the progression of dysbiosis. In the past two decades, much progress has been made in better understanding of production of immunoglobulin A and its association with commensal microbiota. The present minireview summarizes the recent findings in the gut microbiota dysbiosis and dysfunction of immunoglobulin A induced by the imbalance of pathogenic bacteria and commensal microbiota. We also propose the potentials of dietary carotenoids, such as β-carotene and astaxanthin, in the improvement of the gut immune system maturation and immunoglobulin A production, and the consequent promotion of the gut health. Impact statement The concept of carotenoid metabolism in the gut health has not been well established in the literature. Here, we review and discuss the roles of retinoic acid and carotenoids, including pro-vitamin A carotenoids and xanthophylls in the maturation of the gut immune system and IgA production. This is the first review article about the carotenoid supplements and the metabolites in the regulation of the gut microbiome. We hope this review would provide a new direction for the management of the gut microbiota dysbiosis by application of bioactive carotenoids and the metabolites.
Collapse
Affiliation(s)
- Yi Lyu
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Lei Wu
- 2 Department of Nutritional Sciences, 7618 Oklahoma State University, Stillwater , OK 74078, USA
| | - Fang Wang
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Xinchun Shen
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Dingbo Lin
- 2 Department of Nutritional Sciences, 7618 Oklahoma State University, Stillwater , OK 74078, USA
| |
Collapse
|
13
|
Markowiak P, Śliżewska K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017; 9:E1021. [PMID: 28914794 PMCID: PMC5622781 DOI: 10.3390/nu9091021] [Citation(s) in RCA: 1207] [Impact Index Per Article: 150.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/06/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
The human gastrointestinal tract is colonised by a complex ecosystem of microorganisms. Intestinal bacteria are not only commensal, but they also undergo a synbiotic co-evolution along with their host. Beneficial intestinal bacteria have numerous and important functions, e.g., they produce various nutrients for their host, prevent infections caused by intestinal pathogens, and modulate a normal immunological response. Therefore, modification of the intestinal microbiota in order to achieve, restore, and maintain favourable balance in the ecosystem, and the activity of microorganisms present in the gastrointestinal tract is necessary for the improved health condition of the host. The introduction of probiotics, prebiotics, or synbiotics into human diet is favourable for the intestinal microbiota. They may be consumed in the form of raw vegetables and fruit, fermented pickles, or dairy products. Another source may be pharmaceutical formulas and functional food. This paper provides a review of available information and summarises the current knowledge on the effects of probiotics, prebiotics, and synbiotics on human health. The mechanism of beneficial action of those substances is discussed, and verified study results proving their efficacy in human nutrition are presented.
Collapse
Affiliation(s)
- Paulina Markowiak
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Lodz University of Technology, 90-924 Łódź, Poland.
| | - Katarzyna Śliżewska
- Institute of Fermentation Technology and Microbiology, Department of Biotechnology and Food Sciences, Lodz University of Technology, 90-924 Łódź, Poland.
| |
Collapse
|
14
|
Mohan A, Quek SY, Gutierrez-Maddox N, Gao Y, Shu Q. Effect of honey in improving the gut microbial balance. FOOD QUALITY AND SAFETY 2017. [DOI: 10.1093/fqs/fyx015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Mohan A, Quek SY, Gutierrez-Maddox N, Gao Y, Shu Q. Effect of honey in improving the gut microbial balance. FOOD QUALITY AND SAFETY 2017. [DOI: 10.1093/fqsafe/fyx015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Wang C, Ma Y, Hu Q, Xie T, Wu J, Zeng F, Song F. Bifidobacterial recombinant thymidine kinase-ganciclovir gene therapy system induces FasL and TNFR2 mediated antitumor apoptosis in solid tumors. BMC Cancer 2016; 16:545. [PMID: 27464624 PMCID: PMC4964087 DOI: 10.1186/s12885-016-2608-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 07/25/2016] [Indexed: 01/10/2023] Open
Abstract
Background Directly targeting therapeutic suicide gene to a solid tumor is a hopeful approach for cancer gene therapy. Treatment of a solid tumor by an effective vector for a suicide gene remains a challenge. Given the lack of effective treatments, we constructed a bifidobacterial recombinant thymidine kinase (BF-rTK) -ganciclovir (GCV) targeting system (BKV) to meet this requirement and to explore antitumor mechanisms. Methods Bifidobacterium (BF) or BF-rTK was injected intratumorally with or without ganciclovir in a human colo320 intestinal xenograft tumor model. The tumor tissues were analyzed using apoptosis antibody arrays, real time PCR and western blot. The colo320 cell was analyzed by the gene silencing method. Autophagy and necroptosis were also detected in colo320 cell. Meanwhile, three human digestive system xenograft tumor models (colorectal cancer colo320, gastric cancer MKN-45 and liver cancer SSMC-7721) and a breast cancer (MDA-MB-231) model were employed to validate the universality of BF-rTK + GCV in solid tumor gene therapy. The survival rate was evaluated in three human cancer models after the BF-rTK + GCV intratumor treatment. The analysis of inflammatory markers (TNF-α) in tumor indicated that BF-rTK + GCV significantly inhibited TNF-α expression. Results The results suggested that BF-rTK + GCV induced tumor apoptosis without autophagy and necroptosis occurrence. The apoptosis was transduced by multiple signaling pathways mediated by FasL and TNFR2 and mainly activated the mitochondrial control of apoptosis via Bid and Bim, which was rescued by silencing Bid or/and Bim. However, BF + GCV only induced apoptosis via Fas/FasL signal pathway accompanied with increased P53 expression. We further found that BF-rTK + GCV inhibited the expression of the inflammatory maker of TNF-α. However, BF-rTK + GCV did not result in necroptosis and autophagy. Conclusions BF-rTK + GCV induced tumor apoptosis mediated by FasL and TNFR2 through the mitochondrial control of apoptosis via Bid and Bim without inducing necroptosis and autophagy. Furthermore, BF-rTK + GCV showed to repress the inflammation of tumor through downregulating TNF-α expression. Survival analysis results of multiple cancer models confirmed that BF-rTK + GCV system has a wide field of application in solid tumor gene therapy.
Collapse
Affiliation(s)
- Changdong Wang
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| | - Yongping Ma
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China.
| | - Qiongwen Hu
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| | - Tingting Xie
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| | - Jiayan Wu
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| | - Fan Zeng
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| | - Fangzhou Song
- Department of Biochemistry & Molecular Biology, Molecular Medicine & Cancer Research Center, Chongqing Medical University, Yuzhong District, Yi XueYuan Road, No 1, Chongqing, 400016, People's Republic of China
| |
Collapse
|
17
|
Abstract
Antibiotics are by far the most common medications prescribed for children. Recent epidemiological data suggests an association between early antibiotic use and disease phenotypes in adulthood. Antibiotic use during infancy induces imbalances in gut microbiota, called dysbiosis. The gut microbiome's responses to antibiotics and its potential link to disease development are especially complex to study in the changing infant gut. Here, we synthesize current knowledge linking antibiotics, dysbiosis, and disease and propose a framework for studying antibiotic-related dysbiosis in children. We recommend future studies into the microbiome-mediated effects of antibiotics focused on four types of dysbiosis: loss of keystone taxa, loss of diversity, shifts in metabolic capacity, and blooms of pathogens. Establishment of a large and diverse baseline cohort to define healthy infant microbiome development is essential to advancing diagnosis, interpretation, and eventual treatment of pediatric dysbiosis. This approach will also help provide evidence-based recommendations for antibiotic usage in infancy.
Collapse
Affiliation(s)
- Pajau Vangay
- Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tonya Ward
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Jeffrey S Gerber
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Dan Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA; Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
18
|
Varankovich NV, Nickerson MT, Korber DR. Probiotic-based strategies for therapeutic and prophylactic use against multiple gastrointestinal diseases. Front Microbiol 2015; 6:685. [PMID: 26236287 PMCID: PMC4500982 DOI: 10.3389/fmicb.2015.00685] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 06/22/2015] [Indexed: 12/18/2022] Open
Abstract
Probiotic bacteria offer a number of potential health benefits when administered in sufficient amounts that in part include reducing the number of harmful organisms in the intestine, producing antimicrobial substances and stimulating the body's immune response. However, precisely elucidating the probiotic effect of a specific bacterium has been challenging due to the complexity of the gut's microbial ecosystem and a lack of definitive means for its characterization. This review provides an overview of widely used and recently described probiotics, their impact on the human's gut microflora as a preventative treatment of disease, human/animal models being used to help show efficacy, and discusses the potential use of probiotics in gastrointestinal diseases associated with antibiotic administration.
Collapse
Affiliation(s)
| | | | - Darren R. Korber
- Department of Food and Bioproduct Sciences, University of Saskatchewan, SaskatoonSK, Canada
| |
Collapse
|
19
|
Madej J, Stefaniak T, Bednarczyk M. Effect of in ovo-delivered prebiotics and synbiotics on lymphoid-organs’ morphology in chickens. Poult Sci 2015; 94:1209-19. [DOI: 10.3382/ps/pev076] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2015] [Indexed: 11/20/2022] Open
|
20
|
Williams C, Walton G, Jiang L, Plummer S, Garaiova I, Gibson G. Comparative Analysis of Intestinal Tract Models. Annu Rev Food Sci Technol 2015; 6:329-50. [PMID: 25705934 DOI: 10.1146/annurev-food-022814-015429] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- C.F. Williams
- Cultech, Baglan Industrial Park, Port Talbot, West Glamorgan SA12 7BZ, United Kingdom; , ,
| | - G.E. Walton
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading RG6 6AP, United Kingdom;
| | - L. Jiang
- Cultech, Baglan Industrial Park, Port Talbot, West Glamorgan SA12 7BZ, United Kingdom; , ,
| | - S. Plummer
- Cultech, Baglan Industrial Park, Port Talbot, West Glamorgan SA12 7BZ, United Kingdom; , ,
| | - I. Garaiova
- Cultech, Baglan Industrial Park, Port Talbot, West Glamorgan SA12 7BZ, United Kingdom; , ,
| | - G.R. Gibson
- Department of Food and Nutritional Sciences, School of Chemistry, Food and Pharmacy, University of Reading, Reading RG6 6AP, United Kingdom;
| |
Collapse
|
21
|
Cooper DM. The presence and management of contaminants in non-certified, agriculturally sourced food items used as enrichment for laboratory animals. Lab Anim (NY) 2015; 44:60-4. [PMID: 25602396 DOI: 10.1038/laban.664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/08/2014] [Indexed: 10/24/2022]
Abstract
One enrichment strategy for laboratory animals is the provision of food variety and foraging opportunities. Fresh agricultural items, including produce or packaged human food items, provide variation in palatability, texture and complexity and can therefore be used as enrichment for lab animals. But concerns are often raised that these food items might sometimes carry contaminants that could affect research subjects and confound experimental results. The author discusses the potential for agriculturally sourced foods used as enrichment for lab animals to be contaminated with mycotoxins, microorganisms and pesticide residues and the effects these contaminants might have on lab animals. He also suggests strategies for reducing the risk of contamination.
Collapse
|
22
|
Hallam MC, Barile D, Meyrand M, German JB, Reimer RA. Maternal high-protein or high-prebiotic-fiber diets affect maternal milk composition and gut microbiota in rat dams and their offspring. Obesity (Silver Spring) 2014; 22:2344-51. [PMID: 25056822 DOI: 10.1002/oby.20849] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 07/09/2014] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Maternal gut microbiota and milk composition could modify offspring microbiota and therefore disease susceptibility. The effect of maternal high-protein (HP) or prebiotic diets on maternal milk composition and gut microbiota in rat dams and offspring was examined. METHODS Wistar rat dams were fed a control, HP (40% wt/wt), or high-prebiotic-fiber (21.6% wt/wt) (HF) diet throughout pregnancy and lactation. Pups were challenged with a high-fat/sucrose diet from 14.5 to 22.5 weeks of age. Dam milk was analyzed for fat, protein, and oligosaccharides (OS). Fecal microbiota was analyzed in dams at parturition and 2 weeks post-partum and in offspring at 5 and 22 weeks along with cecal digesta at termination. RESULTS Maternal milk differed only in OS content, each diet group being distinguishable. HF1 and HP1 offspring had decreased plasma lipopolysaccharide compared with C1. Offspring sex, maternal diet, and time (5 weeks vs. 22 weeks of age) affected the microbial groups examined. Bifidobacteria was higher in HF dams and offspring. CONCLUSIONS Increasing protein or fiber content in maternal diet during pregnancy and lactation modifies milk OS content and gut microbiota of dams which may influence establishment of gut microbiota in offspring.
Collapse
Affiliation(s)
- Megan C Hallam
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | |
Collapse
|
23
|
Hoeft B, Eggersdorfer M, Heck S. Quality and safety aspects of food products addressing the needs of pregnant women and infants. ANNALS OF NUTRITION AND METABOLISM 2014; 65:29-33. [PMID: 25227726 DOI: 10.1159/000365796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Food safety is a primary concern for pregnant women and infants as the immune system is weakened during pregnancy and not developed enough in infants, which makes them especially vulnerable to suffering from the negative effects of nonquality food products. However, food contaminations not only affect an individual's health but also a country's economic development, social harmony, food trade and even politics, as seen during the Chinese infant formula crisis in 2008. Thus, quality control is crucial in the production processes in order to have safe food products on the market. But quality control alone is not enough: manufacturers must embrace quality beyond classic in-process parameters and perform a final microbiological analysis at the end of the production process. This requires a clear and trustworthy approach to quality and safety and the involvement of all stakeholders from industry, government and academia over policy makers to consumers. This paper provides an introductory context for current quality management systems and gives real-life examples of challenges that manufacturers face during quality management and control throughout the production process.
Collapse
Affiliation(s)
- Birgit Hoeft
- DSM Nutritional Products Ltd., Kaiseraugst, Switzerland
| | | | | |
Collapse
|
24
|
Bifidobacteria-host interactions--an update on colonisation factors. BIOMED RESEARCH INTERNATIONAL 2014; 2014:960826. [PMID: 25295282 PMCID: PMC4177770 DOI: 10.1155/2014/960826] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/20/2014] [Accepted: 08/20/2014] [Indexed: 01/10/2023]
Abstract
Bifidobacteria are one of the predominant bacterial groups of the human intestinal microbiota and have important functional properties making them interesting for the food and dairy industries. Numerous in vitro and preclinical studies have shown beneficial effects of particular bifidobacterial strains or strain combinations on various health parameters of their hosts. This indicates the potential of bifidobacteria in alternative or supplementary therapeutic approaches in a number of diseased states. Based on these observations, bifidobacteria have attracted considerable interest by the food, dairy, and pharmaceutical industries and they are widely used as so-called probiotics. As a consequence of the rapidly increasing number of available bifidobacterial genome sequences and their analysis, there has been substantial progress in the identification of bifidobacterial structures involved in colonisation of and interaction with the host. With the present review, we aim to provide an update on the current knowledge on the mechanisms by which bifidobacteria colonise their hosts and exert health promoting effects.
Collapse
|
25
|
Ray D, Alpini G, Glaser S. Probiotic Bifidobacterium species: potential beneficial effects in diarrheal disorders. Focus on "Probiotic Bifidobacterium species stimulate human SLC26A3 gene function and expression in intestinal epithelial cells". Am J Physiol Cell Physiol 2014; 307:C1081-3. [PMID: 25209264 DOI: 10.1152/ajpcell.00300.2014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Debolina Ray
- Division of Gastroenterology, Department of Medicine, Texas A & M University Health Science Center, Temple, Texas
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White Healthcare, Temple, Texas; and Division of Gastroenterology, Department of Medicine, Texas A & M University Health Science Center, Temple, Texas
| | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, Texas; Scott & White Digestive Disease Research Center, Scott & White Healthcare, Temple, Texas; and Division of Gastroenterology, Department of Medicine, Texas A & M University Health Science Center, Temple, Texas
| |
Collapse
|
26
|
Umenai T, Shime N, Asahara T, Nomoto K, Itoi T. A pilot study of Bifidobacterium breve in neonates undergoing surgery for congenital heart disease. J Intensive Care 2014; 2:36. [PMID: 25960878 PMCID: PMC4424709 DOI: 10.1186/2052-0492-2-36] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 05/23/2014] [Indexed: 02/07/2023] Open
Abstract
Background Probiotics have currently been widely used in patients undergoing various types of surgeries and improved their clinical outcomes, while data in pediatric cardiac surgery have been lacking. We investigated the safety and effects on the intestinal microbiota of the probiotic Bifidobacterium breve in neonates undergoing surgery for congenital heart disease. Methods This pilot, randomized study was performed in a single-center, university hospital-based pediatric intensive care unit (PICU). Twenty-one neonates undergoing surgery for congenital heart disease at >7 days after birth were randomly allocated to two groups: group A received 3 × 109 colony-forming units (CFU)/day of enteral B. breve strain Yakult (BBG-01), which was started 1 week before and terminated 1 week after surgery (n = 10), and group B did not receive BBG-01 (n = 11). Results The characteristics of the patients were similar in both groups. The postoperative days until fulfillment of the criteria for discharge from the PICU tended to be fewer in group A (8 [7–8] days) than in group B (9 [8–14] days) (p = 0.10). Likewise, the postoperative days to enteral nutrition or achievement of caloric goal tended to be fewer in group A than in group B. The Bifidobacterium in fecal samples after initiating BBG-01 in group A were significantly higher in number than that in group B. Enterobacteriaceae were significantly fewer in group A than in group B immediately (7.0 [3.9–7.7] vs. 8.5 [8.0–9.1] log10 cells/g) and 1 week (7.7 [7.0–8.1] vs. 9.3 [8.6–9.5] log10 cells/g) after surgery (p < 0.05 for both comparisons). The number of Pseudomonas after 1 week was significantly lower in group A than in group B (p = 0.04). The concentrations of total organic and acetic acids were also significantly higher in group A than in group B. The postoperative course was uncomplicated and all neonates were discharged alive from the PICU. Conclusions The perioperative administration of a probiotic to neonates undergoing surgery for congenital heart disease was safe and significantly improved their intestinal environment. The positive effects of this treatment on clinically significant outcomes remain to be investigated.
Collapse
Affiliation(s)
- Takako Umenai
- Department of Anesthesiology, Shimada Hospital, Osaka 583-0875, Japan.,Department of Anesthesiology and Intensive Care, Postgraduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Nobuaki Shime
- Department of Anesthesiology and Intensive Care, Postgraduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.,Department of Emergency and Critical Care Medicine, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Takashi Asahara
- Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Koji Nomoto
- Yakult Central Institute for Microbiological Research, Tokyo 186-8650, Japan
| | - Toshiyuki Itoi
- Department of Pediatric Cardiology and Nephrology, Postgraduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
27
|
Yang LC, Lu TJ, Lin WC. The prebiotic arabinogalactan of Anoectochilus formosanus prevents ovariectomy-induced osteoporosis in mice. J Funct Foods 2013. [DOI: 10.1016/j.jff.2013.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
28
|
Mego M, Holec V, Drgona L, Hainova K, Ciernikova S, Zajac V. Probiotic bacteria in cancer patients undergoing chemotherapy and radiation therapy. Complement Ther Med 2013; 21:712-23. [PMID: 24280481 DOI: 10.1016/j.ctim.2013.08.018] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 08/01/2013] [Accepted: 08/23/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Probiotics are live microorganisms, which as drugs or food supplements help to maintain health beneficial microbial balance in the digestive tract of a human or other host. Probiotics by their properties may help strengthen homeostasis and thus reduce side effects associated with cancer treatment. Experimental evidence suggests that probiotics might have beneficial effect on the toxicity of anticancer therapy. METHODS A computer-based literature search was carried out using PubMed (keywords: "probiotic" and "lactic acid bacteria" in association with the search terms "cancer" or "oncology" or "chemotherapy" or "radiation"); data reported at international meetings were included. RESULTS Probiotics might have beneficial effects on some aspects of toxicity related to anticancer treatment especially radiation therapy. However, reported trials vary in utilized probiotic strains, dose of probiotics and vast majority of them are small trials with substantial risk of bias. Despite limited data, it seems that probiotic bacteria as live microorganisms could be safely administered even in the setting of neutropenia. CONCLUSIONS Current evidence supporting probiotic use as adjunctive therapy to anticancer treatment is limited, especially in cancer patients treated with chemotherapy. Well designed clinical trials are needed to find true role of probiotics in oncology.
Collapse
Affiliation(s)
- Michal Mego
- Department of Medical Oncology, Faculty of Medicine, Comenius University and National Cancer Institute, Klenova 1, 833 10 Bratislava, Slovak Republic.
| | | | | | | | | | | |
Collapse
|
29
|
Saad N, Delattre C, Urdaci M, Schmitter J, Bressollier P. An overview of the last advances in probiotic and prebiotic field. Lebensm Wiss Technol 2013. [DOI: 10.1016/j.lwt.2012.05.014] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Di Bartolomeo F, Startek JB, Van den Ende W. Prebiotics to fight diseases: reality or fiction? Phytother Res 2012; 27:1457-73. [PMID: 23280537 DOI: 10.1002/ptr.4901] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Revised: 11/10/2012] [Accepted: 11/12/2012] [Indexed: 12/18/2022]
Abstract
Bacteria living in the gastrointestinal tract are crucial for human health and disease occurrence. Increasing the beneficial intestinal microflora by consumption of prebiotics, which are 'functional foods', could be an elegant way to limit the number and incidence of disorders and to recover from dysbiosis or antibiotic treatments. This review focuses on the short-chain low-digestible carbohydrates (LDCs) which are metabolized by gut microbiota serving as energy source, immune system enhancers or facilitators of mineral uptake. Intake of foods containing LDCs can improve the state of health and may prevent diseases as for example certain forms of cancer. Given the large number of different molecules belonging to LDCs, we focused our attention on fructans (inulin, fructo-oligosaccharides), galacto-oligosaccharides and resistant starches and their therapeutic and protective applications. Evidence is accumulating that LDCs can inhibit bacterial and viral infections by modulating host defense responses and by changing the interactions between pathogenic and beneficial bacteria. Animal studies and studies on small groups of human subjects suggest that LDCs might help to counteract colorectal cancer, diabetes and metabolic syndrome. The action mechanisms of LDCs in the human body might be broader than originally thought, perhaps also including reactive oxygen species scavenging and signaling events.
Collapse
Affiliation(s)
- F Di Bartolomeo
- Laboratory of Molecular Plant Biology and Leuven Food Science and Nutrition Research Centre (LFoRCe), Kasteelpark Arenberg 31, 3001, Leuven, Belgium; University of the Studies of Molise - Department of Agriculture Food and Environment (D.A.A.A.) - Terzo edificio polifunzionale, Via de Sanctis, 86100, Campobasso, Italy
| | | | | |
Collapse
|
31
|
Thum C, Cookson AL, Otter DE, McNabb WC, Hodgkinson AJ, Dyer J, Roy NC. Can nutritional modulation of maternal intestinal microbiota influence the development of the infant gastrointestinal tract? J Nutr 2012; 142:1921-8. [PMID: 22990463 DOI: 10.3945/jn.112.166231] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The gastrointestinal microbiota plays an important role in maintaining host health by preventing the colonization of pathogens, fermenting dietary compounds, and maintaining normal mucosal immunity. Particularly in early life, the composition of the microbiota profoundly influences the development and maturation of the gastrointestinal tract (GIT) mucosa, which may affect health in later life. Therefore, strategies to manipulate the microbiota during infancy may prevent the development of some diseases later in adult life. Earlier research suggested that term fetuses are sterile and that the initial bacterial colonization of the newborn GIT occurs only after the baby transits through the birth canal. However, recent studies have demonstrated that the colonization and/or contact of the fetus with the maternal GIT microbiota may start in utero. After vaginal birth, the colonization of the neonate GIT continues through contact with maternal feces and vaginal bacteria, leading to a relatively simple microbial community that is influenced by feeding type (breast vs. formula feeding). Maternal GIT microbiota, vaginal microbiota, and breast milk composition are influenced by maternal diet. Alterations of the maternal GIT microbiota composition via supplementation with probiotics and prebiotics have been shown; however, transfer of these benefits to the offspring remains to be demonstrated. This review focuses on the influence of maternal GIT microbiota during the pre- and postpartum periods on the colonization of the infant GIT. In particular, it examines the manipulation of the maternal GIT microbiota composition through the use of probiotics and/or prebiotics and subsequent consequences for the health of the offspring.
Collapse
Affiliation(s)
- Caroline Thum
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch Grasslands, Palmerston North, New Zealand
| | | | | | | | | | | | | |
Collapse
|
32
|
Tomoda K, Kubo K, Asahara T, Nomoto K, Nishii Y, Yamamoto Y, Yoshikawa M, Kimura H. Suppressed anti-oxidant capacity due to a cellulose-free diet declines further by cigarette smoke in mice. J Toxicol Sci 2012; 37:575-85. [PMID: 22687997 DOI: 10.2131/jts.37.575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Dietary fiber, maintaining the gut environment, contributes to better lung function among smokers. This study was aimed to investigate the role of dietary fiber on the anti-oxidant capacity and gut environment during exposure to cigarette smoke. The anti-oxidant capacity as well as caecal levels of organic acids and population of micro-flora in the gut was measured after 4 months' exposure to cigarette smoke in mice (C57BL/6NcrSlc) fed with a cellulose-free diet. Animals were divided into control diet (AIN-93G)/non-smoke, cellulose-free diet/non-smoke, control diet/smoke, and cellulose-free diet/smoke groups. The anti-oxidant capacity in plasma was significantly suppressed by the cellulose-free diet in the non-smoke exposed mice. The suppression in the anti-oxidant capacity further declined following exposure to cigarette smoke. Both these changes in the anti-oxidant capacity were accompanied with changes in some organic acids levels in caecal contents. The anti-oxidant activity was significantly inversely correlated to succinic acid / acetic acid levels balance in caecal contents. In conclusion the cellulose-free diet suppressed the anti-oxidant capacity in mice, and the suppression further decreased by exposure to cigarette smoke. These changes in the anti-oxidant capacity may be related with changes in the gut environment.
Collapse
Affiliation(s)
- Koichi Tomoda
- Second Department of Internal Medicine, Nara Medical University, Nara Japan.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Gibson GR, Probert HM, Loo JV, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev 2012; 17:259-75. [PMID: 19079930 DOI: 10.1079/nrr200479] [Citation(s) in RCA: 1372] [Impact Index Per Article: 105.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prebiotics are non-digestible (by the host) food ingredients that have a beneficial effect through their selective metabolism in the intestinal tract. Key to this is the specificity of microbial changes. The present paper reviews the concept in terms of three criteria: (a) resistance to gastric acidity, hydrolysis by mammalian enzymes and gastrointestinal absorption; (b) fermentation by intestinal microflora; (c) selective stimulation of the growth and/or activity of intestinal bacteria associated with health and wellbeing. The conclusion is that prebiotics that currently fulfil these three criteria are fructo-oligosaccharides, galacto-oligosaccharides and lactulose, although promise does exist with several other dietary carbohydrates. Given the range of food vehicles that may be fortified by prebiotics, their ability to confer positive microflora changes and the health aspects that may accrue, it is important that robust technologies to assay functionality are used. This would include a molecular-based approach to determine flora changes. The future use of prebiotics may allow species-level changes in the microbiota, an extrapolation into genera other than the bifidobacteria and lactobacilli, and allow preferential use in disease-prone areas of the body.
Collapse
Affiliation(s)
- Glenn R Gibson
- Food Microbial Sciences Unit, School of Food Biosciences, The University of Reading, Reading, UK
| | | | | | | | | |
Collapse
|
34
|
Prebiotics in foods. Curr Opin Biotechnol 2012; 23:187-91. [DOI: 10.1016/j.copbio.2011.12.028] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 12/12/2011] [Accepted: 12/21/2011] [Indexed: 01/04/2023]
|
35
|
|
36
|
|
37
|
Abstract
Probiotic bacteria are increasingly incorporated into food products intended to confer health benefits in the human gut and beyond. Little is known about how the food matrix and product formulation impacts probiotic functionality, even though such information is essential to scientific understanding and regulatory substantiation of health benefits. The food format has the potential to affect probiotic survival, physiology, and potentially efficacy, but few comparative studies in humans have been conducted. Human studies should account for the effects of the food base on human health and the bioactive components present in the foods that may augment or diminish interactions of the probiotic with the human host. Some studies show that food ingredients such as prebiotics and milk components can improve probiotic survival during the shelf life of foods, which may enhance probiotic efficacy through increased dose effects. Furthermore, there are indications that synbiotic products are more effective than either probiotics or prebiotics alone. Identification of probiotic adaptations to the food and gut environments holds promise for determining the specific cell components and potential bacterial-food interactions necessary for health benefits and determining how these factors are affected by changes in food formulation and host diet. These studies, combined with controlled human studies, are important future research activities for advancing this field.
Collapse
|
38
|
Singh AP, Preet S, Rishi P. Augmentation of antimicrobial activity of conventional antibiotics by cell-free extract of L. plantarum. J Antibiot (Tokyo) 2011; 64:795-8. [PMID: 22008700 DOI: 10.1038/ja.2011.92] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Aman Preet Singh
- Department of Microbiology, Basic Medical Sciences Building, Panjab University, Chandigarh, India
| | | | | |
Collapse
|
39
|
Figueroa-González I, Quijano G, Ramírez G, Cruz-Guerrero A. Probiotics and prebiotics--perspectives and challenges. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2011; 91:1341-1348. [PMID: 21445871 DOI: 10.1002/jsfa.4367] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 02/04/2011] [Accepted: 02/04/2011] [Indexed: 05/30/2023]
Abstract
Owing to their health benefits, probiotics and prebiotics are nowadays widely used in yogurts and fermented milks, which are leader products of functional foods worldwide. The world market for functional foods has grown rapidly in the last three decades, with an estimated size in 2003 of ca US$ 33 billion, while the European market estimation exceeded US$ 2 billion in the same year. However, the production of probiotics and prebiotics at industrial scale faces several challenges, including the search for economical and abundant raw materials for prebiotic production, the low-cost production of probiotics and the improvement of probiotic viability after storage or during the manufacturing process of the functional food. In this review, functional foods based on probiotics and prebiotics are introduced as a key biotechnological field with tremendous potential for innovation. A concise state of the art addressing the fundamentals and challenges for the development of new probiotic- and prebiotic-based foods is presented, the niches for future research being clearly identified and discussed.
Collapse
Affiliation(s)
- Ivonne Figueroa-González
- Departamento de Biotecnología, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco No. 186 Col. Vicentina, CP 09340, Distrito Federal, Mexico
| | | | | | | |
Collapse
|
40
|
Effect of the continuous intake of probiotic-fermented milk containing Lactobacillus casei strain Shirota on fever in a mass outbreak of norovirus gastroenteritis and the faecal microflora in a health service facility for the aged. Br J Nutr 2011; 106:549-56. [DOI: 10.1017/s000711451100064x] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
For conducting effective risk management in long-stay elderly people at a health service facility, we performed an open case-controlled study to evaluate the effect of the intake of probiotic-fermented milk containing Lactobacillus casei strain Shirota (LcS-fermented milk) on norovirus gastroenteritis occurring in the winter season during the intake period. A total of seventy-seven elderly people (mean age 84 years) were enrolled in the study. During a 1-month period, there was no significant difference in the incidence of norovirus gastroenteritis between the LcS-fermented milk-administered (n 39) and the non-administered (n 38) groups; however, the mean duration of fever of >37°C after the onset of gastroenteritis was 1·5 (sd 1·7) d in the former and 2·9 (sd 2·3) d in the latter group, showing a significant shortening in the former group (P < 0·05). RT-quantitative PCR analysis targeting ribosomal RNA showed both Bifidobacterium and Lactobacillus to be significantly dominant, whereas Enterobacteriaceae decreased in faecal samples from the administered group (n 10, mean age 83 years), with a significant increase in faecal acetic acid concentration. Continuous intake of LcS-fermented milk could positively contribute to the alleviation of fever caused by norovirus gastroenteritis by correcting the imbalance of the intestinal microflora peculiar to the elderly, although such consumption could not protect them from the disease.
Collapse
|
41
|
Tomoda K, Kubo K, Asahara T, Andoh A, Nomoto K, Nishii Y, Yamamoto Y, Yoshikawa M, Kimura H. Cigarette smoke decreases organic acids levels and population of bifidobacterium in the caecum of rats. J Toxicol Sci 2011; 36:261-6. [DOI: 10.2131/jts.36.261] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
| | - Kaoru Kubo
- Laboratory Animal Research Center, Nara Medical University
| | | | - Akira Andoh
- Division of Mucosal Immunology, Graduate School of Medicine, Shiga University of Medical Science
| | - Koji Nomoto
- Yakult Central Institute for Microbiological Research
| | | | | | | | | |
Collapse
|
42
|
Asahara T, Shimizu K, Takada T, Kado S, Yuki N, Morotomi M, Tanaka R, Nomoto K. Protective effect of Lactobacillus casei strain Shirota against lethal infection with multi-drug resistant Salmonella enterica serovar Typhimurium DT104 in mice. J Appl Microbiol 2010; 110:163-73. [PMID: 21059159 DOI: 10.1111/j.1365-2672.2010.04884.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
AIMS The anti-infectious activity of lactobacilli against multi-drug resistant Salmonella enterica serovar Typhimurium DT104 (DT104) was examined in a murine model of an opportunistic antibiotic-induced infection. METHODS AND RESULTS Explosive intestinal growth and subsequent lethal extra-intestinal translocation after oral infection with DT104 during fosfomycin (FOM) administration was significantly inhibited by continuous oral administration of Lactobacillus casei strain Shirota (LcS), which is naturally resistant to FOM, at a dose of 10(8) colony-forming units per mouse daily to mice. Comparison of the anti-Salmonella activity of several Lactobacillus type strains with natural resistance to FOM revealed that Lactobacillus brevis ATCC 14869(T) , Lactobacillus plantarum ATCC 14917(T) , Lactobacillus reuteri JCM 1112(T) , Lactobacillus rhamnosus ATCC 7469(T) and Lactobacillus salivarius ATCC 11741(T) conferred no activity even when they obtained the high population levels almost similar to those of the effective strains such as LcS, Lact. casei ATCC 334(T) and Lactobacillus zeae ATCC 15820(T) . The increase in concentration of organic acids and maintenance of the lower pH in the intestine because of Lactobacillus colonization were correlated with the anti-infectious activity. Moreover, heat-killed LcS was not protective against the infection, suggesting that the metabolic activity of lactobacilli is important for the anti-infectious activity. CONCLUSION These results suggest that certain lactobacilli in combination with antibiotics may be useful for prophylaxis against opportunistic intestinal infections by multi-drug resistant pathogens, such as DT104. SIGNIFICANCE AND IMPACT OF THE STUDY Antibiotics such as FOM disrupt the metabolic activity of the intestinal microbiota that produce organic acids, and that only probiotic strains that are metabolically active in vivo should be selected to prevent intestinal infection when used clinically in combination with certain antibiotics.
Collapse
Affiliation(s)
- T Asahara
- Yakult Central Institute for Microbiological Research, Kunitachi, Tokyo, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Petersen A, Bergström A, Andersen J, Hansen M, Lahtinen S, Wilcks A, Licht T. Analysis of the intestinal microbiota of oligosaccharide fed mice exhibiting reduced resistance to Salmonella infection. Benef Microbes 2010; 1:271-81. [DOI: 10.3920/bm2010.0016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Certain indigestible carbohydrates, known as prebiotics, are claimed to be beneficial for gut health through a selective stimulation of certain gut microbes including bifidobacteria. However, stimulation of such microbes does not necessarily imply a preventive effect against pathogen infection. We recently demonstrated a reduced resistance to Salmonella infection in mice fed diets containing fructo-oligosaccharides (FOS) or xylo-oligosaccharides (XOS). In the present study, faecal and caecal samples from the same mice were analysed in order to study microbial changes potentially explaining the observed effects on the pathogenesis of Salmonella. Denaturing gradient gel electrophoresis revealed that the microbiota in faecal samples from mice fed FOS or XOS were different from faecal samples collected before the feeding trial as well as from faecal profiles generated from control animals. This difference was not seen for caecal profiles. Further analysis of faecal samples by real-time PCR demonstrated a significant increase in the Bacteroidetes phylum, the Bacteroides fragilis group and in Bifidobacterium spp. in mice fed FOS or XOS. The observed bifidogenic effect was more pronounced for XOS than for FOS. The Firmicutes phylum and the Clostridium coccoides group were reduced by both FOS and XOS. Surprisingly, no significant differences were detected between faecal samples collected before and after pathogen challenge in any of the groups. Furthermore, no effect of diets on caecal concentrations of short-chain fatty acids was recorded. In conclusion, diets supplemented with FOS or XOS induced a number of microbial changes in the faecal microbiota of mice. The observed effects of XOS were qualitatively similar to those of FOS, but the most prominent bifidogenic effect was seen for XOS. An increased level of bifidobacteria is thus not in itself preventive against Salmonella infection, since the same XOS or FOS-fed mice were previously reported to be more severely affected by Salmonella than control animals.
Collapse
Affiliation(s)
- A. Petersen
- National Food Institute, Division of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| | - A. Bergström
- National Food Institute, Division of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| | - J. Andersen
- National Food Institute, Division of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| | - M. Hansen
- National Food Institute, Division of Toxicology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| | - S. Lahtinen
- Danisco Health & Nutrition, Sokeritehtaantie 20, 02460 Kantvik, Finland
| | - A. Wilcks
- National Food Institute, Division of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| | - T. Licht
- National Food Institute, Division of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, 2860 Soeborg, Denmark
| |
Collapse
|
44
|
Lkhagvadorj E, Nagata S, Wada M, Bian L, Wang C, Chiba Y, Yamashiro Y, Shimizu T, Asahara T, Nomoto K. Anti-infectious activity of synbiotics in a novel mouse model of methicillin-resistant Staphylococcus aureus infection. Microbiol Immunol 2010; 54:265-75. [DOI: 10.1111/j.1348-0421.2010.00224.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Eradication of the commensal intestinal microflora by oral antimicrobials interferes with the host response to lipopolysaccharide. Eur J Clin Microbiol Infect Dis 2010; 29:633-41. [PMID: 20300949 DOI: 10.1007/s10096-010-0905-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2009] [Accepted: 02/20/2010] [Indexed: 01/04/2023]
Abstract
The host components and commensal microorganisms of the intestinal microenvironment play roles in the development and maintenance of the host defence. Recent observations have suggested that toll-like receptors (TLRs) are involved in the recognition of innate immunity against intestinal microbes. However, little is known regarding the role of TLR in the maintenance of systemic host defence by intestinal microorganisms. We studied the expression and function of TLR4 and TLR2 on alveolar and peritoneal macrophages in mice after 3 weeks of oral administration of streptomycin and cefotaxime. After active treatment, the intestinal microorganisms were nearly completely eradicated, and the surface expression of TLR4 and TLR2 on the peritoneal macrophages was prominently downregulated. When the actively treated mice were challenged with lipopolysaccharide (LPS), a TLR4 ligand, the host response was markedly impaired. Our results suggest that the oral administration of antimicrobials downregulates the expression of surface TLR on the peritoneal macrophages and modulates the host immune responses against LPS by modifying the intestinal environment.
Collapse
|
46
|
Asahara T. Preventive Effect of Probiotic Bifidobacteria against Shiga Toxin-Producing <i>Escherichia Coli</i> and <i>Salmonella</i> Infections. Biosci Microflora 2010. [DOI: 10.12938/bifidus.29.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
47
|
Petersen A, Heegaard PMH, Pedersen AL, Andersen JB, Sørensen RB, Frøkiaer H, Lahtinen SJ, Ouwehand AC, Poulsen M, Licht TR. Some putative prebiotics increase the severity of Salmonella enterica serovar Typhimurium infection in mice. BMC Microbiol 2009; 9:245. [PMID: 19948011 PMCID: PMC2789089 DOI: 10.1186/1471-2180-9-245] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 11/30/2009] [Indexed: 11/23/2022] Open
Abstract
Background Prebiotics are non-digestible food ingredients believed to beneficially affect host health by selectively stimulating the growth of the beneficial bacteria residing in the gut. Such beneficial bacteria have been reported to protect against pathogenic infections. However, contradicting results on prevention of Salmonella infections with prebiotics have been published. The aim of the present study was to examine whether S. Typhimurium SL1344 infection in mice could be prevented by administration of dietary carbohydrates with different structures and digestibility profiles. BALB/c mice were fed a diet containing 10% of either of the following carbohydrates: inulin, fructo-oligosaccharide, xylo-oligosaccharide, galacto-oligosaccharide, apple pectin, polydextrose or beta-glucan for three weeks prior to oral Salmonella challenge (107 CFU) and compared to mice fed a cornstarch-based control diet. Results The mice fed with diets containing fructo-oligosaccharide (FOS) or xylo-oligosaccharide (XOS) had significantly higher (P < 0.01 and P < 0.05) numbers of S. Typhimurium SL1344 in liver, spleen and mesenteric lymph nodes when compared to the mice fed with the cornstarch-based control diet. Significantly increased amounts (P < 0.01) of Salmonella were detected in ileal and fecal contents of mice fed with diets supplemented with apple pectin, however these mice did not show significantly higher numbers of S. Typhimyrium in liver, spleen and lymph nodes than animals from the control group (P < 0.20). The acute-phase protein haptoglobin was a good marker for translocation of S. Typhimurium in mice. In accordance with the increased counts of Salmonella in the organs, serum concentrations of haptoglobin were significantly increased in the mice fed with FOS or XOS (P < 0.001). Caecum weight was increased in the mice fed with FOS (P < 0.01), XOS (P < 0.01), or polydextrose (P < 0.001), and caecal pH was reduced in the mice fed with polydextrose (P < 0.001). In vitro fermentation in monocultures revealed that S. Typhimurium SL1344 is capable of fermenting FOS, beta-glucan and GOS with a corresponding decline in pH. Conclusion Supplementing a cornstarch-based rodent diet with 10% FOS or XOS was found to increase the translocation of S. Typhimurium SL1344 to internal organs in mice, while 10% apple pectin was found to increase the numbers of S. Typhimurium in intestinal content and feces.
Collapse
Affiliation(s)
- Anne Petersen
- The National Food Institute, Department of Microbiology and Risk Assessment, Technical University of Denmark, Moerkhoej Bygade 19, DK-2860 Soeborg, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Watanabe T, Nishio H, Tanigawa T, Yamagami H, Okazaki H, Watanabe K, Tominaga K, Fujiwara Y, Oshitani N, Asahara T, Nomoto K, Higuchi K, Takeuchi K, Arakawa T. Probiotic Lactobacillus casei strain Shirota prevents indomethacin-induced small intestinal injury: involvement of lactic acid. Am J Physiol Gastrointest Liver Physiol 2009; 297:G506-13. [PMID: 19589943 DOI: 10.1152/ajpgi.90553.2008] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Inflammatory responses triggered by activation of the lipopolysaccharide (LPS)/Toll-like receptor (TLR) 4 signaling pathway are a key mechanism in nonsteroidal anti-inflammatory drug-induced enteropathy. The aim of this study was to investigate the probiotic effect of Lactobacillus casei strain Shirota (LcS) on indomethacin-induced small intestinal injury. Rats pretreated with viable LcS or heat-killed LcS once or once daily for a week were administered indomethacin by gavage to induce injury. Anti-inflammatory effects of L-lactic acid (1-15 mM) were evaluated in vitro by use of THP-1 cells. One-week treatment with viable LcS prevented indomethacin-induced intestinal injury with increase in the concentration of lactic acid in small intestinal content and inhibited increases in myeloperoxidase activity and expression of mRNA for tumor necrosis factor-alpha (TNF-alpha) while affecting neither TLR4 expression nor the number of gram-negative bacteria in intestinal content, whereas neither heat-killed LcS nor a single dose of viable LcS inhibited intestinal injury. Prevention of this injury was also observed in rats given l-lactic acid in drinking water. Both L-lactic acid and LcS culture supernatant containing 10 mM lactic acid inhibited NF-kappaB activation and increases in TNF-alpha mRNA expression and TNF-alpha protein secretion in THP-1 cells treated with LPS. Western blot analyses showed that both L-lactic acid and LcS culture supernatants suppressed phosphorylation and degradation of I-kappaB-alpha induced by LPS without affecting expression of TLR4. These findings suggest that LcS exhibits a prophylactic effect on indomethacin-induced enteropathy by suppressing the LPS/TLR4 signaling pathway and that this probiotic effect of LcS may be mediated by L-lactic acid.
Collapse
Affiliation(s)
- Toshio Watanabe
- Dept. of Gastroenterology, Osaka City Univ. Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wada M, Nagata S, Saito M, Shimizu T, Yamashiro Y, Matsuki T, Asahara T, Nomoto K. Effects of the enteral administration of Bifidobacterium breve on patients undergoing chemotherapy for pediatric malignancies. Support Care Cancer 2009; 18:751-9. [PMID: 19685085 DOI: 10.1007/s00520-009-0711-6] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Accepted: 07/20/2009] [Indexed: 12/21/2022]
Abstract
PURPOSE Probiotics are expected to be effective in prophylaxis of infection in cancer patient, since infections in neutropenics are mainly caused by endogenous flora through the intestinal mucosa. However, the experience with the use of probiotics in immunocompromised patients is limited, and precise fecal bacteria analysis has not been reported. The aim of the study was to evaluate the effects of the enteral administration of the probiotic, Bifidobacterium breve strain Yakult, on its ability to prevent infection, fecal micro flora, and intestinal environments in cancer patients on chemotherapy. METHODS A placebo-controlled trial was performed at Juntendo University Hospital. Patients with malignancies admitted for chemotherapy (n = 42) were randomized into two groups receiving probiotic or placebo. The effects on infectious complications, natural killer cells, fecal micro flora, fecal organic acid concentrations, and fecal pH were studied. RESULTS The frequency of fever and the use of intravenous antibiotics were lower in the probiotic group than the placebo group. The probiotic administration enhanced the habitation of anaerobes. Disruption of the intestinal microbiota after chemotherapy such as the increase in the population levels of Enterobacteriaceae was observed at more pronounced manner in the placebo group in comparison to the probiotic group. The concentrations of total organic acids were maintained most of the time at the normal level, which constantly maintained the pH below 7.0 only in the probiotic group. CONCLUSION These data, although based on a limited number of patients and samples, suggest that administration of B. breve strain Yakult could be an effective approach for achieving clinical benefits in immunocompromised hosts by improving their intestinal environments.
Collapse
Affiliation(s)
- Mariko Wada
- Department of Laboratory for Probiotics Research, Juntendo University School of Medicine, Tokyo 113-8421, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Rishi P, Mavi SK, Bharrhan S, Shukla G, Tewari R. Protective efficacy of probiotic alone or in conjunction with a prebiotic in Salmonella-induced liver damage. FEMS Microbiol Ecol 2009; 69:222-30. [PMID: 19496820 DOI: 10.1111/j.1574-6941.2009.00703.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In view of the increasing interest in the bioecological and nutritional control of diseases, use of probiotics alone or in combination with prebiotics (synbiotics) appears as a therapeutic option for various diseases. In this study, an attempt was made to explore the protective potential of Lactobacillus acidophilus as a probiotic, inulin as a prebiotic and both L. acidophilus and inulin as synbiotic against Salmonella-induced liver damage in a murine model. The probiotic, prebiotic and synbiotic supplementation resulted in decreased bacterial translocation in the liver of mice challenged with Salmonella typhimurium and decreased levels of serum aminotransferases, suggesting their protective role against Salmonella infection. Mice supplemented with these preparations before Salmonella challenge also revealed decreased levels of lipid peroxidation, increased levels of superoxide dismutase and glutathione, along with reduced levels of nitric oxide. Thus, bacteriological and biochemical alterations correlated well with the histological evidence. Protection afforded by supplementation with the probiotic alone was found to be more effective. None of the observations was suggestive of the synergistic effect in the synbiotic-supplemented animals. Thus, it is indicated that the probiotic and the prebiotic used in the present study may act by different mechanisms involved in affording protection against Salmonella-induced liver damage.
Collapse
Affiliation(s)
- Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India.
| | | | | | | | | |
Collapse
|