1
|
Li Q, Liu D, Liang M, Zhu Y, Yousaf M, Wu Y. Mechanism of probiotics in the intervention of colorectal cancer: a review. World J Microbiol Biotechnol 2024; 40:306. [PMID: 39160377 DOI: 10.1007/s11274-024-04112-w] [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: 06/24/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The human microbiome interacts with the host mainly in the intestinal lumen, where putrefactive bacteria are suggested to promote colorectal cancer (CRC). In contrast, probiotics and their isolated components and secreted substances, display anti-tumor properties due to their ability to modulate gut microbiota composition, promote apoptosis, enhance immunity, resist oxidation and alter metabolism. Probiotics help to form a solid intestinal barrier against damaging agents via altering the gut microbiota and preventing harmful microbes from colonization. Probiotic strains that specifically target essential proteins involved in the process of apoptosis can overcome CRC resistance to apoptosis. They can increase the production of anti-inflammatory cytokines, essential in preventing carcinogenesis, and eliminate cancer cells by activating T cell-mediated immune responses. There is a clear indication that probiotics optimize the antioxidant system, decrease radical generation, and detect and degrade potential carcinogens. In this review, the pathogenic mechanisms of pathogens in CRC and the recent insights into the mechanism of probiotics in CRC prevention and therapy are discussed to provide a reference for the actual application of probiotics in CRC.
Collapse
Affiliation(s)
- Qinqin Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Dongmei Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Minghua Liang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Yichao Zhu
- Laboratory of Cell Engineering, Research Unit of Cell Death Mechanism, Beijing Institute of Biotechnology, Chinese Academy of Medical Sciences (2021RU008), Beijing, 100071, China
| | - Muhammad Yousaf
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Yaping Wu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| |
Collapse
|
2
|
Hesser LA, Puente AA, Arnold J, Ionescu E, Mirmira A, Talasani N, Lopez J, Maccio-Maretto L, Mimee M, Nagler CR. A synbiotic of Anaerostipes caccae and lactulose prevents and treats food allergy in mice. Cell Host Microbe 2024; 32:1163-1176.e6. [PMID: 38906158 PMCID: PMC11239278 DOI: 10.1016/j.chom.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 03/26/2024] [Accepted: 05/28/2024] [Indexed: 06/23/2024]
Abstract
Depletion of beneficial microbes by modern lifestyle factors correlates with the rising prevalence of food allergies. Re-introduction of allergy-protective bacteria may be an effective treatment strategy. We characterized the fecal microbiota of healthy and food-allergic infants and found that the anaerobe Anaerostipes caccae (A. caccae) was representative of the protective capacity of the healthy microbiota. We isolated a strain of A. caccae from the feces of a healthy infant and identified lactulose as a prebiotic to optimize butyrate production by A. caccae in vitro. Administration of a synbiotic composed of our isolated A. caccae strain and lactulose increased luminal butyrate in gnotobiotic mice colonized with feces from an allergic infant and in antibiotic-treated specific pathogen-free (SPF) mice, and prevented or treated an anaphylactic response to allergen challenge. The synbiotic's efficacy in two models and microbial contexts suggests that it may be a promising approach for the treatment of food allergy.
Collapse
Affiliation(s)
- Lauren A Hesser
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Armando A Puente
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Jack Arnold
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Edward Ionescu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | - Anjali Mirmira
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Nidhi Talasani
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Jacqueline Lopez
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | - Mark Mimee
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA; Committee on Microbiology, The University of Chicago, Chicago, IL, USA
| | - Cathryn R Nagler
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA; Department of Pathology, The University of Chicago, Chicago, IL, USA; Committee on Immunology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Odriozola A, González A, Odriozola I, Álvarez-Herms J, Corbi F. Microbiome-based precision nutrition: Prebiotics, probiotics and postbiotics. ADVANCES IN GENETICS 2024; 111:237-310. [PMID: 38908901 DOI: 10.1016/bs.adgen.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Microorganisms have been used in nutrition and medicine for thousands of years worldwide, long before humanity knew of their existence. It is now known that the gut microbiota plays a key role in regulating inflammatory, metabolic, immune and neurobiological processes. This text discusses the importance of microbiota-based precision nutrition in gut permeability, as well as the main advances and current limitations of traditional probiotics, new-generation probiotics, psychobiotic probiotics with an effect on emotional health, probiotic foods, prebiotics, and postbiotics such as short-chain fatty acids, neurotransmitters and vitamins. The aim is to provide a theoretical context built on current scientific evidence for the practical application of microbiota-based precision nutrition in specific health fields and in improving health, quality of life and physiological performance.
Collapse
Affiliation(s)
- Adrián Odriozola
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Adriana González
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Iñaki Odriozola
- Health Department of Basque Government, Donostia-San Sebastián, Spain
| | - Jesús Álvarez-Herms
- Phymo® Lab, Physiology, and Molecular Laboratory, Collado Hermoso, Segovia, Spain
| | - Francesc Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), Centre de Lleida, Universitat de Lleida (UdL), Lleida, Spain
| |
Collapse
|
4
|
Nayman EI, Schwartz BA, Polmann M, Gumabong AC, Nieuwdorp M, Cickovski T, Mathee K. Differences in gut microbiota between Dutch and South-Asian Surinamese: potential implications for type 2 diabetes mellitus. Sci Rep 2024; 14:4585. [PMID: 38403716 PMCID: PMC10894869 DOI: 10.1038/s41598-024-54769-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
Gut microbiota, or the collection of diverse microorganisms in a specific ecological niche, are known to significantly impact human health. Decreased gut microbiota production of short-chain fatty acids (SCFAs) has been implicated in type 2 diabetes mellitus (T2DM) disease progression. Most microbiome studies focus on ethnic majorities. This study aims to understand how the microbiome differs between an ethnic majority (the Dutch) and minority (the South-Asian Surinamese (SAS)) group with a lower and higher prevalence of T2DM, respectively. Microbiome data from the Healthy Life in an Urban Setting (HELIUS) cohort were used. Two age- and gender-matched groups were compared: the Dutch (n = 41) and SAS (n = 43). Microbial community compositions were generated via DADA2. Metrics of microbial diversity and similarity between groups were computed. Biomarker analyses were performed to determine discriminating taxa. Bacterial co-occurrence networks were constructed to examine ecological patterns. A tight microbiota cluster was observed in the Dutch women, which overlapped with some of the SAS microbiota. The Dutch gut contained a more interconnected microbial ecology, whereas the SAS network was dispersed, i.e., contained fewer inter-taxonomic correlational relationships. Bacteroides caccae, Butyricicoccus, Alistipes putredinis, Coprococcus comes, Odoribacter splanchnicus, and Lachnospira were enriched in the Dutch gut. Haemophilus, Bifidobacterium, and Anaerostipes hadrus discriminated the SAS gut. All but Lachnospira and certain strains of Haemophilus are known to produce SCFAs. The Dutch gut microbiome was distinguished from the SAS by diverse, differentially abundant SCFA-producing taxa with significant cooperation. The dynamic ecology observed in the Dutch was not detected in the SAS. Among several potential gut microbial biomarkers, Haemophilus parainfluenzae likely best characterizes the ethnic minority group, which is more predisposed to T2DM. The higher prevalence of T2DM in the SAS may be associated with the gut dysbiosis observed.
Collapse
Affiliation(s)
- Eric I Nayman
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA.
| | - Brooke A Schwartz
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - Michaela Polmann
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Alayna C Gumabong
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA
| | - Max Nieuwdorp
- Amsterdam Diabetes Center, Department of Internal Medicine, Academic Medical Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Trevor Cickovski
- Bioinformatics Research Group, Knight Foundation School of Computing and Information Sciences, College of Engineering and Computing, Florida International University, Miami, FL, USA.
| | - Kalai Mathee
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Biomolecular Sciences Institute, Florida International University, Miami, FL, USA.
| |
Collapse
|
5
|
Zhao S, Lau R, Zhong Y, Chen MH. Lactate cross-feeding between Bifidobacterium species and Megasphaera indica contributes to butyrate formation in the human colonic environment. Appl Environ Microbiol 2024; 90:e0101923. [PMID: 38126785 PMCID: PMC10807433 DOI: 10.1128/aem.01019-23] [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: 06/19/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Butyrate, a physiologically active molecule, can be synthesized through metabolic interactions among colonic microorganisms. Previously, in a fermenting trial of human fecal microbiota, we observed that the butyrogenic effect positively correlated with the increasing Bifidobacterium population and an unidentified Megasphaera species. Therefore, we hypothesized that a cross-feeding phenomenon exists between Bifidobacterium and Megasphaera, where Megasphaera is the butyrate producer, and its growth relies on the metabolites generated by Bifidobacterium. To validate this hypothesis, three bacterial species (B. longum, B. pseudocatenulatum, and M. indica) were isolated from fecal cultures fermenting hydrolyzed xylan; pairwise cocultures were conducted between the Bifidobacterium and M. indica isolates; the microbial interactions were determined based on bacterial genome information, cell growth, substrate consumption, metabolite quantification, and metatranscriptomics. The results indicated that two Bifidobacterium isolates contained distinct gene clusters for xylan utilization and expressed varying substrate preferences. In contrast, M. indica alone scarcely grew on the xylose-based substrates. The growth of M. indica was significantly elevated by coculturing it with bifidobacteria, while the two Bifidobacterium species responded differently in the kinetics of cell growth and substrate consumption. Coculturing led to the depletion of lactate and increased the formation of butyrate. An RNA-seq analysis further revealed the upregulation of M. indica genes involved in the lactate utilization and butyrate formation pathways. We concluded that lactate generated by Bifidobacterium through catabolizing xylose fueled the growth of M. indica and triggered the synthesis of butyrate. Our findings demonstrated a novel cross-feeding mechanism to generate butyrate in the human colon.IMPORTANCEButyrate is an important short-chain fatty acid that is produced in the human colon through microbial fermentation. Although many butyrate-producing bacteria exhibit a limited capacity to degrade nondigestible food materials, butyrate can be formed through cross-feeding microbial metabolites, such as acetate or lactate. Previously, the literature has explicated the butyrate-forming links between Bifidobacterium and Faecalibacterium prausnitzii and between Bifidobacterium and Eubacterium rectale. In this study, we provided an alternative butyrate synthetic pathway through the interaction between Bifidobacterium and Megasphaera indica. M. indica is a species named in 2014 and is indigenous to the human intestinal tract. Scientific studies explaining the function of M. indica in the human colon are still limited. Our results show that M. indica proliferated based on the lactate generated by bifidobacteria and produced butyrate as its end metabolic product. The pathways identified here may contribute to understanding butyrate formation in the gut microbiota.
Collapse
Affiliation(s)
- Sainan Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Raymond Lau
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Yang Zhong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Department of Clinical Translational Research, Singapore General Hospital, Singapore, Singapore
| | - Ming-Hsu Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
6
|
Li N, Wang H, Zhao H, Wang M, Cai J, Hao Y, Yu J, Jiang Y, Lü X, Liu B. Cooperative interactions between Veillonella ratti and Lactobacillus acidophilus ameliorate DSS-induced ulcerative colitis in mice. Food Funct 2023; 14:10475-10492. [PMID: 37934670 DOI: 10.1039/d3fo03898j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Veillonella and Lactobacillus species are key regulators of a healthy gut environment through metabolic cross-feeding, influencing lactic acid and short-chain fatty acid (SCFA) levels, which are crucial for gut health. This study aims to investigate how Veillonella ratti (V. ratti) and Lactobacillus acidophilus (LA) interact with each other and alleviate dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in a mouse model. We assess their metabolic interactions regarding carbon sources through co-culturing in a modified medium. In the in vitro experiments, V. ratti and LA were inoculated in mono-cultures and co-culture, and viable cell counts, OD600, pH, lactic acid, glucose and SCFAs were measured. For the in vivo experiment, 60 C57BL/6 mice were randomly divided into five groups and administered V. ratti and LA alone or in combination via oral gavage (1 × 109 CFU mL-1 per day per mouse) for 14 days. On the seventh day, 2.5% DSS was added to the drinking water to induce colitis. The effects of these probiotics on UC were evaluated by assessing intestinal barrier integrity and intestinal inflammation in the gut microenvironment. In vitro results demonstrated that co-culturing V. ratti with LA significantly increased viable cell numbers, lactic acid production, and SCFA production, while reducing pH and glucose levels in the medium. In vivo findings revealed that intervention with V. ratti, particularly in combination with LA, alleviated symptoms, including weight loss, colon shortening, and tissue damage. These probiotics mitigated intestinal inflammation by down-regulating pro-inflammatory molecules, such as IL-6, IL-1β, IL-γ, iNOS, and IFN-γ, as well as oxidative stress markers, including MDA and MPO. Concurrently, they upregulated the activity of anti-inflammatory enzymes, namely, SOD and GSH, and promoted the production of SCFAs. The combined intervention of V. ratti and LA significantly increased acetic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, and total SCFAs in cecal contents. Furthermore, the intervention of V. ratti and LA increased the abundance of beneficial bacteria, such as Akkermansia, while reducing the abundance of harmful bacteria, such as Escherichia-Shigella and Desulfovibrio, thereby mitigating excessive inflammation. These findings highlight the enhanced therapeutic effects resulting from the interactions between V. ratti and LA, demonstrating the potential of this combined probiotic approach.
Collapse
Affiliation(s)
- Na Li
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Hejing Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Huizhu Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Mengyang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jin Cai
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yi Hao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Jia Yu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Yun Jiang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Bianfang Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
7
|
Lee DH, Jee JJ, Lee YS, Kim DY, Bang JY, Lee HW, Koh H, Bae SH. Fecal microbiota transplantation improves hepatic fibro-inflammation via regulating oxidative stress in experimental NASH. Dig Liver Dis 2023; 55:1521-1532. [PMID: 37380586 DOI: 10.1016/j.dld.2023.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/09/2023] [Accepted: 06/14/2023] [Indexed: 06/30/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is associated with imbalance of gut microbiome, indicating participation of gut environment in hepatic health status. Therefore, modulating gut environment via fecal microbiota transplantation (FMT) is a promising therapeutic procedure for NASH patients. However, the effect and mechanism of the FMT remains largely unknown. Here, we investigated the gut-liver axis to understand the FMT-mediated hepatic improvement in NASH. Feces from specific pathogen free mice were infused allogeneically into gastrointestinal tract of mice fed with high fat, high cholesterol and fructose (HFHCF), resulting in suppressing hepatic pathogenic events, featured by decreasing inflammatory and fibrotic mediators. The FMT elevated NF-E2-related factor 2 (NRF2), a key transcription factor that regulates antioxidant enzymes, in livers. The HFHCF-induced NASH increased intestinal permeability with abundant Facklamia and Aerococcus, an imbalanced gut environment that was significantly improved by the FMT, characterized with restoration of intestinal barrier function and an enrichment of Clostridium. Notably, the gut environment created by FMT was inferred to produce metabolites from the aromatic biogenic amine degradation pathway, specifically 4-hydroxyphenylacetic acid (4-HPA), which is known to ameliorate liver injury. We suggest that gut-derived molecules, related to hepatic improvement such as 4-HPA are the potential therapeutic agents for preventing and treating NASH.
Collapse
Affiliation(s)
- Da Hyun Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Jai J Jee
- Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Republic of Korea
| | - Yu Seol Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea
| | - Da Ye Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea
| | - Ji Yun Bang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea
| | - Hye Won Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Hong Koh
- Department of Pediatrics, Yonsei University College of Medicine, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Seoul, Republic of Korea.
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea; Severance Biomedical Science Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Republic of Korea.
| |
Collapse
|
8
|
Mysonhimer AR, Cannavale CN, Bailey MA, Khan NA, Holscher HD. Prebiotic Consumption Alters Microbiota but Not Biological Markers of Stress and Inflammation or Mental Health Symptoms in Healthy Adults: A Randomized, Controlled, Crossover Trial. J Nutr 2023; 153:1283-1296. [PMID: 36841506 DOI: 10.1016/j.tjnut.2023.02.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/26/2023] [Accepted: 02/08/2023] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Chronic stress contributes to systemic inflammation and diminished mental health. Although animal work suggests strong links with the microbiota-gut-brain axis, clinical trials investigating the effectiveness of prebiotics in improving mental health and reducing inflammation are lacking. OBJECTIVES We aimed to determine fructooligosaccharide (FOS) and galactooligosaccharide (GOS) effects on biological markers of stress and inflammation and mental health symptoms in adults. Secondary outcomes included fecal microbiota and metabolites, digestive function, emotion, and sleep. METHODS Twenty-four healthy adults (25-45 y; 14 females, 10 males; BMI, 29.3 ± 1.8 kg/m2) from central Illinois participated in a 2-period, randomized, controlled, single-blinded crossover trial. Interventions included the prebiotic (PRE) treatment (237 mL/d Lactaid low-fat 1% milk, 5 g/d FOS, 5 g/d GOS) and control (CON) (237 mL/d Lactaid), which were consumed in counterbalanced order for 4 wk each, separated by ≥4-wk washout. Inflammatory markers were measured in blood plasma (>10-h fast) and cortisol in urine. The Depression Anxiety Stress Scales-42 assessed mental health symptoms. Fecal samples were collected for 16S rRNA gene (V4 region) sequencing and analysis. Emotion was measured by rating images from a computer task. Sleep was assessed using 7-d records and accelerometers. Change scores were analyzed using linear mixed models with treatment and baseline covariate as fixed effects and participant ID as the random effect. RESULTS There were no differences in change scores between PRE and CON treatments on biological markers of stress and inflammation or mental health. PRE increased change in percent sequences (q = 0.01) of Actinobacteriota (CON: 0.46 ± 0.70%; PRE: 5.40 ± 1.67%) and Bifidobacterium (CON: -1.72 ± 0.43%; PRE: 4.92 ± 1.53%). There were also no differences in change scores between treatments for microbial metabolites, digestive function, emotion, or sleep quality. CONCLUSIONS FOS+GOS did not affect biological markers of stress and inflammation or mental health symptoms in healthy adults; however, it increased Bifidobacterium. CLINICAL TRIAL REGISTRY NCT04551937, www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
| | | | - Melisa A Bailey
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA
| | - Naiman A Khan
- Neuroscience Program, University of Illinois, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA; Department of Kinesiology and Community Health, University of Illinois, Urbana, IL, USA
| | - Hannah D Holscher
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, USA; Division of Nutritional Sciences, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
9
|
Singh V, Lee G, Son H, Koh H, Kim ES, Unno T, Shin JH. Butyrate producers, "The Sentinel of Gut": Their intestinal significance with and beyond butyrate, and prospective use as microbial therapeutics. Front Microbiol 2023; 13:1103836. [PMID: 36713166 PMCID: PMC9877435 DOI: 10.3389/fmicb.2022.1103836] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/28/2022] [Indexed: 01/15/2023] Open
Abstract
Gut-microbial butyrate is a short-chain fatty acid (SCFA) of significant physiological importance than the other major SCFAs (acetate and propionate). Most butyrate producers belong to the Clostridium cluster of the phylum Firmicutes, such as Faecalibacterium, Roseburia, Eubacterium, Anaerostipes, Coprococcus, Subdoligranulum, and Anaerobutyricum. They metabolize carbohydrates via the butyryl-CoA: acetate CoA-transferase pathway and butyrate kinase terminal enzymes to produce most of butyrate. Although, in minor fractions, amino acids can also be utilized to generate butyrate via glutamate and lysine pathways. Butyrogenic microbes play a vital role in various gut-associated metabolisms. Butyrate is used by colonocytes to generate energy, stabilizes hypoxia-inducible factor to maintain the anaerobic environment in the gut, maintains gut barrier integrity by regulating Claudin-1 and synaptopodin expression, limits pro-inflammatory cytokines (IL-6, IL-12), and inhibits oncogenic pathways (Akt/ERK, Wnt, and TGF-β signaling). Colonic butyrate producers shape the gut microbial community by secreting various anti-microbial substances, such as cathelicidins, reuterin, and β-defensin-1, and maintain gut homeostasis by releasing anti-inflammatory molecules, such as IgA, vitamin B, and microbial anti-inflammatory molecules. Additionally, butyrate producers, such as Roseburia, produce anti-carcinogenic metabolites, such as shikimic acid and a precursor of conjugated linoleic acid. In this review, we summarized the significance of butyrate, critically examined the role and relevance of butyrate producers, and contextualized their importance as microbial therapeutics.
Collapse
Affiliation(s)
- Vineet Singh
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - GyuDae Lee
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - HyunWoo Son
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hong Koh
- Department of Pediatrics, Severance Fecal Microbiota Transplantation Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eun Soo Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tatsuya Unno
- Faculty of Biotechnology, School of Life Sciences, SARI, Jeju National University, Jeju, Republic of Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, Republic of Korea
- Department of Integrative Biotechnology, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
10
|
Tincati C, Ficara M, Ferrari F, Augello M, Dotta L, Tagliabue C, Diana A, Camelli V, Iughetti L, Badolato R, Cellini M, Marchetti G. Gut-dependent inflammation and alterations of the intestinal microbiota in individuals with perinatal HIV exposure and different HIV serostatus. AIDS 2022; 36:1917-1925. [PMID: 35848569 PMCID: PMC9612678 DOI: 10.1097/qad.0000000000003324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/24/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE HIV-exposed infected (HEI) and uninfected (HEU) children represent the two possible outcomes of maternal HIV infection. Modifications of the intestinal microbiome have been linked to clinical vulnerability in both settings, yet whether HEI and HEU differ in terms of gut impairment and peripheral inflammation/activation is unknown. DESIGN We performed a cross-sectional, pilot study on fecal and plasma microbiome as well as plasma markers of gut damage, microbial translocation, inflammation and immune activation in HIV-infected and uninfected children born from an HIV-infected mother. METHODS Fecal and plasma microbiome were determined by means of 16S rDNA amplification with subsequent qPCR quantification. Plasma markers were quantified via ELISA. RESULTS Forty-seven HEI and 33 HEU children were consecutively enrolled. The two groups displayed differences in fecal beta-diversity and relative abundance, yet similar microbiome profiles in plasma as well as comparable gut damage and microbial translocation. In contrast, monocyte activation (sCD14) and systemic inflammation (IL-6) were significantly higher in HEI than HEU. CONCLUSION In the setting of perinatal HIV infection, enduring immune activation and inflammation do not appear to be linked to alterations within the gut. Given that markers of activation and inflammation are independent predictors of HIV disease progression, future studies are needed to understand the underlying mechanisms of such processes and elaborate adjuvant therapies to reduce the clinical risk in individuals with perinatal HIV infection.
Collapse
Affiliation(s)
- Camilla Tincati
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Monica Ficara
- Division of Paediatric Oncology-Haematology, Policlinico Hospital, Modena
| | - Francesca Ferrari
- Division of Paediatric Oncology-Haematology, Policlinico Hospital, Modena
| | - Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| | - Laura Dotta
- Pediatric Clinic and ‘A. Nocivelli’ Institute for Molecular Medicine, Spedali Civili Hospital, Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | | | - Alfredo Diana
- Section of Pediatrics, Department of Translational Medical Science, University Federico II, Naples
| | - Vittoria Camelli
- Department of Sciences of Public Health and Pediatrics, University of Turin, Turin
| | - Lorenzo Iughetti
- Division of Paediatric Oncology-Haematology, Policlinico Hospital, Modena
- Pediatric Unit, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Raffaele Badolato
- Pediatric Clinic and ‘A. Nocivelli’ Institute for Molecular Medicine, Spedali Civili Hospital, Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Monica Cellini
- Division of Paediatric Oncology-Haematology, Policlinico Hospital, Modena
| | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, San Paolo Hospital, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan
| |
Collapse
|
11
|
Marchesi JR, Allen S, Scott E, Jenkins H, Sadlier C, Thomas S. An observational investigation of the faical microbiota and metabonome of gastrostomy fed children, on blended and formula diets. Gut Microbes 2022; 14:2138661. [PMID: 36284401 PMCID: PMC9621064 DOI: 10.1080/19490976.2022.2138661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gastrostomy fed children traditionally have a Formulae diet (FD), which fulfills nutritional requirements; however, many families are adopting Blended diets (BD), which are what the whole family would eat. We undertook an observational investigation of the colonic microbiota and metabonome in a small group of gastrostomy fed children, who were either on an FD or BD, and compared, where possible to their siblings (17 FD, 28 BD, 19 HS). There was no increase in complications in tube blockage or infection rates, but a significant improvement in the prevalence of bowel problems, a reduction in medication and an increase in quality of life. Metataxonomic analysis showed that the FD group was significantly different to the Sibling group, and that families did not cluster together. Whole sample metabonomics showed no differences between groups; however, univariate analysis of biologically important metabolites did differ. Changing to a BD resulted in no increase in complications or risks, but improved the overall quality of life for the children and families.
Collapse
Affiliation(s)
- Julian R. Marchesi
- Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital, Imperial College London, London, UK,CONTACT Julian R. Marchesi Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital, Imperial College London, South Wharf Road, LondonW2 1NYUK
| | - Sophie Allen
- School of Biosciences, Cardiff University, CardiffUK
| | - Emma Scott
- Department of Metabolism, Digestion and Reproduction, St Mary’s Hospital, Imperial College London, London, UK
| | - Huw Jenkins
- Department of Paediatric Gastroenterology, Noah’s Ark Children’s Hospital for Wales, Cardiff, UK
| | - Claire Sadlier
- Department of Child Health, University Hospital of Wales, CardiffUK
| | - Sian Thomas
- Aneurin Bevan University Health Board, Royal Gwent Hospital, Newport, UK
| |
Collapse
|
12
|
Li S, Luo X, Liao Z, Liang M, Xu H, Mai K, Zhang Y. Effects of Lysophosphatidylcholine on Intestinal Health of Turbot Fed High-Lipid Diets. Nutrients 2022; 14:4398. [PMID: 36297082 PMCID: PMC9611283 DOI: 10.3390/nu14204398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/30/2022] Open
Abstract
An 8-week feeding trial was conducted, where turbot were fed four experimental diets, containing different LPC levels (0%, 0.1%, 0.25%, and 0.5%, named LPC0, LPC0.1, LPC0.25, and LPC0.5, respectively). The intestinal morphology results showed that there were no widened lamina propria and mixed inflammatory cells in the LPC-supplemented groups. Dietary LPC remarkably decreased the expression of TLRs (TLR3, TLR8, TLR9, and TLR22), MyD88, and signaling molecules (NF-κB, JNK, and AP-1). Similarly, diets with LPC supplementation markedly depressed the gene expression of NF-κB and JNK signaling pathway downstream genes (TNF-α, IL-1β, Bax, Caspase9, and Caspase-3). Furthermore, dietary LPC modified the intestinal microbial profiles, increasing the relative abundance of short-chain fatty acids-producers, lactic acid bacteria, and digestive enzyme-producing bacteria. Predictive functions of intestinal microbiota showed that turbot fed LPC diets had a relatively higher abundance of functions, such as lipid metabolism and immune system, but a lower abundance of functions, such as metabolic diseases and immune system diseases. The activities of intestinal acid phosphatase and alkaline phosphatase were also increased by dietary LPC. In conclusion, LPC supplementation could regulate the intestinal mucosal barrier via the TLR signaling pathway and alter the intestinal microbiota profile of turbot fed high-lipid diets.
Collapse
Affiliation(s)
- Sihui Li
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture), The Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Xing Luo
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Zhangbin Liao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
| | - Mengqing Liang
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
- Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Houguo Xu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 106 Nanjing Road, Qingdao 266071, China
- Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Kangsen Mai
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture), The Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| | - Yanjiao Zhang
- The Key Laboratory of Aquaculture Nutrition and Feed (Ministry of Agriculture), The Key Laboratory of Mariculture (Ministry of Education), Ocean University of China, 5 Yushan Road, Qingdao 266003, China
- Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao 266237, China
| |
Collapse
|
13
|
Niikura M, Atobe S, Takahashi A, Kado Y, Sugimoto T, Tsuji H, Shimizu K, Ogura H, Asahara T. Development of a rapid and sensitive analytical system for Pseudomonas aeruginosa based on reverse transcription quantitative PCR targeting of rRNA molecules. Emerg Microbes Infect 2021; 10:677-686. [PMID: 33734032 PMCID: PMC8023615 DOI: 10.1080/22221751.2021.1906164] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/10/2021] [Accepted: 03/14/2021] [Indexed: 12/05/2022]
Abstract
For Pseudomonas aeruginosa (PA), infection control and appropriate antimicrobial treatment have become important issues. Diagnosis is critical in managing PA infection, but conventional methods are not highly accurate or rapid. We developed a new PA quantification system based on 23S rRNA-targeted reverse transcription quantitative PCR (RT-qPCR). We confirmed that RT-qPCR can quantify PA directly from clinical samples quickly (within 6 h) and with high sensitivity (blood, 1 cell/mL; stool, 100 cells/g) and without cross-reaction. Also, under antibiotic treatment, PA viable counts detected by this system correlated well with the inflammatory response of infected Caco-2 cells compared to other methods such as culturing and qPCR. Next, we utilized this system on fecal samples collected from 65 septic ICU patients and 44 healthy volunteers to identify ICU infection status. We confirmed that the PA detection ratio in ICU patients was significantly higher than that in healthy volunteers (49.2% vs. 13.6%, P < 0.05). Additionally, we monitored drug-resistant PA in 4 ICU patients by this system. The trends in PA counts accurately reflected various treatment backgrounds such as antibiotic use and mechanical ventilator use. Our results suggest that this RT-qPCR system is beneficial for the early diagnosis and evaluation of appropriate antibacterial treatment and may be a useful tool in combating PA infection.
Collapse
Affiliation(s)
- Mai Niikura
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Satomi Atobe
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Akira Takahashi
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Yukiko Kado
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Takuya Sugimoto
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Hirokazu Tsuji
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| | - Kentaro Shimizu
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroshi Ogura
- Department of Traumatology and Acute Critical Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co., Ltd., Kunitachi, Tokyo, Japan
| |
Collapse
|
14
|
Neelis EG, de Koning BAE, Hulst JM, Papadopoulou R, Kerbiriou C, Rings EHHM, Wijnen RMH, Nichols B, Gerasimidis K. Gut microbiota and its diet-related activity in children with intestinal failure receiving long-term parenteral nutrition. JPEN J Parenter Enteral Nutr 2021; 46:693-708. [PMID: 33982321 PMCID: PMC9255855 DOI: 10.1002/jpen.2188] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background This study characterized gut microbiota and its diet‐related activity in children with intestinal failure (IF) receiving parenteral nutrition (PN) compared with those of healthy controls (HC) and in relation to disease characteristics. Methods The fecal microbiota and short‐chain fatty acids (SCFAs) were measured in 15 IF patients (n = 68) and 25 HC (n = 25). Results Patients with IF had a lower bacterial load (P = .003), diversity (P < .001), evenness (P < .001) and richness (P = 0.006) than HC. Patients with surgical IF had lower diversity (P < .039) than those with functional IF. Propionic acid and butyric acid (p < .001) were lower and d‐lactate and l‐lactate were higher (p < 0.001) in IF patients than in HC. The energy supplied by PN (%PN) was negatively associated with microbiota diversity and SCFA profile. IF patients had more Escherichia‐Shigella (P = .006), Cronobacter (P = .001), and Staphylococcus (Operational Taxonomic Unit 14, P < .001) and less Faecalibacterium (P < 0.001) and Ruminococcus 1 and 2 (P < .001). Duration of PN (P = .005), %PN (P = .005), and fiber intake (P = .011) were predictive of microbiota structure. Higher intake of enteral nutrition was associated with microbiota structure and function closer to those of HC. Conclusions Microbiota composition and its diet‐related function are altered in IF, with depletion of beneficial SCFAs and species and supraphysiological increase of potentially harmful pathobionts. The influence of this compositional and functional microbial dysbiosis on patients’ outcomes and management warrants further exploration.
Collapse
Affiliation(s)
- Esther G Neelis
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Barbara A E de Koning
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Jessie M Hulst
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rodanthi Papadopoulou
- Human Nutrition, School of Medicine, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Caroline Kerbiriou
- Human Nutrition, School of Medicine, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Edmond H H M Rings
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatric Gastroenterology, Leiden University Medical Center-Willem Alexander Children's Hospital, Leiden, The Netherlands
| | - René M H Wijnen
- Department of Pediatric Surgery, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Ben Nichols
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Konstantinos Gerasimidis
- Department of Pediatric Gastroenterology, Erasmus Medical Center-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Takada T, Chinda D, Mikami T, Shimizu K, Oana K, Hayamizu S, Miyazawa K, Arai T, Katto M, Nagara Y, Makino H, Kushiro A, Oishi K, Fukuda S. Dynamic analysis of human small intestinal microbiota after an ingestion of fermented milk by small-intestinal fluid perfusion using an endoscopic retrograde bowel insertion technique. Gut Microbes 2020; 11:1662-1676. [PMID: 32552401 PMCID: PMC7524281 DOI: 10.1080/19490976.2020.1766942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Probiotic products have been shown to have beneficial effects on human hosts, but what happens in the gastrointestinal tract after its ingestion remains unclear. Our aim was to investigate the changes within the small intestines after a single intake of a fermented milk product containing a probiotic. We have periodically collected the small-intestinal fluids from the terminal ileum of seven healthy subjects for up to 7 h after ingestion by small-intestinal fluid perfusion using an endoscopic retrograde bowel insertion technique. The bacterial composition of the terminal ileum clearly revealed that the ingested probiotics (Lactobacillus casei strain Shirota: LcS and Bifidobacterium breve strain Yakult: BbrY) occupied the ileal microbiota for several hours, temporarily representing over 90% of the ileal microbiota in several subjects. Cultivation of ileal fluids showed that under a dramatic pH changes before reaching the terminal ileum, a certain number of the ingested bacteria survived (8.2 ± 6.4% of LcS, 7.8 ± 11.0% of BbrY). This means that more than 1 billion LcS and BbrY cells reached the terminal ileum with their colony-forming ability intact. These results indicate that there is adequate opportunity for the ingested probiotics to continuously stimulate the host cells in the small intestines. Our data suggest that probiotic fermented milk intake affects intestinal microbes and the host, explaining part of the process from the intake of probiotics to the exertion of their beneficial effects on the host.
Collapse
Affiliation(s)
- Toshihiko Takada
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan,CONTACT Toshihiko Takada Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo, 186-8650, Japan
| | - Daisuke Chinda
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| | - Tatsuya Mikami
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| | - Kensuke Shimizu
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Kosuke Oana
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Shiro Hayamizu
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| | - Kuniaki Miyazawa
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| | - Tetsu Arai
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| | - Miyuki Katto
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Yusuke Nagara
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Hiroshi Makino
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Akira Kushiro
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Kenji Oishi
- Laboratory of Applied Microbiology, Microbiological Research Department, Yakult Central Institute, Tokyo, Japan
| | - Shinsaku Fukuda
- Laboratory of Gastroenterology and Hematology, Graduate School of Medicine, Hirosaki University, Aomori, Japan
| |
Collapse
|
16
|
1-Kestose supplementation mitigates the progressive deterioration of glucose metabolism in type 2 diabetes OLETF rats. Sci Rep 2020; 10:15674. [PMID: 32973311 PMCID: PMC7515885 DOI: 10.1038/s41598-020-72773-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The fructooligosaccharide 1-kestose cannot be hydrolyzed by gastrointestinal enzymes, and is instead fermented by the gut microbiota. Previous studies suggest that 1-kestose promotes increases in butyrate concentrations in vitro and in the ceca of rats. Low levels of butyrate-producing microbiota are frequently observed in the gut of patients and experimental animals with type 2 diabetes (T2D). However, little is known about the role of 1-kestose in increasing the butyrate-producing microbiota and improving the metabolic conditions in type 2 diabetic animals. Here, we demonstrate that supplementation with 1-kestose suppressed the development of diabetes in Otsuka Long-Evans Tokushima Fatty (OLETF) rats, possibly through improved glucose tolerance. We showed that the cecal contents of rats fed 1-kestose were high in butyrate and harbored a higher proportion of the butyrate-producing genus Anaerostipes compared to rats fed a control diet. These findings illustrate how 1-kestose modifications to the gut microbiota impact glucose metabolism of T2D, and provide a potential preventative strategy to control glucose metabolism associated with dysregulated insulin secretion.
Collapse
|
17
|
Administration of Bifidobacterium bifidum CGMCC 15068 modulates gut microbiota and metabolome in azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced colitis-associated colon cancer (CAC) in mice. Appl Microbiol Biotechnol 2020; 104:5915-5928. [PMID: 32367312 DOI: 10.1007/s00253-020-10621-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/31/2022]
Abstract
The gut microbiota plays an important role in colorectal cancer (CRC), and the use of probiotics might be a promising intervention method. The aim of our study was to investigate the beneficial effect of Bifidobacterium bifidum CGMCC 15068 on an azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced colitis-associated CRC (CAC) mouse model. CAC was induced by an intra-peritoneal injection of AOM (10 mg/kg) and three 7-day cycles of 2% DSS in drinking water with a 14-day recovery period between two consecutive DSS administrations. B. bifidum CGMCC 15068 (3 × 109 CFU/mL) was gavaged once daily during the recovery period. Then, the faecal microbial composition and metabolome were profiled using the 16S rRNA sequencing technology and gas chromatography-mass spectrometry (GC-MS), respectively. The administration of B. bifidum CGMCC 15068 attenuated tumourigenesis in the CAC mouse model. In addition, B. bifidum CGMCC 15068 pre-treatment increased the relative abundance of Akkermansia, Desulfovibrionaceae, Romboutsia, Turicibacter, Verrucomicrobiaceae, Ruminococcaceae_UCG_013, Lachnospiraceae_UCG_004, and Lactobacillus. Meanwhile, B. bifidum CGMCC 15068 altered metabolites involved in the citrate cycle (TCA cycle), glycolysis, butyrate metabolism, fatty acid biosynthesis, and galactose metabolism. Several significant correlations were identified between the differentially abundant microbes and metabolites. These findings supported the beneficial role of B. bifidum CGMCC 15068 in intestinal health by modulating dysbiosis and the gut metabolic profile. The manipulation of the gut microbial composition using probiotics might be a promising prevention strategy for CRC. Long-term and large-scale clinical trials are warranted for the potential clinical applications of this strategy in the future.
Collapse
|
18
|
Wang Q, Ye J, Fang D, Lv L, Wu W, Shi D, Li Y, Yang L, Bian X, Wu J, Jiang X, Wang K, Wang Q, Hodson MP, Thibaut LM, Ho JWK, Giannoulatou E, Li L. Multi-omic profiling reveals associations between the gut mucosal microbiome, the metabolome, and host DNA methylation associated gene expression in patients with colorectal cancer. BMC Microbiol 2020; 20:83. [PMID: 32321427 PMCID: PMC7178946 DOI: 10.1186/s12866-020-01762-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022] Open
Abstract
Background The human gut microbiome plays a critical role in the carcinogenesis of colorectal cancer (CRC). However, a comprehensive analysis of the interaction between the host and microbiome is still lacking. Results We found correlations between the change in abundance of microbial taxa, butyrate-related colonic metabolites, and methylation-associated host gene expression in colonic tumour mucosa tissues compared with the adjacent normal mucosa tissues. The increase of genus Fusobacterium abundance was correlated with a decrease in the level of 4-hydroxybutyric acid (4-HB) and expression of immune-related peptidase inhibitor 16 (PI16), Fc Receptor Like A (FCRLA) and Lymphocyte Specific Protein 1 (LSP1). The decrease in the abundance of another potentially 4-HB-associated genus, Prevotella 2, was also found to be correlated with the down-regulated expression of metallothionein 1 M (MT1M). Additionally, the increase of glutamic acid-related family Halomonadaceae was correlated with the decreased expression of reelin (RELN). The decreased abundance of genus Paeniclostridium and genus Enterococcus were correlated with increased lactic acid level, and were also linked to the expression change of Phospholipase C Beta 1 (PLCB1) and Immunoglobulin Superfamily Member 9 (IGSF9) respectively. Interestingly, 4-HB, glutamic acid and lactic acid are all butyrate precursors, which may modify gene expression by epigenetic regulation such as DNA methylation. Conclusions Our study identified associations between previously reported CRC-related microbial taxa, butyrate-related metabolites and DNA methylation-associated gene expression in tumour and normal colonic mucosa tissues from CRC patients, which uncovered a possible mechanism of the role of microbiome in the carcinogenesis of CRC. In addition, these findings offer insight into potential new biomarkers, therapeutic and/or prevention strategies for CRC.
Collapse
Affiliation(s)
- Qing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.,Computational Genomics Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia
| | - Jianzhong Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Daiqiong Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Wenrui Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Yating Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Liya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiaoyuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jingjing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Kaicen Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Mark P Hodson
- Freedman Foundation Metabolomics Facility, Victor Chang Innovation Centre, Victor Chang Cardiac Research Institute, Sydney, Australia.,School of Pharmacy, University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Loïc M Thibaut
- Computational Genomics Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia.,School of Mathematics and Statistics, UNSW Sydney, Sydney, Australia
| | - Joshua W K Ho
- Bioinformatics and Systems Medicine Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Eleni Giannoulatou
- Computational Genomics Laboratory, Victor Chang Cardiac Research Institute, Sydney, Australia. .,St Vincent's Clinical School, UNSW Sydney, Sydney, Australia.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China. .,Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.
| |
Collapse
|
19
|
Metagenomic study of endophytic bacterial community of sweet potato (Ipomoea batatas) cultivated in different soil and climatic conditions. World J Microbiol Biotechnol 2019; 35:176. [PMID: 31673867 DOI: 10.1007/s11274-019-2754-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
The aim of this study was to clarify effects of soil and climatic conditions on community structure of sweet potato bacterial endophytes by applying locked nucleic acid oligonucleotide-PCR clamping technique and metagenomic analysis. For this purpose, the soil samples in three locations were transferred each other and sweet potato nursery plants from the same farm were cultivated for ca. 3 months. After removal of plastid, mitochondria and undefined sequences, the averaged numbers of retained sequences and operational taxonomic units per sample were 20,891 and 846, respectively. Proteobacteria (85.0%), Bacteroidetes (6.6%) and Actinobacteria (6.3%) were the three most dominant phyla, accounting for 97.9% of the reads, and γ-Proteobacteria (66.3%) being the most abundant. Top 10 genera represented 81.2% of the overall reads in which Pseudomonas (31.9-45.0%) being the most predominant. The overall endophytic bacterial communities were similar among the samples which indicated that the soil and the climatic conditions did not considerably affect the entire endophytic community. The original endophytic bacterial community might be kept during the cultivation period.
Collapse
|
20
|
Dietary intervention using (1,3)/(1,6)-β-glucan, a fungus-derived soluble prebiotic ameliorates high-fat diet-induced metabolic distress and alters beneficially the gut microbiota in mice model. Eur J Nutr 2019; 59:2617-2629. [DOI: 10.1007/s00394-019-02110-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/04/2019] [Indexed: 01/21/2023]
|
21
|
Muthuramalingam K, Singh V, Choi C, Choi SI, Park S, Kim YM, Unno T, Cho M. Effect of mushroom (Schizophyllum spp.) derived β-glucan on low-fiber diet induced gut dysbiosis. ACTA ACUST UNITED AC 2019. [DOI: 10.3839/jabc.2019.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Karthika Muthuramalingam
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Vineet Singh
- Faculty of Biotechnology, College of Applied Life Sciences, SARI, Jeju National University, Jeju 63243, Republic of Korea
| | - Changmin Choi
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Seung In Choi
- Department of Pharmaceutical Research Institute, Quegen Biotech Co. Ltd., Seoul 429931, Republic of Korea
| | - Sanggyu Park
- Division of Life & Environmental Science, Daegu University, Daegu 712-714, Republic of Korea
| | - Young Mee Kim
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Tatsuya Unno
- Faculty of Biotechnology, College of Applied Life Sciences, SARI, Jeju National University, Jeju 63243, Republic of Korea
- Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju 63243, Republic of Korea
| | - Moonjae Cho
- Department of Biochemistry, School of Medicine, Jeju National University, Jeju 63241, Republic of Korea
- Department of Biochemistry, School of Medicine, Institute of Medical Sciences, Jeju National University, Jeju 63241, Republic of Korea
| |
Collapse
|
22
|
Karetkin BA, Guseva EV, Evdokimova SA, Mishchenko AS, Khabibulina NV, Grosheva VD, Menshutina NV, Panfilov VI. A quantitative model of Bacillus cereus ATCC 9634 growth inhibition by bifidobacteria for synbiotic effect evaluation. World J Microbiol Biotechnol 2019; 35:89. [PMID: 31134431 DOI: 10.1007/s11274-019-2665-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
The present study is dedicated to the development of novel criteria for assessing the synbiotic effect of prebiotic and probiotic composition against a specific pathogen. These criteria were obtained from the quantitative model of Bifidobacterium adolescentis ATCC 15703 and Bacillus cereus ATCC 9634 (as a model food contaminant) competition in co-culture fermentation. The model is based on the hypothesis that probiotics can reduce the specific growth rate of non-probiotics by producing short-chain fatty acids. To define the relationship between the specific growth rate of non-probiotics and short-chain fatty acid yields, the inhibition constants were determined separately for each inhibitor produced by bifidobacteria (lactic, acetic and propionic acids) in a pure culture of bacilli. Two different equations based on the minimum inhibitor concentration (MIC) and inhibition constant (Ki) were used to connect the specific growth rate and concentrations of inhibitors. The yields of the inhibitors mentioned above were obtained from co-culture experiments. The experimental values and the values predicted by the model of Bacillus count did not differ significantly (R2 not less than 0.83) in the competition experiments. Therefore, the general criterion of the synbiotic effect was derived from the model and presents the coefficient of non-probiotic specific growth rate reduction as a result of probiotic growth and inhibitor formation in the final co-culture fermentation. This criterion has been examined for different commercial prebiotics coupled with the Bifidobacterium adolescentis strain. The synergistic combination of inulin GR with bifidobacteria had the best effect against Bacillus cereus ATCC 9634.
Collapse
Affiliation(s)
- Boris A Karetkin
- Department of Biotechnology, Faculty of Biotechnology and Industrial Ecology, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia.
| | - Elena V Guseva
- Department of Cybernetics of Chemical Engineering Processes, Faculty of Information Technologies and Management, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Svetlana A Evdokimova
- Department of Biotechnology, Faculty of Biotechnology and Industrial Ecology, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Anastasia S Mishchenko
- Department of Cybernetics of Chemical Engineering Processes, Faculty of Information Technologies and Management, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Natalia V Khabibulina
- Department of Biotechnology, Faculty of Biotechnology and Industrial Ecology, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Veronika D Grosheva
- Department of Biotechnology, Faculty of Biotechnology and Industrial Ecology, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Natalia V Menshutina
- Department of Cybernetics of Chemical Engineering Processes, Faculty of Information Technologies and Management, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| | - Victor I Panfilov
- Department of Biotechnology, Faculty of Biotechnology and Industrial Ecology, Dmitry Mendeleev University of Chemical Technology of Russia, Miusskaya Sq., 9, 125047, Moscow, Russia
| |
Collapse
|
23
|
Lordan C, Thapa D, Ross RP, Cotter PD. Potential for enriching next-generation health-promoting gut bacteria through prebiotics and other dietary components. Gut Microbes 2019; 11:1-20. [PMID: 31116628 PMCID: PMC6973326 DOI: 10.1080/19490976.2019.1613124] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The human intestinal commensal microbiota and associated metabolic products have long been regarded as contributors to host health. As the identity and activities of the various members of this community have become clearer, newly identified health-associated bacteria, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, Ruminococcus bromii and Roseburia species, have emerged. Notably, the abundance of many of these bacteria is inversely correlated to several disease states. While technological and regulatory hurdles may limit the use of strains from these taxa as probiotics, it should be possible to utilize prebiotics and other dietary components to selectively enhance their growth in situ. Dietary components of potential relevance include well-established prebiotics, such as galacto-oligosaccharides, fructo-oligosaccharides and inulin, while other putative prebiotics, such as other oligosaccharides, polyphenols, resistant starch, algae and seaweed as well as host gut metabolites such as lactate and acetate, may also be applied with the aim of selectively and/or differentially affecting the beneficial bacterial community within the gastrointestinal environment. The present review provides an overview of the dietary components that could be applied in this manner.
Collapse
Affiliation(s)
- Cathy Lordan
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland,School of Microbiology, University College Cork, Ireland
| | - Dinesh Thapa
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - R. Paul Ross
- School of Microbiology, University College Cork, Ireland,APC Microbiome Ireland, University College Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland,APC Microbiome Ireland, University College Cork, Ireland,CONTACT Paul D. Cotter
| |
Collapse
|
24
|
Aoe S, Nakamura F, Fujiwara S. Effect of Wheat Bran on Fecal Butyrate-Producing Bacteria and Wheat Bran Combined with Barley on Bacteroides Abundance in Japanese Healthy Adults. Nutrients 2018; 10:nu10121980. [PMID: 30558159 PMCID: PMC6316639 DOI: 10.3390/nu10121980] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/18/2022] Open
Abstract
Wheat bran (WB) is rich in insoluble arabinoxylan, while BARLEYmax (BM) is a barley line that is rich in fructan, resistant starch, and β-glucan. In the present study, we investigated which of these two fiber sources would produce more favorable changes in the fecal variables of healthy subjects. Sixty healthy subjects were randomly divided into four groups (n = 15 per group) and fed twice daily for 4 weeks with baked cereal bars containing neither WB nor BM (WB−BM−), WB without BM (WB+BM−), BM without WB (WB−BM+), or WB and BM (WB+BM+). At baseline and after 4 weeks, the fecal microbiota composition and the concentrations of short-chain fatty acids were measured. A significant interactive effect of WB and BM on the abundance of genus Bacteroides was observed at week 4. The abundance of butyrate-producing bacteria and the fecal concentration of n-butyrate were significantly higher in the WB+ groups than in the WB− groups. In conclusion, WB was associated with elevated fecal concentrations of short-chain fatty acids including butyrate owing to an increase in the abundance of butyrate-producing bacteria. Additionally, the combination of WB and BM was associated with an increase in the abundance of genus Bacteroides. Therefore, both WB alone and WB combined with BM favorably influenced the fecal variables of healthy subjects.
Collapse
Affiliation(s)
- Seiichiro Aoe
- Department of Food Science, Faculty of Home Economics, Otsuma Women's University, 12 Sanban-cho, Chiyoda-ku, Tokyo 102-8357, Japan.
| | - Fumiko Nakamura
- CPCC Company Limited, 3-3-5 Uchikanda, Chiyoda-ku, Tokyo 101-0047, Japan.
| | - Suguru Fujiwara
- CPCC Company Limited, 3-3-5 Uchikanda, Chiyoda-ku, Tokyo 101-0047, Japan.
| |
Collapse
|
25
|
Burlina A, Tims S, van Spronsen F, Sperl W, Burlina AP, Kuhn M, Knol J, Rakhshandehroo M, Coşkun T, Singh RH, MacDonald A. The potential role of gut microbiota and its modulators in the management of propionic and methylmalonic acidemia. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1536540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Alberto Burlina
- Division of Inherited Metabolic Diseases Reference Centre Expanded Newborn Screening, Padova, Italy
| | - Sebastian Tims
- Gut and Microbiology Platform, Nutricia Research, Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Francjan van Spronsen
- Division of Metabolic Diseases, Beatrix Children’s Hospital, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | - Wolfgang Sperl
- Salzburger Landeskliniken and Paracelsus Medical University Salzburg, Salzburg, Austria
| | | | - Mirjam Kuhn
- Research Department of Paediatric Care and Metabolic Control, Nutricia Research, Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Jan Knol
- Gut and Microbiology Platform, Nutricia Research, Advanced Medical Nutrition, Utrecht, The Netherlands
- Lab of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Maryam Rakhshandehroo
- Research Department of Paediatric Care and Metabolic Control, Nutricia Research, Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Turgay Coşkun
- Department of Pediatrics Division of Metabolism and Nutrition, Hacettepe University Faculty of Medicine, Turkey
| | - Rani H Singh
- Division of Medical Genetics, Nutrition Section, Emory University, Atlanta, USA
| | - Anita MacDonald
- Gut and Microbiology Platform, Nutricia Research, Advanced Medical Nutrition, Utrecht, The Netherlands
- Birmingham Children’s Hospital, Birmingham, UK
| |
Collapse
|
26
|
Hua C, Geng Y, Chen Q, Niu L, Cai L, Tao S, Ni Y, Zhao R. Chronic dexamethasone exposure retards growth without altering the digestive tract microbiota composition in goats. BMC Microbiol 2018; 18:112. [PMID: 30200878 PMCID: PMC6131888 DOI: 10.1186/s12866-018-1253-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023] Open
Abstract
Background Dexamethasone (Dex), an artificially synthetic cortisol substitute, is commonly used as an anti-inflammatory drug, and is also employed to mimic the stress state experimentally. It is well known that chronic stress disturbs the gut microbiota community and digestive functions. However, no relevant studies have been conducted in ruminants. Results In this study, a low dosage of Dex (0.2 mg/kg body weight, Dex group, n = 5) was consecutively injected intramuscularly for 21 days to simulate chronic stress in growing goats. Goats were injected with saline (0.2 mg/kg body weight) as the control group (Con, n = 5). Dex-treated goats showed a higher number of white blood cells and blood glucose levels (p < 0.01), but lower dry matter intake (DMI) and body weight (p < 0.01) than those of saline-injected goats. Plasma cortisol concentration decreased significantly in response to the Dex injection compared to the control (p < 0.05). The Dex treatment did not change most ruminal volatile fatty acid (VFAs) concentrations before the morning feeding after 1–21 days of treatment (p > 0.05); however, ruminal VFA concentrations decreased dramatically 2, 4, 6, and 8 h after the morning feeding on day 21 of the Dex injections. In this study, chronic Dex exposure did not alter the community structure of microbes or methanogenes in the rumen, caecum, or colonic digesta. Only Prevotella increased on days 7 and 14 of Dex treatment, but decreased on day 21, and Methanosphaera was the only genus of methanogene that decreased. Conclusions Our results suggest that chronic Dex exposure retards growth by decreasing DMI, which may be mediated by higher levels of blood glucose and lower ruminal VFA production. Microbiota in the digestive tract was highly resistant to chronic Dex exposure. Electronic supplementary material The online version of this article (10.1186/s12866-018-1253-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Canfeng Hua
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Yali Geng
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Qu Chen
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Liqiong Niu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Liuping Cai
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Shiyu Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Yingdong Ni
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| |
Collapse
|
27
|
Yoshikawa S, Araoka R, Kajihara Y, Ito T, Miyamoto H, Kodama H. Valerate production by Megasphaera elsdenii isolated from pig feces. J Biosci Bioeng 2018; 125:519-524. [DOI: 10.1016/j.jbiosc.2017.12.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/29/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023]
|
28
|
Wopereis H, Sim K, Shaw A, Warner JO, Knol J, Kroll JS. Intestinal microbiota in infants at high risk for allergy: Effects of prebiotics and role in eczema development. J Allergy Clin Immunol 2018; 141:1334-1342.e5. [DOI: 10.1016/j.jaci.2017.05.054] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/17/2017] [Accepted: 05/04/2017] [Indexed: 12/18/2022]
|
29
|
Preferential isolation of Megasphaera elsdenii from pig feces. Anaerobe 2017; 48:160-164. [PMID: 28842275 DOI: 10.1016/j.anaerobe.2017.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/20/2017] [Accepted: 08/21/2017] [Indexed: 01/19/2023]
Abstract
Lactic acid produced by intestinal bacteria is fermented by lactate-utilizing bacteria. In this study, we developed a selective culture medium (KMI medium) for Megasphaera elsdenii, a lactate-utilizing bacterium that is abundant in pig intestines. Supplementation of the medium with lactate and beef extract powder was necessary for the preferential growth of M. elsdenii. In addition, we designed a species-specific primer set to detect M. elsdenii. When pig fecal samples were plated on KMI agar medium, approximately 60-100% of the resulting colonies tested positive using the M. elsdenii-specific PCR primers. In fact, nearly all of the large, yellow-white colonies that grew on the KMI agar medium tested positive by PCR with this primer set. The 16S rRNA gene sequences of three representative PCR-positive strains showed strong similarities to that of M. elsdenii ATCC 25940T (98.9-99.2% identity). These three strains were approximately 1.5 μm sized cocci that were primarily arranged in pairs, as was observed for M. elsdenii JCM 1772T. The selective KMI medium and species-specific primer set developed in this study are useful for the isolation and detection of M. elsdenii and will be useful in research aimed at increasing our understanding of intestinal short-chain fatty acid metabolism in pigs.
Collapse
|
30
|
Desguin B, Soumillion P, Hausinger RP, Hols P. Unexpected complexity in the lactate racemization system of lactic acid bacteria. FEMS Microbiol Rev 2017; 41:S71-S83. [DOI: 10.1093/femsre/fux021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/11/2017] [Indexed: 12/30/2022] Open
|
31
|
Isaiah A, Parambeth JC, Steiner JM, Lidbury JA, Suchodolski JS. The fecal microbiome of dogs with exocrine pancreatic insufficiency. Anaerobe 2017; 45:50-58. [DOI: 10.1016/j.anaerobe.2017.02.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/31/2017] [Accepted: 02/13/2017] [Indexed: 02/07/2023]
|
32
|
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory disease of the colon with unclear pathogenesis. A dysbiotic intestinal microbiota is regarded as a key component in the disease process and there has been significant interest in developing new treatments which target the microbiota. AIM To give an overview of the studies to date investigating prebiotics and synbiotics for the treatment of UC. METHODS A literature search of PubMed and related search engines was carried out using the terms "ulcerative colitis" in combination with "prebiotic", "synbiotic" or "dietary fibre". RESULTS In total 17 studies on humans examining the effect of prebiotics in UC were found. Five major groups could be distinguished. Fructo-oligosaccharides were tried in six studies (mean 35 patients included, range 9-121). One study found a clinical response while two demonstrated indirect evidence of an effect. Germinated barley foodstuff was used in 8 studies (mean 38 patients, range 10-63). One study found an endoscopic response, while four noted a clinical response and two some indirect effects. Galacto-oligosaccharides, lactulose and resveratrol were used in one study each (mean 48 patients, range 41-52). One study found an endoscopic response and one a clinical response. CONCLUSION There is yet inadequate evidence - especially in humans - to support any particular prebiotic in the clinical management of UC. However, due to the bulk of evidence supporting the effect of the microbiota on colonic inflammation, there is enough potential to justify further high-quality clinical trials investigating this subject.
Collapse
Affiliation(s)
- Axel Laurell
- a Department of Clinical Sciences Malmö , Lund University, Department of Gastroenterology and Nutrition, Malmö, Skåne University Hospital , Malmö , Sweden
| | - Klas Sjöberg
- a Department of Clinical Sciences Malmö , Lund University, Department of Gastroenterology and Nutrition, Malmö, Skåne University Hospital , Malmö , Sweden
| |
Collapse
|
33
|
Godo T, Saki Y, Nojiri Y, Tsujitani M, Sugahara S, Hayashi S, Kamiya H, Ohtani S, Seike Y. Geosmin-producing Species of Coelosphaerium (Synechococcales, Cyanobacteria) in Lake Shinji, Japan. Sci Rep 2017; 7:41928. [PMID: 28195147 PMCID: PMC5307322 DOI: 10.1038/srep41928] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/04/2017] [Indexed: 11/09/2022] Open
Abstract
In Lake Shinji, Japan, periodic outbreaks of musty odour have occurred since mid-May 2007. Although the substance responsible for the odour was identified as geosmin, the odour-producing organism was unknown. We cultivated an axenic unialgal strain and determined that a species of Coelosphaerium (Synechococcales) was responsible for the production of geosmin in Lake Shinji. Our analysis was conducted using gas chromatography/mass spectrometry to determine the odorous compound. To determine the algae species, it was observed by optical microscopy to describe its morphological characteristics and the polymerase chain reaction was used to characterise the nucleotide sequence of the 16S rRNA gene and the 16S-23S rRNA internal transcribed spacer region. In addition, we explored the relationship between the number of cells of the Coelosphaerium sp. and the concentration of geosmin. In conclusion, geosmin, the cause of the musty odour in Lake Shinji in autumn 2009, was produced by Coelosphaerium sp., and to our knowledge, this is the first report of a geosmin-producing species in the family Coelosphaeriaceae.
Collapse
Affiliation(s)
- T Godo
- Shimane Prefectural Institute of Public Health and Environmental Science, 582-1 Nishihamasada, Matsue, Shimane 690-0122, Japan
| | - Y Saki
- Shimane Prefectural Institute of Public Health and Environmental Science, 582-1 Nishihamasada, Matsue, Shimane 690-0122, Japan
| | - Y Nojiri
- Shimane Prefectural Institute of Public Health and Environmental Science, 582-1 Nishihamasada, Matsue, Shimane 690-0122, Japan
| | - M Tsujitani
- The United Graduate School of Agricultural Sciences, Tottori University, 4-101 Koyama-cho minami, Tottori, Tottori 680-8553, Japan
| | - S Sugahara
- Graduate School of Science and Engineering, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan
| | - S Hayashi
- Faculty of Life and Environmental Science, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan
| | - H Kamiya
- Shimane Prefectural Institute of Public Health and Environmental Science, 582-1 Nishihamasada, Matsue, Shimane 690-0122, Japan
| | - S Ohtani
- Faculty of Education, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan
| | - Y Seike
- Graduate School of Science and Engineering, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan
| |
Collapse
|
34
|
Kumari M, Kozyrskyj AL. Gut microbial metabolism defines host metabolism: an emerging perspective in obesity and allergic inflammation. Obes Rev 2017; 18:18-31. [PMID: 27862824 DOI: 10.1111/obr.12484] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/26/2016] [Accepted: 10/05/2016] [Indexed: 12/14/2022]
Abstract
The presence of >100 trillion microorganisms (collectively called gut microbiota) in our large intestine is essential for the maintenance of health. The gut microbiota starts to develop before birth and matures within first three years of life. The Western diet and lifestyle have been implicated in causing an imbalance of gut microbial communities and their metabolites that consequence in disease states, such as obesity and asthma. With more than 13% of the world population currently living with obesity and one out of 10 children diagnosed with asthma, we explore here the recent developments in the biosynthesis and mode of action of the key metabolites in relation to these two chronic inflammatory conditions.
Collapse
Affiliation(s)
- M Kumari
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - A L Kozyrskyj
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,Department of Obstetrics and Gynecology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.,School of Public Health, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
35
|
Czaja AJ. Factoring the intestinal microbiome into the pathogenesis of autoimmune hepatitis. World J Gastroenterol 2016; 22:9257-9278. [PMID: 27895415 PMCID: PMC5107691 DOI: 10.3748/wjg.v22.i42.9257] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/07/2016] [Accepted: 10/31/2016] [Indexed: 02/06/2023] Open
Abstract
The intestinal microbiome is a reservoir of microbial antigens and activated immune cells. The aims of this review were to describe the role of the intestinal microbiome in generating innate and adaptive immune responses, indicate how these responses contribute to the development of systemic immune-mediated diseases, and encourage investigations that improve the understanding and management of autoimmune hepatitis. Alterations in the composition of the intestinal microflora (dysbiosis) can disrupt intestinal and systemic immune tolerances for commensal bacteria. Toll-like receptors within the intestine can recognize microbe-associated molecular patterns and shape subsets of T helper lymphocytes that may cross-react with host antigens (molecular mimicry). Activated gut-derived lymphocytes can migrate to lymph nodes, and gut-derived microbial antigens can translocate to extra-intestinal sites. Inflammasomes can form within hepatocytes and hepatic stellate cells, and they can drive the pro-inflammatory, immune-mediated, and fibrotic responses. Diet, designer probiotics, vitamin supplements, re-colonization methods, antibiotics, drugs that decrease intestinal permeability, and molecular interventions that block signaling pathways may emerge as adjunctive regimens that complement conventional immunosuppressive management. In conclusion, investigations of the intestinal microbiome are warranted in autoimmune hepatitis and promise to clarify pathogenic mechanisms and suggest alternative management strategies.
Collapse
|
36
|
Sato T, Kusuhara S, Yokoi W, Ito M, Miyazaki K. Prebiotic potential of L-sorbose and xylitol in promoting the growth and metabolic activity of specific butyrate-producing bacteria in human fecal culture. FEMS Microbiol Ecol 2016; 93:fiw227. [PMID: 27810878 DOI: 10.1093/femsec/fiw227] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/18/2016] [Accepted: 10/31/2016] [Indexed: 12/21/2022] Open
Abstract
Dietary low-digestible carbohydrates (LDCs) affect gut microbial metabolism, including the production of short-chain fatty acids. The ability of various LDCs to promote butyrate production was evaluated in in vitro human fecal cultures. Fecal suspensions from five healthy males were anaerobically incubated with various LDCs. L-Sorbose and xylitol markedly promoted butyrate formation in cultures. Bacterial 16S rRNA gene-based denaturing gradient gel electrophoresis analyses of these fecal cultures revealed a marked increase in the abundance of bacteria closely related to the species Anaerostipes hadrus or A. caccae or both, during enhanced butyrate formation from L-sorbose or xylitol. By using an agar plate culture, two strains of A. hadrus that produced butyrate from each substrate were isolated from the feces of two donors. Furthermore, of 12 species of representative colonic butyrate producers, only A. hadrus and A. caccae demonstrated augmented butyrate production from L-sorbose or xylitol. These findings suggest that L-sorbose and xylitol cause prebiotic stimulation of the growth and metabolic activity of Anaerostipes spp. in the human colon.
Collapse
Affiliation(s)
- Tadashi Sato
- Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Shiro Kusuhara
- Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Wakae Yokoi
- Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Masahiko Ito
- Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Kouji Miyazaki
- Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| |
Collapse
|
37
|
Bypass graft infection and bacteremia caused by Anaerostipes caccae: First report of human infection caused by a recently described gut anaerobe. Anaerobe 2016; 42:98-100. [PMID: 27693544 DOI: 10.1016/j.anaerobe.2016.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/26/2016] [Indexed: 11/21/2022]
Abstract
We report a case of bypass graft infection and bacteremia caused by Anaerostipes caccae. A review of the literature shows no reported human infection caused by this microorganism to date. The patient was initially treated with vancomycin and piperacillin-tazobactam on admission and with amoxicillin-clavulanate upon discharge. The slow-growing organism was subsequently found to be susceptible to metronidazole and ertapenem.
Collapse
|
38
|
Etxeberria U, Hijona E, Aguirre L, Milagro FI, Bujanda L, Rimando AM, Martínez JA, Portillo MP. Pterostilbene-induced changes in gut microbiota composition in relation to obesity. Mol Nutr Food Res 2016; 61. [PMID: 27377854 DOI: 10.1002/mnfr.201500906] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 05/25/2016] [Accepted: 06/20/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Usune Etxeberria
- Department of Nutrition, Food Science and Physiology, and Centre for Nutrition Research University of Navarra; Pamplona Spain
| | - Elizabeth Hijona
- Department of Gastroenterology, Donostia Hospital and Biodonostia Institute; University of the Basque Country (UPV/EHU); San Sebastián Spain
- CIBER Hepatic and Digestive Pathologies (CIBERehd), Institute of Health Carlos III; Madrid Spain
| | - Leixuri Aguirre
- CIBER Obesity and Physiopathology of Nutrition (CIBERobn), Institute of Health Carlos III; Madrid Spain
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy; University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre; Vitoria Spain
| | - Fermin I. Milagro
- Department of Nutrition, Food Science and Physiology, and Centre for Nutrition Research University of Navarra; Pamplona Spain
- CIBER Obesity and Physiopathology of Nutrition (CIBERobn), Institute of Health Carlos III; Madrid Spain
| | - Luis Bujanda
- Department of Gastroenterology, Donostia Hospital and Biodonostia Institute; University of the Basque Country (UPV/EHU); San Sebastián Spain
- CIBER Hepatic and Digestive Pathologies (CIBERehd), Institute of Health Carlos III; Madrid Spain
| | - Agnes M. Rimando
- United States Department of Agriculture; Agricultural Research Service Natural Products Utilization Research Unit, University; MS USA
| | - José Alfredo Martínez
- Department of Nutrition, Food Science and Physiology, and Centre for Nutrition Research University of Navarra; Pamplona Spain
- CIBER Obesity and Physiopathology of Nutrition (CIBERobn), Institute of Health Carlos III; Madrid Spain
| | - María P. Portillo
- CIBER Obesity and Physiopathology of Nutrition (CIBERobn), Institute of Health Carlos III; Madrid Spain
- Nutrition and Obesity Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy; University of the Basque Country (UPV/EHU) and Lucio Lascaray Research Centre; Vitoria Spain
| |
Collapse
|
39
|
Arrazuria R, Elguezabal N, Juste RA, Derakhshani H, Khafipour E. Mycobacterium avium Subspecies paratuberculosis Infection Modifies Gut Microbiota under Different Dietary Conditions in a Rabbit Model. Front Microbiol 2016; 7:446. [PMID: 27065994 PMCID: PMC4815054 DOI: 10.3389/fmicb.2016.00446] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/18/2016] [Indexed: 12/18/2022] Open
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) the causative agent of paratuberculosis, produces a chronic granulomatous inflammation of the gastrointestinal tract of ruminants. It has been recently suggested that MAP infection may be associated with dysbiosis of intestinal microbiota in ruminants. Since diet is one of the key factors affecting the balance of microbial populations in the digestive tract, we intended to evaluate the effect of MAP infection in a rabbit model fed a regular or high fiber diet during challenge. The composition of microbiota of the cecal content and the sacculus rotundus was studied in 20 New Zealand white female rabbits. The extracted DNA was subjected to paired-end Illumina sequencing of the V3-V4 hypervariable region of the 16S rRNA gene for microbiota analysis. Microbial richness (Chao1) in the cecal content was significantly increased by MAP infection in regular diet rabbits (p = 0.0043) and marginally increased (p = 0.0503) in the high fiber group. Analysis of beta-diversity showed that MAP infection produces deeper changes in the microbiota of sacculus rotundus than in the cecal content. A lower abundance of Proteobacteria in the cecal content of infected animals fed the high fiber diet and also lower abundance of Bacteroidetes in the sacculus rotundus of infected animals fed the regular diet were observed. Based on OPLS-DA analysis, we observed that some bacteria repeatedly appear to be positively associated with infection in different samples under different diets (families Dehalobacteriaceae, Coriobacteriaceae, and Mogibacteriaceae; genus Anaerofustis). The same phenomenon was observed with some of the bacteria negatively associated with MAP infection (genera Anaerostipes and Coprobacillus). However, other groups of bacteria (Enterobacteriaceae family and ML615J-28 order) were positively associated with infection in some circumstances and negatively associated with infection in others. Data demonstrate that MAP infection and diet changes do interact and result in shifts in the microbiota of the cecal content and sacculus rotundus of rabbits.
Collapse
Affiliation(s)
- Rakel Arrazuria
- Department of Animal Health, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario Derio, Spain
| | - Natalia Elguezabal
- Department of Animal Health, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario Derio, Spain
| | - Ramon A Juste
- Department of Animal Health, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario Derio, Spain
| | - Hooman Derakhshani
- Department of Animal Science, University of Manitoba, Winnipeg MB, Canada
| | - Ehsan Khafipour
- Department of Animal Science, University of Manitoba, WinnipegMB, Canada; Department of Medical Microbiology, University of Manitoba, WinnipegMB, Canada
| |
Collapse
|
40
|
Nozu R, Ueno M, Hayashimoto N. Composition of fecal microbiota of laboratory mice derived from Japanese commercial breeders using 16S rRNA gene clone libraries. J Vet Med Sci 2016; 78:1045-50. [PMID: 26902692 PMCID: PMC4937141 DOI: 10.1292/jvms.15-0454] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The fecal microbiota of six mice derived from three Japanese commercial breeders was analyzed by using 16S
rRNA gene clone libraries to construct a database for analyzing the gut microbiota of laboratory mice. The 566
clones were obtained from the clone libraries generated from the fecal DNA samples derived from BALB/c,
C57BL/6N, DBA/2 and ICR mice. Among these 566 clones, there were 446 unique 16S rRNA gene sequences. When
grouped at the 98% similarity level, the 446 unique sequences consisted of 103 Clostridiales, 43
Bacteroidales, 5 Lactobacillus and 3 Erysipelotricaceae, as well as sequences from 11 other
phyla.
Collapse
Affiliation(s)
- Ryoko Nozu
- ICLAS Monitoring Center, Central Institute for Experimental Animals, 3-25-12 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| | | | | |
Collapse
|
41
|
Tao Y, Hu X, Zhu X, Jin H, Xu Z, Tang Q, Li X. Production of Butyrate from Lactate by a Newly Isolated Clostridium sp. BPY5. Appl Biochem Biotechnol 2016; 179:361-74. [DOI: 10.1007/s12010-016-1999-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/24/2016] [Indexed: 10/22/2022]
|
42
|
Effects of Xylo-Oligosaccharides on Broiler Chicken Performance and Microbiota. Appl Environ Microbiol 2015; 81:5880-8. [PMID: 26092452 DOI: 10.1128/aem.01616-15] [Citation(s) in RCA: 159] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/15/2015] [Indexed: 01/04/2023] Open
Abstract
In broiler chickens, feed additives, including prebiotics, are widely used to improve gut health and to stimulate performance. Xylo-oligosaccharides (XOS) are hydrolytic degradation products of arabinoxylans that can be fermented by the gut microbiota. In the current study, we aimed to analyze the prebiotic properties of XOS when added to the broiler diet. Administration of XOS to chickens, in addition to a wheat-rye-based diet, significantly improved the feed conversion ratio. XOS significantly increased villus length in the ileum. It also significantly increased numbers of lactobacilli in the colon and Clostridium cluster XIVa in the ceca. Moreover, the number of gene copies encoding the key bacterial enzyme for butyrate production, butyryl-coenzyme A (butyryl-CoA):acetate CoA transferase, was significantly increased in the ceca of chickens administered XOS. In this group of chickens, at the species level, Lactobacillus crispatus and Anaerostipes butyraticus were significantly increased in abundance in the colon and cecum, respectively. In vitro fermentation of XOS revealed cross-feeding between L. crispatus and A. butyraticus. Lactate, produced by L. crispatus during XOS fermentation, was utilized by the butyrate-producing Anaerostipes species. These data show the beneficial effects of XOS on broiler performance when added to the feed, which potentially can be explained by stimulation of butyrate-producing bacteria through cross-feeding of lactate and subsequent effects of butyrate on gastrointestinal function.
Collapse
|
43
|
Bruno-Barcena JM, Azcarate-Peril MA. Galacto-oligosaccharides and Colorectal Cancer: Feeding our Intestinal Probiome. J Funct Foods 2015; 12:92-108. [PMID: 25584074 PMCID: PMC4288025 DOI: 10.1016/j.jff.2014.10.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Prebiotics are ingredients selectively fermented by the intestinal microbiota that promote changes in the microbial community structure and/or their metabolism, conferring health benefits to the host. Studies show that β (1-4) galacto-oligosaccharides [β (1-4) GOS], lactulose and fructo-oligosaccharides increase intestinal concentration of lactate and short chain fatty acids, and stool frequency and weight, and they decrease fecal concentration of secondary bile acids, fecal pH, and nitroreductase and β-glucuronidase activities suggesting a clear role in colorectal cancer (CRC) prevention. This review summarizes research on prebiotics bioassimilation, specifically β (1-4) GOS, and their potential role in CRC. We also evaluate research that show that the impact of prebiotics on host physiology can be direct or through modulation of the gut intestinal microbiome, specifically the probiome (autochtonous beneficial bacteria), we present studies on a potential role in CRC progression to finally describe the current state of β (1-4) GOS generation for industrial production.
Collapse
Affiliation(s)
- Jose M. Bruno-Barcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina
| | - M. Andrea Azcarate-Peril
- Department of Cell Biology and Physiology, and Microbiome Core Facility, University of North Carolina School of Medicine, Chapel Hill
| |
Collapse
|
44
|
Bernard H, Desseyn JL, Bartke N, Kleinjans L, Stahl B, Belzer C, Knol J, Gottrand F, Husson MO. Dietary Pectin–Derived Acidic Oligosaccharides Improve the Pulmonary Bacterial Clearance ofPseudomonas aeruginosaLung Infection in Mice by Modulating Intestinal Microbiota and Immunity. J Infect Dis 2014; 211:156-65. [DOI: 10.1093/infdis/jiu391] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
45
|
Motelica-Wagenaar AM, Nauta A, van den Heuvel EGHM, Kleerebezem R. Flux analysis of the human proximal colon using anaerobic digestion model 1. Anaerobe 2014; 28:137-48. [PMID: 24880006 DOI: 10.1016/j.anaerobe.2014.05.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 04/30/2014] [Accepted: 05/19/2014] [Indexed: 02/07/2023]
Abstract
The colon can be regarded as an anaerobic digestive compartment within the gastro intestinal tract (GIT). An in silico model simulating the fluxes in the human proximal colon was developed on basis of the anaerobic digestion model 1 (ADM1), which is traditionally used to model waste conversion to biogas. Model calibration was conducted using data from in vitro fermentation of the proximal colon (TIM-2), and, amongst others, supplemented with the bio kinetics of prebiotic galactooligosaccharides (GOS) fermentation. The impact of water and solutes absorption by the host was also included. Hydrolysis constants of carbohydrates and proteins were estimated based on total short chain fatty acids (SCFA) and ammonia production in vitro. Model validation was established using an independent dataset of a different in vitro model: an in vitro three-stage continuous culture system. The in silico model was shown to provide quantitative insight in the microbial community structure in terms of functional groups, and the substrate and product fluxes between these groups as well as the host, as a function of the substrate composition, pH and the solids residence time (SRT). The model confirms the experimental observation that methanogens are washed out at low pH or low SRT-values. The in silico model is proposed as useful tool in the design of experimental setups for in vitro experiments by giving insight in fermentation processes in the proximal human colon.
Collapse
Affiliation(s)
- Anne Marieke Motelica-Wagenaar
- Delft University of Technology, Department of Biotechnology, Julianalaan 67, 2628BC Delft, The Netherlands; FrieslandCampina, Stationsplein 4, 3818 LE Amersfoort, The Netherlands.
| | - Arjen Nauta
- FrieslandCampina, Stationsplein 4, 3818 LE Amersfoort, The Netherlands.
| | - Ellen G H M van den Heuvel
- FrieslandCampina, Stationsplein 4, 3818 LE Amersfoort, The Netherlands; EMGO Institute for Health and Care Research, Department of Epidemiology and Biostatistics, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | - Robbert Kleerebezem
- Delft University of Technology, Department of Biotechnology, Julianalaan 67, 2628BC Delft, The Netherlands.
| |
Collapse
|
46
|
Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep 2014; 4:4548. [PMID: 24686447 PMCID: PMC4070089 DOI: 10.1038/srep04548] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 03/13/2014] [Indexed: 12/12/2022] Open
Abstract
Prevention of quality of life (QOL) deterioration is associated with the inhibition of geriatric diseases and the regulation of brain function. However, no substance is known that prevents the aging of both body and brain. It is known that polyamine concentrations in somatic tissues (including the brain) decrease with increasing age, and polyamine-rich foods enhance longevity in yeast, worms, flies, and mice, and protect flies from age-induced memory impairment. A main source of exogenous polyamines is the intestinal lumen, where they are produced by intestinal bacteria. We found that arginine intake increased the concentration of putrescine in the colon and increased levels of spermidine and spermine in the blood. Mice orally administered with arginine in combination with the probiotic bifidobacteria LKM512 long-term showed suppressed inflammation, improved longevity, and protection from age-induced memory impairment. This study shows that intake of arginine and LKM512 may prevent aging-dependent declines in QOL via the upregulation of polyamines.
Collapse
|
47
|
Keenan MJ, Janes M, Robert J, Martin RJ, Raggio AM, McCutcheon KL, Pelkman C, Tulley R, Goita M, Durham HA, Zhou J, Senevirathne RN. Resistant starch from high amylose maize (HAM-RS2) reduces body fat and increases gut bacteria in ovariectomized (OVX) rats. Obesity (Silver Spring) 2013; 21:981-4. [PMID: 23784900 PMCID: PMC4826615 DOI: 10.1002/oby.20109] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 07/29/2012] [Accepted: 09/19/2012] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Obesity after menopause is a health concern for older females. Changes in the microbiota are likely to occur with this condition. Modifying the microbiota with a prebiotic is a plausible strategy for improving the health of menopausal females. DESIGN AND METHODS Resistant starch type 2 from high-amylose maize (HAM-RS2) was used as a prebiotic in rats in a 2 × 2 factorial study with two levels of HAM-RS2 (0 or 29.7% of weight of diet) referred to as energy control (EC) and HAM-RS2 diets, respectively; and two levels of surgery, ovariectomized (OVX) and sham. RESULTS In a 6-week, postsurgery recovery period, OVX rats gained more body weight with consumption of a similar amount of food. Subsequently, consumption of HAM-RS2 versus EC diets resulted in reduced abdominal fat in both OVX and sham rats; but when normalized for disemboweled body weight (body weight minus GI tract), there was no effect of surgery, only reduction with HAM-RS2. Targeted bacterial populations were estimated that are known to ferment HAM-RS2 or metabolize the products of that initial fermentation. OVX and sham rats demonstrated increased bacterial levels with dietary HAM-RS2 for all bacteria. Additionally, culture techniques and qPCR provided similar results. CONCLUSION This study shows that, as expected, OVX increases adiposity. However, contrary to previous effects seen in obese mice, this did not prevent fermentation of HAM-RS2 and consequently, the fat gain associated with OVX was attenuated.
Collapse
Affiliation(s)
- Michael J Keenan
- Department of Human Ecology, Louisiana State University Agricultural Center, Baton Rouge, Louisiana, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Butyrate-producing bacteria, including mucin degraders, from the swine intestinal tract. Appl Environ Microbiol 2013; 79:3879-81. [PMID: 23584773 DOI: 10.1128/aem.00589-13] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
To identify bacteria with potential for influencing gut health, 980 anaerobes were cultured from the swine intestinal tract and analyzed for butyrate production. Fifteen isolates in the order Clostridiales produced butyrate and had butyryl coenzyme A (CoA):acetate CoA transferase activity. Three of the isolates grew on mucin, suggesting an intimate association with host intestinal mucosa.
Collapse
|
49
|
In vitro comparative evaluation of the impact of lacto-N-biose I, a major building block of human milk oligosaccharides, on the fecal microbiota of infants. Anaerobe 2013; 19:50-7. [DOI: 10.1016/j.anaerobe.2012.12.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/05/2012] [Accepted: 12/18/2012] [Indexed: 01/06/2023]
|
50
|
Nylund L, Satokari R, Nikkilä J, Rajilić-Stojanović M, Kalliomäki M, Isolauri E, Salminen S, de Vos WM. Microarray analysis reveals marked intestinal microbiota aberrancy in infants having eczema compared to healthy children in at-risk for atopic disease. BMC Microbiol 2013; 13:12. [PMID: 23339708 PMCID: PMC3563445 DOI: 10.1186/1471-2180-13-12] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 11/26/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Deviations in composition and diversity of intestinal microbiota in infancy have been associated with both the development and recurrence of atopic eczema. Thus, we decided to use a deep and global microarray-based method to characterize the diversity and temporal changes of the intestinal microbiota in infancy and to define specific bacterial signatures associated with eczema. Faecal microbiota at 6 and 18 months of age were analysed from 34 infants (15 with eczema and 19 healthy controls) selected from a prospective follow-up study based on the availability of faecal samples. The infants were originally randomized to receive either Lactobacillus rhamnosus GG or placebo. RESULTS Children with eczema harboured a more diverse total microbiota than control subjects as assessed by the Simpson's reciprocal diversity index of the microarray profiles. Composition of the microbiota did not differ between study groups at age of 6 months, but was significantly different at age of 18 months as assessed by MCPP (p=0.01). At this age healthy children harboured 3 -fold greater amount of members of the Bacteroidetes (p=0.01). Microbiota of children suffering from eczema had increased abundance of the Clostridium clusters IV and XIVa, which are typically abundant in adults. Probiotic Lactobacillus rhamnosus GG supplementation in early infancy was observed to have minor long-term effects on the microbiota composition. CONCLUSION A diverse and adult-type microbiota in early childhood is associated with eczema and it may contribute to the perpetuation of eczema.
Collapse
Affiliation(s)
- Lotta Nylund
- Functional Foods Forum, University of Turku, Turku, FI-20014, Finland.
| | | | | | | | | | | | | | | |
Collapse
|