51
|
Collinet A, Grimm P, Julliand S, Julliand V. Multidimensional Approach for Investigating the Effects of an Antibiotic-Probiotic Combination on the Equine Hindgut Ecosystem and Microbial Fibrolysis. Front Microbiol 2021; 12:646294. [PMID: 33841371 PMCID: PMC8027512 DOI: 10.3389/fmicb.2021.646294] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
The equine hindgut ecosystem is specialized in dietary fibers' fermentation to provide horses' energy and contribute to its health. Nevertheless, antibiotics are known to disrupt the hindgut microbiota, affecting the fibrolytic activity of bacteria and the intestinal immune balance, leading to diseases. This in vivo study used a general and comprehensive approach for characterizing the hindgut ecosystem of 9 healthy horses over 28 days in response to a 5-day challenge with oral trimethoprim-sulfadiazine (TMS), with a special emphasis on microbial fibrolytic activity and the host immune response. Horses were supplemented with two doses of Lactobacillus acidophilus, Ligilactobacillus salivarius (formerly L. salivarius), and Bifidobacterium lactis blend or a placebo in a 3 × 3 Latin square design. Changes in fecal microbiota were investigated using 16S rRNA sequencing. Clostridioides difficile was quantified in feces using quantitative polymerase chain reaction. Anaerobic microbiological culture was used to enumerate functional bacterial groups (cellulolytic, amylolytic, and lactic acid-utilizing). The environmental dimensions were assessed by measuring the concentrations of volatile fatty acids (VFAs) and lactic acid using biochemical methods, and changes in pH and dry matter weight. Systemic and local inflammation was evaluated by determination of cytokine and immunoglobulin (Ig)A concentrations in the serum and secretory IgA (SIgA) concentrations in the feces using immuno-enzymatic methods. Oral TMS treatment strongly altered the whole hindgut ecosystem by 2 days after the first administration. Bacterial diversity decreased in proportion to the relative abundance of fibrolytic genera, which coincided with the decrease in the concentration of cellulolytic bacteria. At the same time, the composition of microbiota members was reorganized in terms of relative abundances, probably to support the alteration in fibrolysis. C. difficile DNA was not found in these horses, but the relative abundances of several potential pathobiont genera increased. 2 days after the first TMS administration, fecal concentrations of VFAs and SIgA increased in parallel with fecal water content, suggesting an alteration of the integrity of the hindgut mucosa. Recovery in bacterial composition, functions, and immune biomarkers took 2-9 days after the end of TMS administration. Supplementation with this bacterial blend did not limit bacterial alteration but might have interesting mucosal immunomodulatory effects.
Collapse
Affiliation(s)
- Axelle Collinet
- Lab To Field, Dijon, France.,Univ. Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, Dijon, France
| | | | | | - Véronique Julliand
- Univ. Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, Dijon, France
| |
Collapse
|
52
|
Lactobacillus animalis pZL8a: a potential probiotic isolated from pig feces for further research. 3 Biotech 2021; 11:132. [PMID: 33680697 DOI: 10.1007/s13205-021-02681-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/05/2021] [Indexed: 12/13/2022] Open
Abstract
This study aimed to screen a potential anti-diarrheal probiotic for pigs to meet the growing demand for antibiotic alternatives in livestock. Six intestinal pathogens, Escherichia coli (O157: H7) ATCC 43888, Staphylococcus aureus ATCC 6538, Listeria monocytogenes ATCC 19115, Salmonella Typhimurium ATCC 14028, Shigella boydii ATCC 9207, and Staphylococcus haemolyticus ZSY2 were employed as indicator bacteria. Our result showed that Lactobacillus animalis pZL8a isolated from pig feces had extensive and higher antibacterial activity against indicator pathogens among 9 tested strains. In addition, valuable attributes of pZL8a such as great tolerance of low pH (3.0) and bile salts (0.3%), high-level adhesion to Caco-2 cells, and similar susceptibility to the reference strain Lactobacillus rhamnosus GG (LGG) were observed. Compared with control, pZL8a supplement significantly improved the level of immunoglobulin G (IgG), immunoglobulin M (IgM), and interleukin-2 (IL-2) in mouse serum. Therefore, L. animalis pZL8a was proposed as a potential probiotic for further research and hope to reduce or replace the application of antibiotics in animal production.
Collapse
|
53
|
Yang L, Zhang Q, Huang J, Liu D, Lan Y, Yuan L, Chen Q. Xianglian Pill ameliorates antibiotic-associated diarrhea by restoring intestinal microbiota and attenuating mucosal damage. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113377. [PMID: 32920136 DOI: 10.1016/j.jep.2020.113377] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 08/13/2020] [Accepted: 09/06/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xianglian Pill (XLP), a traditional Chinese pharmaceutical preparation for the treatment of gastrointestinal disease, possessing anti-inflammatory, anti-microbial and analgesic activities, may represent a promising candidate for the treatment of antibiotic-associated diarrhea (AAD). AIM OF THE STUDY This study aimed to unravel the underlying mechanism of XLP on the amelioration of AAD. MATERIALS AND METHODS AAD was induced by intragastric administration of a mixture of cefuroxime and levofoxacin (300 mg/kg. bw + 200 mg/kg. bw) for five consecutive days. Then AAD mice were treated with XLP at the dose of 500, 1000 and 2000 mg/kg. bw, respectively for 5 days. The physical manifestations, diarrhea status were monitored during the drug delivery. Histopathology of colon, intestinal microbiota, inflammatory cytokines, tight junction protein and short chain fat acids (SCFAs) were determined. RESULTS Mice received cefuroxime and levofoxacin for 5 days developed medium to severe diarrhea. XLP treatment, however, mitigated the diarrhea status. Further evaluation revealed that XLP promoted the recovery of mucosa, maintained the integrity of tight junction, attenuated the inflammatory disorders, restored intestinal microbiota and increased SCFAs level in feces. CONCLUSION XLP ameliorates AAD by restoring intestinal microbiota and attenuating mucosal damage.
Collapse
Affiliation(s)
- Lujia Yang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Qian Zhang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Jieyao Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Danning Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Yunfei Lan
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China
| | - Lujiang Yuan
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| | - Qianfeng Chen
- College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
54
|
Yang S, Xie X, Ma J, He X, Li Y, Du M, Li L, Yang L, Wu Q, Chen W, Zhang J. Selective Isolation of Bifidobacterium From Human Faeces Using Pangenomics, Metagenomics, and Enzymology. Front Microbiol 2021; 12:649698. [PMID: 33967985 PMCID: PMC8096985 DOI: 10.3389/fmicb.2021.649698] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Bifidobacterium, an important genus for human health, is difficult to isolate. We applied metagenomics, pangenomics, and enzymology to determine the dominant glycoside hydrolase (GH) families of Bifidobacterium and designed selective medium for Bifidobacterium isolation. Pangenomics results showed that the GH13, GH3, GH42, and GH43 families were highly conserved in Bifidobacterium. Metagenomic analysis of GH families in human faecal samples was performed. The results indicated that Bifidobacterium contains core GHs for utilizing raffinose, D-trehalose anhydrous, D(+)-cellobiose, melibiose, lactulose, lactose, D(+)-sucrose, resistant starch, pullulan, xylan, and glucan. These carbohydrates as the main carbon sources were applied for selective media, which were more conducive to the growth of bifidobacteria. In the medium with lactose, raffinose and xylan as the main carbon sources, the ratio of cultivable bifidobacteria to cultivable microorganisms were 89.39% ± 2.50%, 71.45% ± 0.99%, and 53.95% ± 1.22%, respectively, whereas the ratio in the ordinary Gifu anaerobic medium was only 17.90% ± 0.58%. Furthermore, the species significantly (p < 0.05) varied among samples from different individuals. Results suggested that xylan might be a prebiotic that benefits host health, and it is feasible to screen and isolate bifidobacteria using the oligosaccharides corresponding to the specific GHs of bifidobacteria as the carbon sources of the selective media.
Collapse
Affiliation(s)
- Shuanghong Yang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jun Ma
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Xingxiang He
- Department of Gastroenterology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Mingzhu Du
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Lingshuang Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Wei Chen
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
55
|
Hao Y, Wang X, Yuan S, Wang Y, Liao X, Zhong M, He Q, Shen H, Liao W, Shen J. Flammulina velutipes polysaccharide improves C57BL/6 mice gut health through regulation of intestine microbial metabolic activity. Int J Biol Macromol 2020; 167:1308-1318. [PMID: 33202270 DOI: 10.1016/j.ijbiomac.2020.11.085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 01/07/2023]
Abstract
Flammulina velutipes polysaccharides (FVP) can improve gut health through gut microbiota and metabolism regulation. In this study, the 28-days fed experiment was used to investigate gut microbime and metabolic profiling induced by FVP. After treatment, intestinal tissue section showed the higher villus height and villus height/crypt depth (V/C) value in FVP-treated group. The 16 s rRNA gene sequencing revealed microbiota composition alteration caused by FVP, as the Firmicutes phylum increased while Bacteroidetes phylum slightly decreased. The metabolic profiling was detected by LC/MS and results showed 56 and 99 compounds were dramatically changed after FVP treatment in positive and negative ion mode, respectively. Annotation in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways displayed the adjustment of energy metabolism, amino acid metabolism, nucleotide metabolism and other related basic pathways after FVP treatment. Our study suggested that FVP can be developed as a dietary supplement for intestine health promotion.
Collapse
Affiliation(s)
- Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Xiangdong Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Sijie Yuan
- Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), No.1 of Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong Province, China
| | - Yingyi Wang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Xiaoshan Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Meiling Zhong
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Qiangnan He
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Haibin Shen
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, China.
| | - Jie Shen
- Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), No.1 of Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong Province, China.
| |
Collapse
|
56
|
Mulder M, Radjabzadeh D, Kiefte-de Jong JC, Uitterlinden AG, Kraaij R, Stricker BH, Verbon A. Long-term effects of antimicrobial drugs on the composition of the human gut microbiota. Gut Microbes 2020; 12:1795492. [PMID: 32991820 PMCID: PMC7781642 DOI: 10.1080/19490976.2020.1791677] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [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
INTRODUCTION Antimicrobial drugs are known to have effects on the human gut microbiota. We studied the long-term temporal relationship between several antimicrobial drug groups and the composition of the human gut microbiota determined in feces samples. METHODS Feces samples were obtained from a community-dwelling cohort of middle-aged and elderly individuals (Rotterdam Study). Bacterial DNA was isolated and sequenced using V3/V4 16 S ribosomal RNA sequencing (Illumina MiSeq). The time between the last prescription of several antimicrobial drug groups and the day of sampling was categorized into 0-12, 12-24, 24-48 and >48 months. The effects of the antimicrobial drug groups on the Shannon alpha-diversity (diversity), the Bray-Curtis beta-diversity (community structure), the Firmicutes/Bacteroidetes (F/B) ratio and individual genera were determined. RESULTS We studied the gut microbiota of 1413 individuals (57.5% female, median age 62.6 years). The alpha-diversity was significantly lower up to 4 years after prescriptions of macrolides and lincosamides. It was also lower in the first year after the use of beta-lactams. The community structure (beta-diversity) of the microbiota was significantly different up to 4 years for macrolides and lincosamides, the first year for beta-lactams and at least the first year for quinolones. For the F/B ratio, drugs with a high anaerobic activity shifted the ratio toward Firmicutes in the first year whereas other antimicrobial drugs shifted the ratio toward Bacteroidetes. CONCLUSION Use of antimicrobial drugs is associated with a shift in the composition of the gut microbiota.These effects differ in strength and duration, depending on the antimicrobial drug group used. These findings should be considered when prescribing antimicrobial drugs.
Collapse
Affiliation(s)
- M. Mulder
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands,Youth and Healthcare Inspectorate, Heerlen, The Netherlands
| | - D. Radjabzadeh
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - J. C. Kiefte-de Jong
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands,Department of Public Health and Primary Care, LUMC, The Hague, The Netherlands
| | - A. G. Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands,Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - R. Kraaij
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - B. H. Stricker
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands,Youth and Healthcare Inspectorate, Heerlen, The Netherlands,Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands,CONTACT B. H. Stricker Department of Epidemiology, Erasmus MC Rotterdam, PO Box 2040, Rotterdam, CA 3000, The Netherlands
| | - A. Verbon
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands,Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
57
|
Figueroa LM, Ramírez-Jiménez AK, Senés-Guerrero C, Santacruz A, Pacheco A, Gutiérrez-Uribe JA. Assessment of the bacterial diversity of agave sap concentrate, resistance to in vitro gastrointestinal conditions and short-chain fatty acids production. Food Res Int 2020; 140:109862. [PMID: 33648180 DOI: 10.1016/j.foodres.2020.109862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/12/2020] [Accepted: 10/30/2020] [Indexed: 10/23/2022]
Abstract
Low bacterial diversity in the gut has been associated with the development of several diseases. Agave sap concentrate (ASC) is obtained from the thermal treatment of the fresh sap called "aguamiel", an artisanal Mexican food. In this study, we assessed the microbial diversity from three different ASC producing regions in Mexico using high-throughput sequencing of the 16S rRNA gene and evaluated their resistance to an in vitro gastrointestinal process as well as their ability to produce short-chain fatty acids (SCFA). Seven phyla and 120 genera were detected in ASC samples; Firmicutes had the highest relative read abundance at the phylum level, whereas Bacillus was the most abundant genus. Bacterial diversity at phylum and genus levels was highly dependent on the region where ASC was produced. The microbiota from a selected sample was resistant to low pH conditions, bile salts and intestinal enzymes. Moreover, bacteria were able to survive and grow in the colonic environment. SCFA production was comparable with that observed for a well-known probiotic, Lactobacillus plantarum 299v, that was used as control. These findings demonstrate that ASC contains a bacterial ecosystem with potential probiotic benefits.
Collapse
Affiliation(s)
- Luis M Figueroa
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849 Monterrey, Nuevo León, Mexico
| | - Aurea K Ramírez-Jiménez
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Toluca, Avenida Eduardo Monroy Cárdenas 2000 San Antonio Buenavista, 50110 Toluca de Lerdo, Mexico
| | - Carolina Senés-Guerrero
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849 Monterrey, Nuevo León, Mexico
| | - Arlette Santacruz
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849 Monterrey, Nuevo León, Mexico
| | - Adriana Pacheco
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849 Monterrey, Nuevo León, Mexico
| | - Janet A Gutiérrez-Uribe
- Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Centro de Biotecnología-FEMSA, Av. Eugenio Garza Sada 2501 Sur, C.P. 64849 Monterrey, Nuevo León, Mexico; Tecnologico de Monterrey, Escuela de Ingeniería y Ciencias, Campus Puebla, Vía Atlixcayotl 5718, C.P. 72453 Puebla, Puebla, Mexico.
| |
Collapse
|
58
|
Hu JS, Huang YY, Kuang JH, Yu JJ, Zhou QY, Liu DM. Streptococcus thermophiles DMST-H2 Promotes Recovery in Mice with Antibiotic-Associated Diarrhea. Microorganisms 2020; 8:microorganisms8111650. [PMID: 33114373 PMCID: PMC7693992 DOI: 10.3390/microorganisms8111650] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/23/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) is the most common side effect of antibiotics and is routinely treated with probiotics in clinical. Streptococcus thermophiles, extensively utilized for producing dairy foods, has recently been regarded as a new promising probiotic candidate. In this study, the efficacy of Streptococcus thermophiles DMST-H2 (DMST-H2) for AAD treatment in mice was investigated. DMST-H2 was isolated from Chinese traditional yogurt, proved to be non-toxic, and presented tolerance against simulated gastrointestinal conditions in vitro. Additionally, genomic analysis revealed that it possessed genes related to acid tolerance, bile salt tolerance, adhesion, oxidative stress and bacteriocin production. The animal experiment results showed that both DMST-H2 treatment and natural recovery could reduce fecal water content. Compared with spontaneous recovery, DMST-H2 accelerated the recovery of the enlarged caecum and intestinal barrier injury from AAD, and further decreased endotoxin (ET), D-lactate (D-LA) and diamine oxidase (DAO) content in serum. Moreover, pro-inflammatory cytokines (TNF-α) were reduced, while interferon-γ (IFN-γ) and anti-inflammatory cytokines (IL-10) increased after treating with DMST-H2. Furthermore, DMST-H2 better restored the structure of intestinal flora. At the phylum level, Firmicutes increased and Proteobacteria decreased. These findings indicate that DMST-H2 could promote recovery in mice with antibiotic-associated diarrhea.
Collapse
|
59
|
Bailey MJ, Naik NN, Wild LE, Patterson WB, Alderete TL. Exposure to air pollutants and the gut microbiota: a potential link between exposure, obesity, and type 2 diabetes. Gut Microbes 2020; 11:1188-1202. [PMID: 32347153 PMCID: PMC7524284 DOI: 10.1080/19490976.2020.1749754] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Work has shown that increased exposure to air pollutants independently contributes to obesity and type 2 diabetes risk, yet the exact mechanisms underlying these associations have not been fully characterized. The current review summarizes recent findings regarding the impact of inhaled and ingested air pollutants on the gut microbiota. Animal and human studies provide evidence that air pollutants, such as particulate matter, nitrogen oxides, and ozone, have the potential to alter the gut microbiota. Further, studies suggest that such exposure-induced alterations to the gut microbiota may contribute to increased risk for obesity and type 2 diabetes through inflammatory pathways. Future work is needed to fully understand the complex interactions between air pollution, the gut microbiome, and human health. Additionally, advanced sequencing methods for gut microbiome research present unique opportunities to study the underlying pathways that link increased air pollution exposure with obesity and type 2 diabetes risk.
Collapse
Affiliation(s)
- Maximillian J. Bailey
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Noopur N. Naik
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Laura E. Wild
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA,CONTACT Tanya L. Alderete University of Colorado Boulder, Department of Integrative Physiology, Ramaley Biology Building, 1800 Colorado Avenue, N379, Boulder, CO80309
| |
Collapse
|
60
|
Shao H, Zhang C, Wang C, Tan Z. Intestinal mucosal bacterial diversity of antibiotic-associated diarrhea (AAD) mice treated with Debaryomyces hansenii and Qiweibaizhu powder. 3 Biotech 2020; 10:392. [PMID: 32832342 DOI: 10.1007/s13205-020-02383-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023] Open
Abstract
The aim was to investigate the combined effect of Debaryomyces hansenii and Qiweibaizhu powder (QWBZP) on the bacterial diversity of the intestinal mucosa of antibiotic-associated diarrhea (AAD) mice, for the potential treatment of diarrhea, especially which is induced by administration of antibiotics. Eighteen (18) mice were randomly assigned to three equal groups of six mice, namely Normal (mn group), Placebo control (mm group) and D. hansenii and QWBZP (DQ) treatment (mdq group). Mice were gavaged with a solution (23.33 mL·kg-1·day-1) consisting of gentamicin and cefradine to establish AAD. The DQ treatment group was gavaged with DQ for 4 days, and sterile water was used as a placebo control. The metagenome DNA of the intestinal mucosal microbiota was extracted, and the 16S rRNA gene was sequenced. Analysis showed that there were 288 OTUs for the normal group, 443 for the placebo control group, and 229 for the DQ treatment group. Phylogenetically, the gut microbiota of the DQ treatment group and the normal group were closer to each other than to the placebo control group. Both the DQ and placebo-treated groups included Stenotrophomonas, Robinsoniella, Bacteroidales S24-7 group norank, Citrobacter, and Glutamicibacter, but their abundances were significantly higher in the DQ treatment group than in the placebo control group. This suggested that the combined use of D. hansenii and QWBZP overcame the influence of dysbacteriosis and could lead to the recovery of intestinal mucosal microbiota homeostasis. This positive effect is likely related to short-chain fatty acid (SCFA)-producing bacteria, such as members of Micrococcaceae, Lachnospiraceae, and Bacteroidales S24-7 group, which could play beneficial roles in protecting the mucosal barrier and stimulating the immune response in mice.
Collapse
|
61
|
Vallianou N, Stratigou T, Christodoulatos GS, Tsigalou C, Dalamaga M. Probiotics, Prebiotics, Synbiotics, Postbiotics, and Obesity: Current Evidence, Controversies, and Perspectives. Curr Obes Rep 2020; 9:179-192. [PMID: 32472285 DOI: 10.1007/s13679-020-00379-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In this review, we summarize current evidence on gut microbiome and obesity; we discuss the role of probiotics, prebiotics, synbiotics, and postbiotics in obesity prevention and management; and we highlight and analyze main limitations, challenges, and controversies of their use. RECENT FINDINGS Overall, the majority of animal studies and meta-analyses of human studies examining the use of probiotics and synbiotics in obesity has shown their beneficial effects on weight reduction and other metabolic parameters via their involvement in gut microbiota modulation. Bifidobacterium and Lactobacillus strains are still the most widely used probiotics in functional foods and dietary supplements, but next generation probiotics, such as Faecalibacterium prausnitzii, Akkermansia muciniphila, or Clostridia strains, have demonstrated promising results. On the contrary, meta-analyses of human studies on the use of prebiotics in obesity have yielded contradictory results. In animal studies, postbiotics, mainly short-chain fatty acids, may increase energy expenditure through induction of thermogenesis in brown adipose tissue as well as browning of the white adipose tissue. The main limitations of studies on biotics in obesity include the paucity of human studies; heterogeneity among the studied subgroups regarding age, gender, and lifestyle; and use of different agents with potential therapeutic effects in different formulations, doses, ratio and different pharmacodynamics/pharmacokinetics. In terms of safety, the supplementation with prebiotics, probiotics, and synbiotics has not been associated with serious adverse effects among immune-competent individuals, with the exception of the use of probiotics and synbiotics in immunocompromised patients. Further large-scale Randomized Controlled Trials (RCTs) in humans are required to evaluate the beneficial properties of probiotics, prebiotics, synbiotics, and postbiotics; their ideal dose; the duration of supplementation; and the durability of their beneficial effects as well as their safety profile in the prevention and management of obesity.
Collapse
Affiliation(s)
- Natalia Vallianou
- Department of Endocrinology, 'Evangelismos' General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece.
| | - Theodora Stratigou
- Department of Endocrinology, 'Evangelismos' General Hospital of Athens, 45-47 Ypsilantou street, 10676, Athens, Greece
| | - Gerasimos Socrates Christodoulatos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Medical School, Democritus University of Thrace, 6th Km Alexandroupolis-Makri, Alexandroupolis, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527, Athens, Greece
| |
Collapse
|
62
|
Ojima MN, Gotoh A, Takada H, Odamaki T, Xiao JZ, Katoh T, Katayama T. Bifidobacterium bifidum Suppresses Gut Inflammation Caused by Repeated Antibiotic Disturbance Without Recovering Gut Microbiome Diversity in Mice. Front Microbiol 2020; 11:1349. [PMID: 32625197 PMCID: PMC7314955 DOI: 10.3389/fmicb.2020.01349] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiome is a dynamic community that significantly affects host health; it is frequently disturbed by medications such as antibiotics. Recently, probiotics have been proposed as a remedy for antibiotic-induced dysbiosis, but the efficacy of such treatments remains uncertain. Thus, the effect of specific antibiotic-probiotic combinations on the gut microbiome and host health warrants further research. We tested the effect vancomycin, amoxicillin, and ciprofloxacin on mice. Antibiotic administration was followed by one of the following recovery treatments: Bifidobacterium bifidum JCM 1254 as a probiotic (PR); fecal transplant (FT); or natural recovery (NR). Each antibiotic administration and recovery treatment was repeated three times over 9 weeks. We used the Shannon Index and Chao1 Index to determine gut microbiome diversity and assessed recovery by quantifying the magnitude of microbial shift using the Bray-Curtis Index of Dissimilarity. We determined the community composition by sequencing the V3-V4 regions of the 16S ribosomal RNA gene. To assess host health, we measured body weight and cecum weight, as well as mRNA expression of inflammation-related genes by reverse-transcription quantitative PCR. Our results show that community response varied by the type of antibiotic used, with vancomycin having the most significant effects. As a result, the effect of probiotics and fecal transplants also varied by antibiotic type. For vancomycin, the first antibiotic disturbance substantially increased the relative abundance of inflammatory species in the phylum Proteobacteria, such as Proteus, but the effect of subsequent disturbances was less pronounced, suggesting that the gut microbiome is affected by past disturbance events. Furthermore, although gut microbiome diversity did not recover, probiotic supplementation was effective in limiting cecum size enlargement and colonic inflammation caused by vancomycin. However, for amoxicillin and ciprofloxacin, the relative abundances of proinflammatory species were not greatly affected, and consequently, the effect of probiotic supplementation on community structure, cecum weight, and expression of inflammation-related genes was comparatively negligible. These results indicate that probiotic supplementation is effective, but only when antibiotics cause proinflammatory species-induced gut inflammation, suggesting that the necessity of probiotic supplementation is strongly influenced by the type of disturbance introduced to the community.
Collapse
Affiliation(s)
- Miriam N. Ojima
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Aina Gotoh
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiromi Takada
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Toshitaka Odamaki
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Jin-Zhong Xiao
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Toshihiko Katoh
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takane Katayama
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
63
|
Liu D, Zeng L, Yan Z, Jia J, Gao J, Wei Y. The mechanisms and safety of probiotics against toxigenic clostridium difficile. Expert Rev Anti Infect Ther 2020; 18:967-975. [PMID: 32520637 DOI: 10.1080/14787210.2020.1778464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Toxigenic Clostridium difficile (C. difficile) is the main cause of antibiotic-associated diarrhea and can induce pseudomembranous colitis and infrequent toxic megacolon, which are potentially fatal. The standard antibiotic therapy for C. difficile infection (CDI) is limited by antibiotics' broad spectrum and further disruptive effects on indigenous microbiota. Probiotics may offer a prospective and alternative strategy for the prevention and treatment of CDI. AREAS COVERED In this article, the mechanisms implying the probiotic effect against C. difficile and the safety profile highlighting the patient groups with inappropriate application of probiotics were reviewed from 2015 to 2020. EXPERT OPINION Although many strains with ability against C. difficile have been reported, the usage of probiotics for CDI prevention and/or treatment is scarce since the number of clinical trials is not sufficient to prove probiotics' efficacy and safety in CDI treatment, especially for premature infant and immunocompromised patient. Especially, there are few well-defined clinical studies supporting safety of probiotics for CDI. A few strains from Lactobacillus and Saccharomyces genus have been studied more extensively than other probiotic strains through clinical trials for CDI. Thus, more clinical intervention studies regarding the benefit and the comprehensive safety assessments of probiotics for CDI are needed.
Collapse
Affiliation(s)
- Dianbin Liu
- School of Stomatology/Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University , Xuzhou, Jiangsu Province, China
| | - Lingbing Zeng
- Department of clinical microbiology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi Province, China
| | - Zhihan Yan
- School of Stomatology/Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University , Xuzhou, Jiangsu Province, China
| | - Junqi Jia
- School of Stomatology/Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University , Xuzhou, Jiangsu Province, China
| | - Jing Gao
- School of Stomatology/Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University , Xuzhou, Jiangsu Province, China
| | - Yanxia Wei
- School of Stomatology/Department of Pathogenic Biology and Immunology, Jiangsu Key Laboratory of Immunity and Metabolism, Xuzhou Medical University , Xuzhou, Jiangsu Province, China
| |
Collapse
|
64
|
Effects of Antibiotics on the Intestinal Microbiota of Mice. Antibiotics (Basel) 2020; 9:antibiotics9040191. [PMID: 32316518 PMCID: PMC7235770 DOI: 10.3390/antibiotics9040191] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
Studies on human and mouse gastrointestinal microbiota have correlated the composition of the microbiota to a variety of diseases, as well as proved it vital to prevent colonization with resistant bacteria, a phenomenon known as colonization resistance. Antibiotics dramatically modify the gut community and there are examples of how antibiotic usage lead to colonization with resistant bacteria [e.g., dicloxacillin usage selecting for ESBL-producing E. coli carriage], as shown by Hertz et al. Here, we investigated the impact of five antibiotics [cefotaxime, cefuroxime, dicloxacillin, clindamycin, and ciprofloxacin] on the intestinal microbiota in mice. Five different antibiotics were each given to groups of five mice. The intestinal microbiotas were profiled by use of the IS-pro analysis; a 16S–23S rDNA interspace [IS]-region-based profiling method. For the mice receiving dicloxacillin and clindamycin, we observed dramatic shifts in dominating phyla from day 1 to day 5. Of note, diversity increased, but overall bacterial load decreased. For ciprofloxacin, cefotaxime, and cefuroxime there were few overall changes. We speculate that antibiotics with efficacy against the abundant anaerobes in the gut, particularly Bacteroidetes, can in fact be selected for resistant bacteria, disregarding the spectrum of activity.
Collapse
|
65
|
Xie G, Wu Y, Zheng T, Shen K, Tan Z. Effect of Debaryomyces hansenii combined with Qiweibaizhu powder extract on the gut microbiota of antibiotic-treated mice with diarrhea. 3 Biotech 2020; 10:127. [PMID: 32140379 DOI: 10.1007/s13205-020-2121-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
The aim of this study was to investigate the effects of an extract of Qiweibaizhu powder combined with Debaryomyces hansenii on the gut microbiota of antibiotic-treated mice with diarrhea. Mice were gavaged with a mixture of gentamycin sulfate and cefradine to induce diarrhea. After diarrhea was observed, 25% dose of ultra-micro Qiweibaizhu powder extract combined with 25% dose of Debaryomyces hansenii (QCD) was gavaged to mice with diarrhea. DNA of intestinal contents in mice was extracted for 16S rRNA gene sequence analysis by high-throughput sequencing following treatment finished. The results showed that the QCD increased the species richness and diversity, but did not recover the diversity to the original level. Antibiotics and QCD significantly altered the composition of gut microbiota at different taxonomic levels. At the genus level, the relative abundance of Bacteroidales S24-7 group_unidentified and Bacteroides returned to baseline after QCD treatment. Additionally, QCD suppressed the growth of Oscillospira and Ruminococcus, and promoted the proliferation of Erysipelotrichaceae_norank and Blautia compared with the healthy and diarrheal mice. Our results indicated that QCD modulated the diversity and composition of the gut microbiota in antibiotic-treated mice with diarrhea. The synergistic effect between Qiweibaizhu powder extract and Debaryomyces hansenii may be related to Bifidobacterium and Bacteroidales S24-7 group_unidentified.
Collapse
Affiliation(s)
- Guozhen Xie
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan China
| | - Yi Wu
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan China
| | - Tao Zheng
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan China
| | - Kejia Shen
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan China
| | - Zhoujin Tan
- Hunan University of Chinese Medicine, Xueshi Road 300, Yuelu District, Changsha, 410208 Hunan China
| |
Collapse
|
66
|
Mekonnen SA, Merenstein D, Fraser CM, Marco ML. Molecular mechanisms of probiotic prevention of antibiotic-associated diarrhea. Curr Opin Biotechnol 2020; 61:226-234. [PMID: 32087535 DOI: 10.1016/j.copbio.2020.01.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 02/08/2023]
Abstract
Antibiotic-associated diarrhea (AAD) is a common and unintended adverse effect of antibiotic treatment. It is characterized by the disruption of the gut microbiota, decreased intestinal short chain fatty acid (SCFA) concentrations, accumulation of luminal carbohydrates and colonic bile acids, altered water absorption, and ultimately diarrhea. Probiotics were shown to prevent AAD in numerous clinical trials. This review examines what is currently known about how probiotics reduce the risk for AAD via modulating the gut microbiota, altering nutrient and bile acid metabolism, inducing epithelial solute transporter activity, supporting intestinal barrier function, and influencing the immune system. Although probiotics are frequently prescribed with antibiotic use, mechanistic evidence verifying how they confer protection against AAD is extremely limited. This information is urgently needed for improving recommendations for sustaining probiotic development and for implementing probiotics in clinical settings.
Collapse
Affiliation(s)
- Solomon A Mekonnen
- Department of Food Science and Technology, University of California, Davis, CA, USA
| | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Claire M Fraser
- Department of Medicine, Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, CA, USA.
| |
Collapse
|
67
|
Pan M, Barrangou R. Combining omics technologies with CRISPR-based genome editing to study food microbes. Curr Opin Biotechnol 2020; 61:198-208. [DOI: 10.1016/j.copbio.2019.12.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/23/2019] [Accepted: 12/25/2019] [Indexed: 12/22/2022]
|
68
|
Mitra A, Grossman Biegert GW, Delgado AY, Karpinets TV, Solley TN, Mezzari MP, Yoshida-Court K, Petrosino JF, Mikkelson MD, Lin L, Eifel P, Zhang J, Ramondetta LM, Jhingran A, Sims TT, Schmeler K, Okhuysen P, Colbert LE, Klopp AH. Microbial Diversity and Composition Is Associated with Patient-Reported Toxicity during Chemoradiation Therapy for Cervical Cancer. Int J Radiat Oncol Biol Phys 2020; 107:163-171. [PMID: 31987960 DOI: 10.1016/j.ijrobp.2019.12.040] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Patients receiving pelvic radiation for cervical cancer experience high rates of acute gastrointestinal (GI) toxicity. The association of changes in the gut microbiome with bowel toxicity from radiation is not well characterized. METHODS AND MATERIALS Thirty-five patients undergoing definitive chemoradiation therapy (CRT) underwent longitudinal sampling (baseline and weeks 1, 3, and 5) of the gut microbiome and prospective assessment of patient-reported GI toxicity. DNA was isolated from stool obtained at rectal examination and analyzed with 16S rRNA sequencing. GI toxicity was assessed with the Expanded Prostate Cancer Index Composite instrument to evaluate frequency, urgency, and discomfort associated with bowel function. Shannon diversity index was used to characterize alpha (within sample) diversity. Weighted UniFrac principle coordinates analysis was used to compare beta (between sample) diversity between samples using permutational multivariate analysis of variance. Linear discriminant analysis effect size highlighted microbial features that best distinguish categorized patient samples. RESULTS Gut microbiome diversity continuously decreased over the course of CRT, with the largest decrease at week 5. Expanded Prostate Cancer Index Composite bowel function scores also declined over the course of treatment, reflecting increased symptom burden. At all individual time points, higher diversity of the gut microbiome was linearly correlated with better patient-reported GI function, but baseline diversity was not predictive of eventual outcome. Patients with high toxicity demonstrated different compositional changes during CRT in addition to compositional differences in Clostridia species. CONCLUSIONS Over time, increased radiation toxicity is associated with decreased gut microbiome diversity. Baseline diversity is not predictive of end-of-treatment bowel toxicity, but composition may identify patients at risk for developing high toxicity.
Collapse
Affiliation(s)
- Aparna Mitra
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Andrea Y Delgado
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tatiana V Karpinets
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Travis N Solley
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Melissa P Mezzari
- Alkek Center for Metagenomics and Microbiome Research Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Kyoko Yoshida-Court
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joe F Petrosino
- Alkek Center for Metagenomics and Microbiome Research Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Megan D Mikkelson
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lilie Lin
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Eifel
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jianhua Zhang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lois M Ramondetta
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anuja Jhingran
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Travis T Sims
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathleen Schmeler
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pablo Okhuysen
- Department of Infectious Diseases, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren E Colbert
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ann H Klopp
- Division of Radiation Oncology University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
69
|
Abstract
The microbiome field is increasingly raising interest among scientists, clinicians, biopharmaceutical entities, and the general public. Technological advances from the past two decades have enabled the rapid expansion of our ability to characterize the human microbiome in depth, highlighting its previously underappreciated role in contributing to multifactorial diseases including those with unknown etiology. Consequently, there is growing evidence that the microbiome could be utilized in medical diagnosis and patient stratification. Moreover, multiple gut microbes and their metabolic products may be bioactive, thereby serving as future potential microbiome-targeting or -associated therapeutics. Such therapies could include new generation probiotics, prebiotics, fecal microbiota transplantations, postbiotics, and dietary modulators. However, microbiome research has also been associated with significant limitations, technical and conceptual challenges, and, at times, "over-hyped" expectations that microbiome research will produce quick solutions to chronic and mechanistically complex human disorders. Herein, we summarize these challenges and also discuss some of the realistic promises associated with microbiome research and its applicability into clinical application.
Collapse
Affiliation(s)
- Sara Federici
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Jotham Suez
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
70
|
Septyaningtrias DE, Lin CW, Ouchida R, Nakai N, Suda W, Hattori M, Morita H, Honda K, Tamada K, Takumi T. Altered microbiota composition reflects enhanced communication in 15q11-13 CNV mice. Neurosci Res 2019; 161:59-67. [PMID: 31863791 DOI: 10.1016/j.neures.2019.12.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorder (ASD) is a complex and heterogeneous neurodevelopmental disorder. In addition to the core symptoms of ASD, many patients with ASD also show comorbid gut dysbiosis, which may lead to various gastrointestinal (GI) problems. Intriguingly, there is evidence that gut microbiota communicate with the central nervous system to modulate behavioral output through the gut-brain axis. To investigate how the microbiota composition is changed in ASD and to identify which microbes are involved in autistic behaviors, we performed a 16S rRNA gene-based metagenomics analysis in an ASD mouse model. Here, we focused on a model with human 15q11-13 duplication (15q dup), the most frequent chromosomal aberration or copy number variation found in ASD. Species diversity of the microbiome was significantly decreased in 15q dup mice. A combination of antibiotics treatment and behavioral analysis showed that neomycin improved social communication in 15q dup mice. Furthermore, comparison of the microbiota composition of mice treated with different antibiotics enabled us to identify beneficial operational taxonomic units (OTUs) for ultrasonic vocalization.
Collapse
Affiliation(s)
- Dian Eurike Septyaningtrias
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan; Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami, Hiroshima, 734-8553, Japan
| | - Chia-Wen Lin
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Rika Ouchida
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Nobuhiro Nakai
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, 230-0045, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, 230-0045, Japan
| | - Hidetoshi Morita
- Graduate School of Environmental and Life Science, Okayama University, Kita, Okayama, 700-8530, Japan
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences, Tsurumi, Yokohama, 230-0045, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku, Tokyo 160-8582, Japan
| | - Kota Tamada
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan
| | - Toru Takumi
- RIKEN Brain Science Institute, Wako, Saitama, 351-0198, Japan; Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami, Hiroshima, 734-8553, Japan; RIKEN Center for Science and Technology Hub, Medical Sciences Innovation Hub Program (MIH), Japan; Department of Physiology and Cell Biology, Kobe University Graduate School of Medicine, Chuo, Kobe, 650-0017, Japan.
| |
Collapse
|
71
|
Gil-Sánchez I, Cueva C, Tamargo A, Quintela JC, de la Fuente E, Walker AW, Moreno-Arribas MV, Bartolomé B. Application of the dynamic gastrointestinal simulator (simgi®) to assess the impact of probiotic supplementation in the metabolism of grape polyphenols. Food Res Int 2019; 129:108790. [PMID: 32036893 DOI: 10.1016/j.foodres.2019.108790] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/20/2023]
Abstract
In this paper, the Dynamic Gastrointestinal Simulator (simgi®) is used as a model to the study the metabolic activity of probiotics at the intestinal level, and in particular, to assess the impact of probiotic supplementation in the microbial metabolism of grape polyphenols. Two independent simulations using fecal samples from two healthy volunteers were carried out. Changes in microbiota composition and in metabolic activity were assessed by qPCR and 16S rRNA gene sequencing and by analyses of phenolic metabolites and ammonium ions (NH4+). The strain Lactobacillus plantarum CLC 17 was successfully implanted in the colon compartments of the simgi® after daily feeding of 2 × 1010 CFU/day for 7 days. Overall, no changes in bacterial diversity were observed after probiotic implantation. In comparison to the digestion of the grape polyphenols on their own, the inclusion of L. plantarum CLC 17 in the simgi® colon compartments led to a greater formation of phenolic metabolites such as benzoic acids, probably by the breakdown of high-molecular-weight procyanidin polymers. These results provide evidence that the probiotic strain Lactobacillus plantarum CLC 17 may improve the metabolism of dietary polyphenols when used as a food ingredient.
Collapse
Affiliation(s)
- Irene Gil-Sánchez
- Institute of Food Science Research (CIAL), CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Carolina Cueva
- Institute of Food Science Research (CIAL), CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Alba Tamargo
- Institute of Food Science Research (CIAL), CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Jose C Quintela
- Natac S A, Parque Científico de Madrid, C/ Faraday 7, 28049 Madrid, Spain
| | | | - Alan W Walker
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, Scotland, UK
| | | | - Begoña Bartolomé
- Institute of Food Science Research (CIAL), CSIC-UAM, C/ Nicolás Cabrera 9, 28049 Madrid, Spain.
| |
Collapse
|
72
|
Li J, Cha R, Zhao X, Guo H, Luo H, Wang M, Zhou F, Jiang X. Gold Nanoparticles Cure Bacterial Infection with Benefit to Intestinal Microflora. ACS NANO 2019; 13:5002-5014. [PMID: 30916928 DOI: 10.1021/acsnano.9b01002] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Antibiotics that are most used to cure bacterial infections in the clinic result in the imbalance of intestinal microflora, destroy the intestinal barrier, and induce bacterial resistance. There is an urgent need for antibacterial agent therapy for bacterial infections that does not destroy intestinal microflora. Herein, we applied 4,6-diamino-2-pyrimidinethiol (DAPT)-coated Au nanoparticles (D-Au NPs) for therapy of bacterial infection induced by Escherichia coli ( E. coli) in the gut. We cultured D-Au NPs and E. coli in an anaerobic atmosphere to evaluate their bactericidal effect. We studied the microflora, distribution of Au, and biomarkers in mice after a 28-day oral administration to analyze the effect of Au NPs on mice. D-Au NPs cured bacterial infections more effectively than levofloxacin without harming intestinal microflora. D-Au NPs showed great potential as alternatives to oral antibiotics.
Collapse
Affiliation(s)
- Juanjuan Li
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology , China University of Geosciences (Beijing) , No. 29 Xueyuan Road , Beijing 100083 , People's Republic of China
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Ruitao Cha
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Xiaohui Zhao
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Hongbo Guo
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Huize Luo
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology , China University of Geosciences (Beijing) , No. 29 Xueyuan Road , Beijing 100083 , People's Republic of China
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Mingzheng Wang
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology , China University of Geosciences (Beijing) , No. 29 Xueyuan Road , Beijing 100083 , People's Republic of China
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
| | - Fengshan Zhou
- Beijing Key Laboratory of Materials Utilization of Nonmetallic Minerals and Solid Wastes, National Laboratory of Mineral Materials, School of Materials Science and Technology , China University of Geosciences (Beijing) , No. 29 Xueyuan Road , Beijing 100083 , People's Republic of China
| | - Xingyu Jiang
- Beijing Engineering Research Center for BioNanotechnology and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for NanoScience and Technology , No. 11 Zhongguancun Beiyitiao , Beijing 100190 , People's Republic of China
- Department of Biomedical Engineering , Southern University of Science and Technology , No. 1088 Xueyuan Road , Nanshan District, Shenzhen , Guangdong 518055 , People's Republic of China
- University of Chinese Academy of Sciences , 19 A Yuquan Road , Shijingshan District, Beijing 100049 , People's Republic of China
| |
Collapse
|
73
|
Suez J, Zmora N, Zilberman-Schapira G, Mor U, Dori-Bachash M, Bashiardes S, Zur M, Regev-Lehavi D, Ben-Zeev Brik R, Federici S, Horn M, Cohen Y, Moor AE, Zeevi D, Korem T, Kotler E, Harmelin A, Itzkovitz S, Maharshak N, Shibolet O, Pevsner-Fischer M, Shapiro H, Sharon I, Halpern Z, Segal E, Elinav E. Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT. Cell 2019; 174:1406-1423.e16. [PMID: 30193113 DOI: 10.1016/j.cell.2018.08.047] [Citation(s) in RCA: 666] [Impact Index Per Article: 133.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 06/05/2018] [Accepted: 08/20/2018] [Indexed: 02/06/2023]
Abstract
Probiotics are widely prescribed for prevention of antibiotics-associated dysbiosis and related adverse effects. However, probiotic impact on post-antibiotic reconstitution of the gut mucosal host-microbiome niche remains elusive. We invasively examined the effects of multi-strain probiotics or autologous fecal microbiome transplantation (aFMT) on post-antibiotic reconstitution of the murine and human mucosal microbiome niche. Contrary to homeostasis, antibiotic perturbation enhanced probiotics colonization in the human mucosa but only mildly improved colonization in mice. Compared to spontaneous post-antibiotic recovery, probiotics induced a markedly delayed and persistently incomplete indigenous stool/mucosal microbiome reconstitution and host transcriptome recovery toward homeostatic configuration, while aFMT induced a rapid and near-complete recovery within days of administration. In vitro, Lactobacillus-secreted soluble factors contributed to probiotics-induced microbiome inhibition. Collectively, potential post-antibiotic probiotic benefits may be offset by a compromised gut mucosal recovery, highlighting a need of developing aFMT or personalized probiotic approaches achieving mucosal protection without compromising microbiome recolonization in the antibiotics-perturbed host.
Collapse
Affiliation(s)
- Jotham Suez
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Niv Zmora
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel; Internal Medicine Department, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel
| | | | - Uria Mor
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Mally Dori-Bachash
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Stavros Bashiardes
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Maya Zur
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Dana Regev-Lehavi
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Rotem Ben-Zeev Brik
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Sara Federici
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Max Horn
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yotam Cohen
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Andreas E Moor
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - David Zeevi
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Tal Korem
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Eran Kotler
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Nitsan Maharshak
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Research Center for Digestive tract and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Oren Shibolet
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Research Center for Digestive tract and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
| | | | - Hagit Shapiro
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Itai Sharon
- Migal Galilee Research Institute, 11016 Kiryat Shmona, Israel; Tel Hai College, Upper Galilee, 1220800, Israel
| | - Zamir Halpern
- Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Research Center for Digestive tract and Liver Diseases, Tel Aviv Sourasky Medical Center, 6423906 Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel.
| | - Eran Segal
- Department of Molecular Cell Biology, Weizmann Institute of Science, 7610001 Rehovot, Israel; Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
74
|
Suez J, Zmora N, Segal E, Elinav E. The pros, cons, and many unknowns of probiotics. Nat Med 2019; 25:716-729. [DOI: 10.1038/s41591-019-0439-x] [Citation(s) in RCA: 690] [Impact Index Per Article: 138.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/28/2019] [Indexed: 02/07/2023]
|
75
|
Abstract
Technological developments, including massively parallel DNA sequencing, gnotobiotics, metabolomics, RNA sequencing and culturomics, have markedly propelled the field of microbiome research in recent years. These methodologies can be harnessed to improve our in-depth mechanistic understanding of basic concepts related to consumption of probiotics, including their rules of engagement with the indigenous microbiome and impacts on the human host. We have recently demonstrated that even during probiotic supplementation, resident gut bacteria in a subset of individuals resist the mucosal presence of probiotic strains, limiting their modulatory effect on the microbiome and on the host gut transcriptional landscape. Resistance is partly alleviated by antibiotics treatment, which enables probiotics to interact with the host at the gut mucosal interface, although rather than promoting reconstitution of the indigenous microbiome and of the host transcriptional profile, they inhibit these components from returning to their naïve pre-antibiotic configurations. In this commentary, we discuss our findings in the context of previous and recent works, and suggest that incorporating the state-of-the-art methods currently utilized in microbiome research into the field of probiotics may lead to improved understanding of their mechanisms of activity, as well as their efficacy and long-term safety.
Collapse
Affiliation(s)
- Jotham Suez
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel,Digestive Center, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel,Internal Medicine Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel,Cancer-Microbiome Research Division, DKFZ, Heidelberg, Germany,CONTACT Eran Elinav Immunology Department, Weizmann Institute of Science, 234 Herzl Street, Rehovot 7610001, Israel
| |
Collapse
|
76
|
Ntemiri A, Ribière C, Stanton C, Ross RP, O'Connor EM, O'Toole PW. Retention of Microbiota Diversity by Lactose-Free Milk in a Mouse Model of Elderly Gut Microbiota. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:2098-2112. [PMID: 30665298 DOI: 10.1021/acs.jafc.8b06414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Prebiotics may improve aging-related dysbiosis. Milk is a source of nutrients including oligosaccharides whose prebiotic potential remains largely unexplored. We used a murine model to explore the effect of milk products on high diversity and lower diversity faecal microbiota from healthy and frail elderly subjects, respectively. Mice were treated with antibiotics and subsequently "humanized" with human faecal microbiota. The mice received lactose-free or whole milk, glycomacropeptide, or soy protein (control) supplemented diets for one month. The faecal microbiota was analyzed by 16S rRNA gene amplicon sequencing. Lactose-free milk diet was as efficient as the control diet in retaining faecal microbiota diversity in mice. Both milk diets had a significant effect on the relative abundance of health-relevant taxa (e.g., Ruminococcaceae, Lachnospiraceae). The glycomacropeptide prebiotic activity previously observed in vitro was not replicated in vivo. However, these data indicate the novel prebiotic potential of bovine milk for human nutrition.
Collapse
Affiliation(s)
- Alexandra Ntemiri
- School of Microbiology, Food Science Building , University College Cork , Western Road , T12 Y337 Cork , Ireland
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
| | - Céline Ribière
- School of Microbiology, Food Science Building , University College Cork , Western Road , T12 Y337 Cork , Ireland
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
| | - Catherine Stanton
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
- Teagasc, Moorepark Food Research Centre , Fermoy , Co Cork P61 C996 , Ireland
| | - R Paul Ross
- School of Microbiology, Food Science Building , University College Cork , Western Road , T12 Y337 Cork , Ireland
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
| | - Eibhlís M O'Connor
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
- Department of Biological Sciences, School of Natural Sciences , University of Limerick , V94 T9PX Limerick , Ireland
- Health Research Institute , University of Limerick , V94 T9PX Limerick , Ireland
| | - Paul W O'Toole
- School of Microbiology, Food Science Building , University College Cork , Western Road , T12 Y337 Cork , Ireland
- APC Microbiome Ireland , University College Cork , Western Road , T12 YN60 Cork , Ireland
| |
Collapse
|
77
|
Gupta S, Fečkaninová A, Lokesh J, Koščová J, Sørensen M, Fernandes J, Kiron V. Lactobacillus Dominate in the Intestine of Atlantic Salmon Fed Dietary Probiotics. Front Microbiol 2019; 9:3247. [PMID: 30700981 PMCID: PMC6343587 DOI: 10.3389/fmicb.2018.03247] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Probiotics, the live microbial strains incorporated as dietary supplements, are known to provide health benefits to the host. These live microbes manipulate the gut microbial community by suppressing the growth of certain intestinal microbes while enhancing the establishment of some others. Lactic acid bacteria (LAB) have been widely studied as probiotics; in this study we have elucidated the effects of two fish-derived LAB types (RII and RIII) on the distal intestinal microbial communities of Atlantic salmon (Salmo salar). We employed high-throughput 16S rRNA gene amplicon sequencing to investigate the bacterial communities in the distal intestinal content and mucus of Atlantic salmon fed diets coated with the LABs or that did not have microbes included in it. Our results show that the supplementation of the microbes shifts the intestinal microbial profile differentially. LAB supplementation did not cause any significant alterations in the alpha diversity of the intestinal content bacteria but RIII feeding increased the bacterial diversity in the intestinal mucus of the fish. Beta diversity analysis revealed significant differences between the bacterial compositions of the control and LAB-fed groups. Lactobacillus was the dominant genus in LAB-fed fish. A few members of the phyla Tenericutes, Proteobacteria, Actinobacteria, and Spirochaetes were also found to be abundant in the LAB-fed groups. Furthermore, the bacterial association network analysis showed that the co-occurrence pattern of bacteria of the three study groups were different. Dietary probiotics can modulate the composition and interaction of the intestinal microbiota of Atlantic salmon.
Collapse
Affiliation(s)
- Shruti Gupta
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Adriána Fečkaninová
- Department of Food Hygiene and Technology, University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Jep Lokesh
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Jana Koščová
- Department of Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Košice, Slovakia
| | - Mette Sørensen
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Jorge Fernandes
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| | - Viswanath Kiron
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
| |
Collapse
|
78
|
Feng P, Ye Z, Kakade A, Virk AK, Li X, Liu P. A Review on Gut Remediation of Selected Environmental Contaminants: Possible Roles of Probiotics and Gut Microbiota. Nutrients 2018; 11:nu11010022. [PMID: 30577661 PMCID: PMC6357009 DOI: 10.3390/nu11010022] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Various environmental contaminants including heavy metals, pesticides and antibiotics can contaminate food and water, leading to adverse effects on human health, such as inflammation, oxidative stress and intestinal disorder. Therefore, remediation of the toxicity of foodborne contaminants in human has become a primary concern. Some probiotic bacteria, mainly Lactobacilli, have received a great attention due to their ability to reduce the toxicity of several contaminants. For instance, Lactobacilli can reduce the accumulation and toxicity of selective heavy metals and pesticides in animal tissues by inhibiting intestinal absorption of contaminants and enhancing intestinal barrier function. Probiotics have also shown to decrease the risk of antibiotic-associated diarrhea possibly via competing and producing antagonistic compounds against pathogenic bacteria. Furthermore, probiotics can improve immune function by enhancing the gut microbiota mediated anti-inflammation. Thus, these probiotic bacteria are promising candidates for protecting body against foodborne contaminants-induced toxicity. Study on the mechanism of these beneficial bacterial strains during remediation processes and particularly their interaction with host gut microbiota is an active field of research. This review summarizes the current understanding of the remediation mechanisms of some probiotics and the combined effects of probiotics and gut microbiota on remediation of foodborne contaminants in vivo.
Collapse
Affiliation(s)
- Pengya Feng
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Ze Ye
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Apurva Kakade
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Amanpreet Kaur Virk
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Xiangkai Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| | - Pu Liu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Tianshuinanlu #222, Lanzhou 730000, Gansu, China.
| |
Collapse
|
79
|
Hertz FB, Nielsen KL, Frimodt-Møller N. Selection of ESBL-Producing E. coli in a Mouse Intestinal Colonization Model. Methods Mol Biol 2018; 1736:105-115. [PMID: 29322463 DOI: 10.1007/978-1-4939-7638-6_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Asymptomatic human carriage of antimicrobially drug-resistant pathogens prior to infection is increasing worldwide. Further investigation into the role of this fecal reservoir is important for combatting the increasing antimicrobial resistance problems. Additionally, the damage on the intestinal microflora due to antimicrobial treatment is still not fully understood. Animal models are powerful tools to investigate bacterial colonization subsequent to antibiotic treatment. In this chapter we present a mouse-intestinal colonization model designed to investigate how antibiotics select for an ESBL-producing E. coli isolate. The model can be used to study how antibiotics with varying effect on the intestinal flora promote the establishment of the multidrug-resistant E. coli. Colonization is successfully investigated by sampling and culturing stool during the days following administration of antibiotics. Following culturing, a precise identification of the bacterial strain found in mice feces is applied to ensure that the isolate found is in fact identical to the strain used for inoculation. For this purpose random amplified of polymorphic DNA (RAPD) PCR specifically developed for E. coli is applied. This method allows us to distinguish E. coli with more than 99.95% genome similarity using a duplex PCR method.
Collapse
Affiliation(s)
| | - Karen Leth Nielsen
- Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
80
|
Neuman H, Forsythe P, Uzan A, Avni O, Koren O. Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev 2018; 42:489-499. [PMID: 29945240 DOI: 10.1093/femsre/fuy018] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 06/23/2018] [Indexed: 12/21/2022] Open
Abstract
Antibiotics are the most common type of medication prescribed to children, including infants, in the Western world. While use of antibiotics has transformed previously lethal infections into relatively minor diseases, antibiotic treatments can have adverse effects as well. It has been shown in children, adults and animal models that antibiotics dramatically alter the gut microbial composition. Since the gut microbiota plays crucial roles in immunity, metabolism and endocrinology, the effects of antibiotics on the microbiota may lead to further health complications. In this review, we present an overview of the effects of antibiotics on the microbiome in children, and correlate them to long-lasting complications of obesity, behavior, allergies, autoimmunity and other diseases.
Collapse
Affiliation(s)
- Hadar Neuman
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel.,Ziv Medical Center, Derech HaRambam St., Safed 13100 Israel.,Zefat Academic College, Jerusalem St. 11, Safed 13206, Israel
| | - Paul Forsythe
- McMaster Brain-Body Institute, St. Joseph's Healthcare Hamilton, L8N 4A6 Hamilton, Ontario, Canada.,Firestone Institute for Respiratory Health and Department of Medicine, 50 Charlton Avenue East, McMaster University, L8N 4A6 Hamilton, Ontario, Canada
| | - Atara Uzan
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Orly Avni
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| | - Omry Koren
- Faculty of Medicine, Bar Ilan University, Henrietta Szold 8, Safed 13115, Israel
| |
Collapse
|
81
|
Bao HD, Pang MD, Olaniran A, Zhang XH, Zhang H, Zhou Y, Sun LC, Schmidt S, Wang R. Alterations in the diversity and composition of mice gut microbiota by lytic or temperate gut phage treatment. Appl Microbiol Biotechnol 2018; 102:10219-10230. [PMID: 30302521 DOI: 10.1007/s00253-018-9378-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/29/2018] [Accepted: 09/05/2018] [Indexed: 12/12/2022]
Abstract
Phages, the most abundant species in the mammalian gut, have numerous advantages as biocontrol agent over antibiotics. In this study, mice were orally treated with the lytic gut phage PA13076 (group B), the temperate phage BP96115 (group C), no phage (group A), or streptomycin (group D) over 31 days. At the end of the experiment, fecal microbiota diversity and composition was determined and compared using high-throughput sequencing of the V3-V4 hyper-variable region of the 16S rRNA gene and virus-like particles (VLPs) were quantified in feces. There was high diversity and richness of microbiota in the lytic and temperate gut phage-treated mice, with the lytic gut phage causing an increased alpha diversity based on the Chao1 index (p < 0.01). However, the streptomycin treatment reduced the microbiota diversity and richness (p = 0.0299). Both phage and streptomycin treatments reduced the abundance of Bacteroidetes at the phylum level (p < 0.01) and increased the abundance of the phylum Firmicutes. Interestingly, two beneficial genera, Lactobacillus and Bifidobacterium, were enhanced by treatment with the lytic and temperate gut phage. The abundance of the genus Escherichia/Shigella was higher in mice after temperate phage administration than in the control group (p < 0.01), but lower than in the streptomycin group. Moreover, streptomycin treatment increased the abundance of the genera Klebsiella and Escherichia/Shigella (p < 0.01). In terms of the gut virome, fecal VLPs did not change significantly after phage treatment. This study showed that lytic and temperate gut phage treatment modulated the composition and diversity of gut microbiota and the lytic gut phage promoted a beneficial gut ecosystem, while the temperate phage may promote conditions enabling diseases to occur.
Collapse
Affiliation(s)
- Hong-Duo Bao
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, China
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg Campus, Private Bag X01, Pietermaritzburg, 3201, South Africa
| | - Mao-da Pang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Ademola Olaniran
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - Xu-Hui Zhang
- College of Forestry, Nanjing Forestry University, Nanjing, 210037, China
| | - Hui Zhang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Yan Zhou
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Li-Chang Sun
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China
| | - Stefan Schmidt
- Discipline of Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu-Natal, Pietermaritzburg Campus, Private Bag X01, Pietermaritzburg, 3201, South Africa.
| | - Ran Wang
- Institute of Food Safety and Nutrition, Key Lab of Food Quality and Safety of Jiangsu Province-State Key Laboratory Breeding Base, Jiangsu Academy of Agricultural Sciences, No. 50 Zhongling Street, Xuanwu Area, Nanjing, 210014, China.
| |
Collapse
|
82
|
Shibayama J, Kuda T, Shikano A, Fukunaga M, Takahashi H, Kimura B, Ishizaki S. Effects of rice bran and fermented rice bran suspensions on caecal microbiota in dextran sodium sulphate-induced inflammatory bowel disease model mice. FOOD BIOSCI 2018. [DOI: 10.1016/j.fbio.2018.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
83
|
Yuan L, Zhang S, Li H, Yang F, Mushtaq N, Ullah S, Shi Y, An C, Xu J. The influence of gut microbiota dysbiosis to the efficacy of 5-Fluorouracil treatment on colorectal cancer. Biomed Pharmacother 2018; 108:184-193. [PMID: 30219675 DOI: 10.1016/j.biopha.2018.08.165] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/28/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer is one of the most frequently diagnosed cancers worldwide. Gut flora can modulate the host response to chemotherapeutic drugs. However, the understanding regarding the relationship between the gut microbiota and the antitumor efficacy of 5- Fluorouracil (5-FU) treatment is limited. Therefore, we compared the tumor size and profiled the gut microbiota of mice treated with 5-FU, combined with probiotics or ABX (an antibiotic cocktail of antibiotics) by using the Colorectal Cancer (CRC) mouse model and high-throughput sequencing. The results elucidated that ABX administration diminished the antitumor efficacy of 5-FU in mice and supplementation of probiotics upon 5-FU treatment could not significantly increase the efficacy of 5-FU treatment, despite improving mice body weight at day 33. There were significant differences in fecal bacteria community among the four groups (ANOSIM p < 0.05). ABX administration reduced microbiota biodiversity and altered microbiota community. The pathogenic bacteria included Escherichia shigella and Enterobacter significantly increased, while other commensal bacterial decreased unidentified Firmicutes increased and the opportunistic pathogens decreased after the administration of Probiotics. In addition, 5-FU treatment also changed the diversity and the community composition of the gut mirobiota. The relative abundance of genus Lachnospiracea_NK4 A136, Bacteroides, Odoribacter, Mucispirillum, and Blautia were significantly increased compared to the control group. Additionally, functional capacity analysis of gut microbiota using PICRUSt showed that genes involved in amino acid metabolism, replication and repair translation, nucleotide metabolism expressed much lower in FU.ABX group than the other groups. The current results suggest that ABX administration disrupted the gut microbiota in mice, which contributed to the reduction of antitumor efficacy of 5-FU.
Collapse
Affiliation(s)
- Lu Yuan
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Siruo Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Huan Li
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Fan Yang
- Department of Neurosurgery, Navy General Hospital of PLA, Beijing, China
| | - Noosheen Mushtaq
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Shakir Ullah
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yi Shi
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China
| | - Cuihong An
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an, China
| | - Jiru Xu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
84
|
Touret T, Oliveira M, Semedo-Lemsaddek T. Putative probiotic lactic acid bacteria isolated from sauerkraut fermentations. PLoS One 2018; 13:e0203501. [PMID: 30192827 PMCID: PMC6128568 DOI: 10.1371/journal.pone.0203501] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022] Open
Abstract
Probiotics are live microorganisms which confer health benefits to the host, and may be isolated from several sources, such as vegetable foodstuffs. Sauerkraut is a cabbage product resulting from fermentation by a lactic acid bacteria microbial succession, and is a potential source for probiotics. The aim of the present study was the isolation and characterization of probiotic microorganisms from sauerkraut fermentations. Four distinct fermentations were performed, from which lactic acid bacteria were recovered. Overall, 114 isolates were obtained, phenotypically and genotypically characterized, identified to the genus level and evaluated regarding safety and probiotic potential. Representative bacteria were selected for further analysis, 52% being Lactobacillus spp. and 33% belonging to Leuconostoc spp. genus. One isolate revealed to be β-hemolytic, 42% possessed potentially mobile antimicrobial resistance, 88% were resistant to bile and 20% to low pH. The six most promising candidates were further characterized and presented antimicrobial activity against Listeria monocytogenes, three being resistant to lower pH values. Thus, global analysis of data gathered during this study highlighted the identification of three Lactobacillus strains with putative probiotic potential, suggesting the applicability of sauerkraut fermentations as a source for probiotic isolation. Due to their origin these strains should be suited for future application in the food industry, namely vegetable products such as sauerkraut itself.
Collapse
Affiliation(s)
- Tiago Touret
- CIISA – Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| | - Manuela Oliveira
- CIISA – Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- * E-mail:
| | - Teresa Semedo-Lemsaddek
- CIISA – Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
85
|
Yasushi Y, Haraguchi Y, Shikano A, Kuda T, Takahashi H, Kimura B. Induction of gut Lactobacillus reuteri
in normal ICR mice by oral administration of L. plantarum
AN1. J Food Biochem 2018. [DOI: 10.1111/jfbc.12589] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Yokota Yasushi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Yutaka Haraguchi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Ayane Shikano
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Takashi Kuda
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Hajime Takahashi
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| | - Bon Kimura
- Department of Food Science and Technology; Tokyo University of Marine Science and Technology; Tokyo Japan
| |
Collapse
|
86
|
Supportive Role of Probiotic Strains in Protecting Rats from Ovariectomy-Induced Cortical Bone Loss. Probiotics Antimicrob Proteins 2018; 11:1145-1154. [DOI: 10.1007/s12602-018-9443-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
87
|
Kuda T, Yokota Y, Haraguchi Y, Takahashi H, Kimura B. Susceptibility of gut indigenous lactic acid bacteria in BALB/c mice to oral administered Lactobacillus plantarum. Int J Food Sci Nutr 2018; 70:53-62. [DOI: 10.1080/09637486.2018.1471590] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Takashi Kuda
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Yasushi Yokota
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Yutaka Haraguchi
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Hajime Takahashi
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Bon Kimura
- Department of Food Science and Technology, Tokyo University of Marine Science and Technology, Tokyo, Japan
| |
Collapse
|
88
|
Wei Y, Yang F, Wu Q, Gao J, Liu W, Liu C, Guo X, Suwal S, Kou Y, Zhang B, Wang Y, Zheng K, Tang R. Protective Effects of Bifidobacterial Strains Against Toxigenic Clostridium difficile. Front Microbiol 2018; 9:888. [PMID: 29867801 PMCID: PMC5952185 DOI: 10.3389/fmicb.2018.00888] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
Probiotics might offer an attractive alternative to prevent and control Clostridium difficile (C. difficile) infection (CDI). Limited information is available on the ability of commercially used bifidobacterial strains to inhibit C. difficile. This study examined the anti-clostridial effects of Bifidobacterium longum JDM301, a widely used commercial probiotic strain in China, in vitro and in vivo. In vitro evaluation revealed a significant reduction in C. difficile counts when JDM301 was co-cultured with C. difficile, which was correlated with the significant decrease in clostridial toxin titres (TcdA and TcdB). Furthermore, the cell-free culture supernatants (CFS) of JDM301 inhibited C. difficile growth and degraded TcdA and TcdB. Notably, the results showed that acid pH promoted the degradation of TcdA by CFS from JDM301. Furthermore, comparative studies among 10 B. longum strains were performed, which showed that the inhibitory effect of CFS from JDM301 was similar with the other 8 B. longum strains and higher than strain BLY1. However, when it was neutralized, the significant different was lost. When present together, it was suggested that the acid pH induced by probiotics not only played important roles in the growth inhibition against C. difficile resulting in the reduction of toxins titres, but also directly promoted the degradation of clostridial toxin. In vivo studies proved that JDM301 partially relieved damage to tissues caused by C. difficile and also decreased the number of C. difficile and toxin levels. In summary, our results demonstrated that the commercial strain, JDM301 could be considered a probiotic able to exert anti-toxin capability and most of the CFS from Bifidobacterium were able to inhibit the growth of C. difficile, depending on acid pH. These results highlighted a potential that JDM301 could be helpful in preventing CDI and that most of the bifidobacterial strains could (at least partially) exert protective effects by reducing toxin titres through growth inhibition against toxigenic C. difficile.
Collapse
Affiliation(s)
- Yanxia Wei
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Fan Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Qiong Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Jing Gao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Wenli Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Chang Liu
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaokui Guo
- Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sharmila Suwal
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Yanbo Kou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Bo Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Yugang Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
89
|
Esaiassen E, Hjerde E, Cavanagh JP, Pedersen T, Andresen JH, Rettedal SI, Støen R, Nakstad B, Willassen NP, Klingenberg C. Effects of Probiotic Supplementation on the Gut Microbiota and Antibiotic Resistome Development in Preterm Infants. Front Pediatr 2018; 6:347. [PMID: 30505830 PMCID: PMC6250747 DOI: 10.3389/fped.2018.00347] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022] Open
Abstract
Objectives: In 2014 probiotic supplementation (Lactobacillus acidophilus and Bifidobacterium longum subspecies infantis; InfloranⓇ) was introduced as standard of care to prevent necrotizing enterocolitis (NEC) in extremely preterm infants in Norway. We aimed to evaluate the influence of probiotics and antibiotic therapy on the developing gut microbiota and antibiotic resistome in extremely preterm infants, and to compare with very preterm infants and term infants not given probiotics. Study design: A prospective, observational multicenter study in six tertiary-care neonatal units. We enrolled 76 infants; 31 probiotic-supplemented extremely preterm infants <28 weeks gestation, 35 very preterm infants 28-31 weeks gestation not given probiotics and 10 healthy full-term control infants. Taxonomic composition and collection of antibiotic resistance genes (resistome) in fecal samples, collected at 7 and 28 days and 4 months age, were analyzed using shotgun-metagenome sequencing. Results: Median (IQR) birth weight was 835 (680-945) g and 1,290 (1,150-1,445) g in preterm infants exposed and not exposed to probiotics, respectively. Two extremely preterm infants receiving probiotic developed NEC requiring surgery. At 7 days of age we found higher median relative abundance of Bifidobacterium in probiotic supplemented infants (64.7%) compared to non-supplemented preterm infants (0.0%) and term control infants (43.9%). Lactobacillus was only detected in small amounts in all groups, but the relative abundance increased up to 4 months. Extremely preterm infants receiving probiotics had also much higher antibiotic exposure, still overall microbial diversity and resistome was not different than in more mature infants at 4 weeks and 4 months. Conclusion: Probiotic supplementation may induce colonization resistance and alleviate harmful effects of antibiotics on the gut microbiota and antibiotic resistome. Clinical Trial Registration: Clinicaltrials.gov: NCT02197468. https://clinicaltrials.gov/ct2/show/NCT02197468.
Collapse
Affiliation(s)
- Eirin Esaiassen
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Erik Hjerde
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Jorunn Pauline Cavanagh
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| | - Tanja Pedersen
- Department of Paediatrics, Haukeland University Hospital, Bergen, Norway
| | - Jannicke H Andresen
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Siren I Rettedal
- Department of Paediatrics, Stavanger University Hospital, Stavanger, Norway
| | - Ragnhild Støen
- Department of Paediatrics, St. Olavs University Hospital, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, University of Science and Technology, Trondheim, Norway
| | - Britt Nakstad
- Department of Paediatric and Adolescents Medicine, Akershus University Hospital, Nordbyhagen, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nils P Willassen
- Department of Chemistry, Norstruct, UiT The Arctic University of Norway, Tromsø, Norway
| | - Claus Klingenberg
- Paediatric Research Group, Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Paediatrics, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
90
|
Dong Q, Xin Y, Wang L, Meng X, Yu X, Lu L, Xuan S. Characterization of Gastric Microbiota in Twins. Curr Microbiol 2016; 74:224-229. [PMID: 27957630 DOI: 10.1007/s00284-016-1176-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022]
Abstract
Contribution of host genetic backgrounds in the development of gastric microbiota has not been clearly defined. This study was aimed to characterize the biodiversity, structure and composition of gastric microbiota among twins. A total of four pairs of twins and eight unrelated individuals were enrolled in the study. Antral biopsies were obtained during endoscopy. The bacterial 16S rRNA gene was amplified and pyrosequenced. Sequences were analyzed for the composition, structure, and α and β diversities of gastric microbiota. Proteobacteria, Firmicutes, Bacteroidetes, Actinobacteria, and Fusobacteria were the most predominant phyla of gastric microbiota. Each individual, twins as well as unrelated individuals, harbored a microbiota of distinct composition. There was no evidence of additional similarity in the richness and evenness of gastric microbiota among co-twins as compared to unrelated individuals. Calculations of θYC and PCoA demonstrated that the structure similarity of gastric microbial community between co-twins did not increase compared to unrelated individuals. In contrast, the structure of microbiota was altered enormously by Helicobacter pylori infection. These results suggest that host genetic backgrounds had little effect in shaping the gastric microbiota. This property of gastric microbiota could facilitate the studies discerning the role of microbiota from genetic grounds in the pathogenesis.
Collapse
Affiliation(s)
- Quanjiang Dong
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Yongning Xin
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Lili Wang
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Xinying Meng
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Xinjuan Yu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Linlin Lu
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China
| | - Shiying Xuan
- Central Laboratories and Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266000, Shandong, China.
| |
Collapse
|
91
|
Characterization of Lactobacillus reuteri BCLR-42 and Lactobacillus plantarum BCLP-51 as novel dog probiotics with innate immune enhancing properties. ACTA ACUST UNITED AC 2016. [DOI: 10.14405/kjvr.2016.56.2.75] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|