101
|
Aslam H, Green J, Jacka FN, Collier F, Berk M, Pasco J, Dawson SL. Fermented foods, the gut and mental health: a mechanistic overview with implications for depression and anxiety. Nutr Neurosci 2018; 23:659-671. [DOI: 10.1080/1028415x.2018.1544332] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hajara Aslam
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Food & Mood Centre, IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Jessica Green
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Food & Mood Centre, IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Austin Health, Heidelberg, Australia
| | - Felice N. Jacka
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Food & Mood Centre, IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Centre for Adolescent Health, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia
| | - Fiona Collier
- Geelong Centre for Emerging Infectious Disease, Barwon Health, Geelong, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- The Department of Psychiatry and the Florey Institute of Neuroscience and Mental Health Department, The Centre of Excellence in Youth Mental Health, The University of Melbourne, Orygen, Australia
| | - Julie Pasco
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Samantha L. Dawson
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Food & Mood Centre, IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Environmental & Genetic Epidemiology Research, Murdoch Children’s Research Institute, Royal Children’s Hospital, Parkville, Australia
| |
Collapse
|
102
|
Guo J, Dougkas A, Elwood PC, Givens DI. Dairy Foods and Body Mass Index over 10-Year: Evidence from the Caerphilly Prospective Cohort Study. Nutrients 2018; 10:E1515. [PMID: 30332779 PMCID: PMC6213600 DOI: 10.3390/nu10101515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/21/2018] [Accepted: 10/12/2018] [Indexed: 11/17/2022] Open
Abstract
The association between dairy product consumption and body mass index (BMI) remains controversial. The aim of the present study was to investigate the association between total dairy, milk, cheese, cream and butter consumption and BMI change over a 10-year follow-up by using long-term follow-up cohort data from the Caerphilly Prospective Cohort Study (CAPS). The CAPS included 2512 men aged 45⁻59 years at baseline, who were followed up at 5-year intervals for over 20-year. A semi-quantitative food frequency questionnaire estimated the intake of dairy consumption, including milk, cheese, cream and butter at baseline, 5-year and 10-year follow-up. In total, men free of cardiovascular disease, diabetes and cancer (n = 1690) were included in current analysis. General linear regression and logistic regression were used for data analysis. The results showed higher cheese consumption was associated with lower BMI at the 5-year follow-up (p = 0.013). There was no evidence that higher consumption of total dairy, milk, cream and butter were significantly associated with BMI during the over the 10-year following-up. This study suggest that cheese consumption have beneficial effects on lowering BMI, which needs further investigation.
Collapse
Affiliation(s)
- Jing Guo
- Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6AR, UK.
| | - Anestis Dougkas
- Institut Paul Bocuse, Chateau Du Vivier, BP 25-69131 Ecully CEDEX, France.
| | - Peter C Elwood
- Department of Primary Care and Public Health, Cardiff University, Cardiff CF10 3AT, UK.
| | - David I Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading RG6 6AR, UK.
| |
Collapse
|
103
|
Lanne ABM, Goode A, Prattley C, Kumari D, Drasbek MR, Williams P, Conde-Álvarez R, Moriyón I, Bonev BB. Molecular recognition of lipopolysaccharide by the lantibiotic nisin. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1861:83-92. [PMID: 30296414 DOI: 10.1016/j.bbamem.2018.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/02/2018] [Accepted: 10/03/2018] [Indexed: 01/01/2023]
Abstract
Nisin is a lanthionine antimicrobial effective against diverse Gram-positive bacteria and is used as a food preservative worldwide. Its action is mediated by pyrophosphate recognition of the bacterial cell wall receptors lipid II and undecaprenyl pyrophosphate. Nisin/receptor complexes disrupt cytoplasmic membranes, inhibit cell wall synthesis and dysregulate bacterial cell division. Gram-negative bacteria are much more tolerant to antimicrobials including nisin. In contrast to Gram-positives, Gram-negative bacteria possess an outer membrane, the major constituent of which is lipopolysaccharide (LPS). This contains surface exposed phosphate and pyrophosphate groups and hence can be targeted by nisin. Here we describe the impact of LPS on membrane stability in response to nisin and the molecular interactions occurring between nisin and membrane-embedded LPS from different Gram-negative bacteria. Dye release from liposomes shows enhanced susceptibility to nisin in the presence of LPS, particularly rough LPS chemotypes that lack an O-antigen whereas LPS from microorganisms sharing similar ecological niches with antimicrobial producers provides only modest enhancement. Increased susceptibility was observed with LPS from pathogenic Klebsiella pneumoniae compared to LPS from enteropathogenic Salmonella enterica and gut commensal Escherichia coli. LPS from Brucella melitensis, an intra-cellular pathogen which is adapted to invade professional and non-professional phagocytes, appears to be refractory to nisin. Molecular complex formation between nisin and LPS was studied by solid state MAS NMR and revealed complex formation between nisin and LPS from most organisms investigated except B. melitensis. LPS/nisin complex formation was confirmed in outer membrane extracts from E. coli.
Collapse
Affiliation(s)
- Alice B M Lanne
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Alice Goode
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Charlotte Prattley
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Divya Kumari
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK
| | - Mette Ryun Drasbek
- DuPont Nutrition Biosciences ApS, Edwin Rahrs Vej 38, DK-8220 Brabrand, Denmark
| | - Paul Williams
- School of Life Sciences, CBS, University of Nottingham, Nottingham NG7 2RD, UK
| | - Raquel Conde-Álvarez
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008 Pamplona, Spain
| | - Ignacio Moriyón
- Instituto de Salud Tropical, Instituto de Investigación Sanitaria de Navarra, and Departamento de Microbiología y Parasitología, Universidad de Navarra, c/Irunlarrea 1, 31008 Pamplona, Spain
| | - Boyan B Bonev
- School of Life Sciences, QMC, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
104
|
Mena-Sánchez G, Babio N, Martínez-González MÁ, Corella D, Schröder H, Vioque J, Romaguera D, Martínez JA, Lopez-Miranda J, Estruch R, Wärnberg J, Bueno-Cavanillas A, Serra-Majem L, Tur JA, Arós F, Tinahones FJ, Sánchez VM, Lapetra J, Pintó X, Vidal J, Vázquez C, Ordovás JM, Delgado-Rodriguez M, Matía-Martín P, Basora J, Buil-Cosiales P, Fernandez-Carrion R, Fitó M, Salas-Salvadó J. Fermented dairy products, diet quality, and cardio-metabolic profile of a Mediterranean cohort at high cardiovascular risk. Nutr Metab Cardiovasc Dis 2018; 28:1002-1011. [PMID: 30207268 DOI: 10.1016/j.numecd.2018.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 05/10/2018] [Accepted: 05/11/2018] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Fermented dairy products have been associated with a better diet quality and cardio-metabolic profile. However, in Mediterranean populations, these associations have not been well characterized. The aim of this study was to assess the diet quality and the associations between the consumption of total fermented dairy products and their subtypes and the prevalence of Metabolic Syndrome (MetS) components in a Mediterranean population at high cardiovascular risk. METHODS AND RESULTS Baseline cross-sectional analyses were conducted on 6,572 men and women (mean age: 65 years) with overweight or obesity and MetS recruited into the PREDIMED-Plus cohort. A 143-item Food Frequency Questionnaire (FFQ) was used, and anthropometrical, biochemical, and blood pressure measurements were recorded. Multivariate-adjusted Cox regressions were fitted to analyze the association between quartiles of consumption of fermented dairy products and their subtypes and MetS components to estimate the relative risk (RR) and 95% confidence intervals (95% CIs). Participants who were high consumers of fermented dairy products reported a higher consumption of fruit, vegetables, fish, nuts, and whole bread and a lower consumption of white bread, alcohol, and cookies. Participants in the higher quartile showed a lower prevalence of the low HDL-cholesterol component of the MetS (RR=0.88; 95% CI: 0.78-0.98) than those in the lowest quartile of cheese consumption. Cheese consumption was inversely associated with the prevalence of hypertriglyceridemia. Total fermented dairy products, yogurt, and its types were not associated with any of the MetS components. CONCLUSIONS Compared to nonconsumers, participants consuming fermented dairy products reported a better diet quality and, particularly, cheese consumers presented a lower prevalence of hypertriglyceridemia and low HDL-cholesterol plasma levels, which are MetS components.
Collapse
Affiliation(s)
- G Mena-Sánchez
- Human Nutrition Unit, University Hospital of Sant Joan de Reus, Department of Biochemistry and Biotechnology, Pere Virgili Institute for Health Research, Rovira i Virgili University, Reus, Spain; CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - N Babio
- Human Nutrition Unit, University Hospital of Sant Joan de Reus, Department of Biochemistry and Biotechnology, Pere Virgili Institute for Health Research, Rovira i Virgili University, Reus, Spain; CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| | - M Á Martínez-González
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine and Public Health, University of Navarra-Navarra Institute for Health Research, Pamplona, Spain; Harvard TH Chan School of Public Health, Department of Nutrition, Boston, USA
| | - D Corella
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Preventive Medicine, University of Valencia, Valencia, Spain
| | - H Schröder
- Cardiovascular Risk and Nutrition Research Group (CARIN), Hospital del Mar Research Institute (IMIM), Barcelona, Spain; CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - J Vioque
- CIBER de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain; Nutritional Epidemiology Research Group, University of Miguel Hernández, Alicante, Spain
| | - D Romaguera
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Instituto de Investigación Sanitaria Illes Balears (IdISBa), University Hospital Son Espases, Palma de Mallorca, Spain
| | - J A Martínez
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Nutrition, Food Sciences, and Physiology, Center for Nutrition Research, University of Navarra, Pamplona, Spain; Madrid Institute for Advanced Studies (IMDEA) Food Institute, Madrid, Spain
| | - J Lopez-Miranda
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Lipids and Atherosclerosis Unit, Department of Internal Medicine, Reina Sofia University Hospital, IMIBIC, University of Córdoba, Córdoba, Spain
| | - R Estruch
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Internal Medicine, Hospital Clínic, IDIBAPS August Pi i Sunyer Biomedical Research Institute, University of Barcelona, Barcelona, Spain
| | - J Wärnberg
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Nursing, University of Malaga, Institute of Biomedical Research in Malaga (IBIMA), Malaga, Spain
| | - A Bueno-Cavanillas
- Nutritional Epidemiology Research Group, University of Miguel Hernández, Alicante, Spain; Departament of Preventive Medicine and Public Health, University of Granada, Spain
| | - L Serra-Majem
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Research Institute of Biomedical and Health Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - J A Tur
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Research Group on Community Nutrition and Oxidative Stress, University of the Balearic Islands, Palma de Mallorca, Spain
| | - F Arós
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Cardiology, University Hospital Araba, Vitoria, Spain
| | - F J Tinahones
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Malaga University, Malaga, Spain
| | - V M Sánchez
- Nutritional Epidemiology Research Group, University of Miguel Hernández, Alicante, Spain; Institute of Biomedicine (IBIOMED), University of León, Spain
| | - J Lapetra
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Family Medicine, Unit Research, Distrito Sanitario Atención Primaria Sevilla, Sevilla, Spain
| | - X Pintó
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Lipid Unit, Department of Internal Medicine, Bellvitge Biomedical Research Institute (IDIBELL)-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Barcelona, Spain
| | - J Vidal
- Department of Lipids, Hospital Clínic, Institut d'Investigacions Biomediques August Pi Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - C Vázquez
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Department of Endocrinology and Nutrition, University Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - J M Ordovás
- Madrid Institute for Advanced Studies (IMDEA) Food Institute, Madrid, Spain; Jean Mayer USDA Human Nutrition Research Center on Aging (JM-USDA-HNRCA), Tufts University, Boston, USA
| | - M Delgado-Rodriguez
- Instituto de Investigación Sanitaria Illes Balears (IdISBa), University Hospital Son Espases, Palma de Mallorca, Spain; Department of Health Sciences, University of Jaen, Jaen, Spain
| | - P Matía-Martín
- Endocrinology and Nutrition Department, Hospital Clínico San Carlos-IdISSC, Madrid, Spain
| | - J Basora
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Primary Health Care Area, Reus, Tarragona, Spain; Institut Català de la Salut, Generalitat de Catalunya, Barcelona, Spain
| | - P Buil-Cosiales
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Primary Health Care, Servicio Navarro de Salud-Osasunbidea, Pamplona, Navarra, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Navarra, Spain
| | - R Fernandez-Carrion
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Harvard TH Chan School of Public Health, Department of Nutrition, Boston, USA
| | - M Fitó
- CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; Cardiovascular Risk and Nutrition Research Group (CARIN), Hospital del Mar Research Institute (IMIM), Barcelona, Spain
| | - J Salas-Salvadó
- Human Nutrition Unit, University Hospital of Sant Joan de Reus, Department of Biochemistry and Biotechnology, Pere Virgili Institute for Health Research, Rovira i Virgili University, Reus, Spain; CIBER de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
105
|
Zhang H, You C. A universal PCR method and its application in sequence-based identification of microorganisms in dairy. Int Dairy J 2018. [DOI: 10.1016/j.idairyj.2018.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
106
|
Progress of analytical tools and techniques for human gut microbiome research. J Microbiol 2018; 56:693-705. [DOI: 10.1007/s12275-018-8238-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/07/2018] [Accepted: 06/08/2018] [Indexed: 12/15/2022]
|
107
|
Skelly E, Kapellas K, Cooper A, Weyrich LS. Consequences of colonialism: A microbial perspective to contemporary Indigenous health. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 167:423-437. [DOI: 10.1002/ajpa.23637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Emily Skelly
- Australian Centre for Ancient DNA, School of Biological Sciences University of Adelaide Adelaide South Australia Australia
| | - Kostas Kapellas
- Australian Research Centre for Population Oral Health, Adelaide Dental School University of Adelaide Adelaide South Australia Australia
| | - Alan Cooper
- Australian Centre for Ancient DNA, School of Biological Sciences University of Adelaide Adelaide South Australia Australia
| | - Laura S. Weyrich
- Australian Centre for Ancient DNA, School of Biological Sciences University of Adelaide Adelaide South Australia Australia
| |
Collapse
|
108
|
Dalziel J, Smolenski G, McKenzie C, Haines S, Day L. Differential effects of sheep and cow skim milk before and after fermentation on gastrointestinal transit of solids in a rat model. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
109
|
A Clinician’s Guide for Trending Cardiovascular Nutrition Controversies. J Am Coll Cardiol 2018; 72:553-568. [DOI: 10.1016/j.jacc.2018.05.030] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/19/2018] [Accepted: 05/20/2018] [Indexed: 12/19/2022]
|
110
|
Velikova P, Petrov K, Lozanov V, Tsvetanova F, Stoyanov A, Wu Z, Liu Z, Petrova P. Microbial diversity and health-promoting properties of the traditional Bulgarian yogurt. BIOTECHNOL BIOTEC EQ 2018. [DOI: 10.1080/13102818.2018.1475255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Petya Velikova
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Kaloyan Petrov
- Department of Chemical and Biochemical Reactors, Institute of Chemical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Valentin Lozanov
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Flora Tsvetanova
- Department of Chemical and Biochemical Reactors, Institute of Chemical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anton Stoyanov
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Zhengjun Wu
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, PR China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, PR China
| | - Penka Petrova
- Department of General Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
111
|
Evaluation of some in vitro probiotic properties of Lactobacillus fermentum Strains. Journal of Food Science and Technology 2018; 55:2801-2807. [PMID: 30042597 DOI: 10.1007/s13197-018-3197-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 12/17/2017] [Accepted: 05/09/2018] [Indexed: 10/16/2022]
Abstract
This study aimed to check the in vitro probiotic properties of eleven Lactobacillus fermentum strains previously isolated from fermented dairy products and infant faeces. These cultures were tested for their tolerance to different pH such as 2.0, 2.5, 3.0, 3.5 and 6.5, bile salt hydrolysis and cell surface hydrophobicity. All the strains were persistent at pH 3.5 for 3 h whereas only faecal origin isolates such as L. fermentum BIF-19, BIF-20, BIF-18 and MTCC 8711 had shown considerable growth at pH 2.5. The strains NCDC-400, MTCC 8711, BIF-18, BIF-19 and BIF-20 showed slight to intense precipitation zone of bile salt hydrolase activity by agar plate assay. The strain L. fermentum BIF-19 exhibited best preliminary probiotic properties was selected for the adhesion to Caco-2 cell lines, which shows similar adhesion to that observed for standard probiotic Lactobacillus rhamnosus GG.
Collapse
|
112
|
Skrypnik K, Suliburska J. Association between the gut microbiota and mineral metabolism. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:2449-2460. [PMID: 28991359 DOI: 10.1002/jsfa.8724] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/23/2017] [Accepted: 09/27/2017] [Indexed: 06/07/2023]
Abstract
The aim of this review is to present the most recent scientific evidence of interactions between the intestinal microbiota and minerals, and the effect of this interaction on the health of the host. The Web of Science database from the years 2013-2017 on this topic was reviewed. Numerous in vitro studies have shown that iron significantly affects the intestinal microbiota. However, Bifidobacteriaceae are capable of binding iron in the large intestine, thereby limiting the formation of free radicals synthesized in the presence of iron, and thus reducing the risk of colorectal cancer. Animal studies have revealed that supplementation with probiotics, prebiotics and synbiotics has a significant effect on bone calcium, phosphate and bone metabolism. The dynamic interaction between microbiota and zinc was shown. Human studies have provided evidence of the influence of probiotic bacteria on parathormone, calcium and phosphate levels and thus on bone resorption. Recent studies have produced new information mainly on the impact of the intestinal bacteria on the metabolism of calcium and iron. From a scientific perspective, the most urgent fields that remain to be investigated are the identification of all human gut microbes and new therapies targeting the interaction between intestinal bacteria and minerals. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Katarzyna Skrypnik
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| | - Joanna Suliburska
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, Poznań, Poland
| |
Collapse
|
113
|
Radziwill-Bienkowska JM, Talbot P, Kamphuis JBJ, Robert V, Cartier C, Fourquaux I, Lentzen E, Audinot JN, Jamme F, Réfrégiers M, Bardowski JK, Langella P, Kowalczyk M, Houdeau E, Thomas M, Mercier-Bonin M. Toxicity of Food-Grade TiO 2 to Commensal Intestinal and Transient Food-Borne Bacteria: New Insights Using Nano-SIMS and Synchrotron UV Fluorescence Imaging. Front Microbiol 2018; 9:794. [PMID: 29740421 PMCID: PMC5928251 DOI: 10.3389/fmicb.2018.00794] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 04/09/2018] [Indexed: 11/13/2022] Open
Abstract
Titanium dioxide (TiO2) is commonly used as a food additive (E171 in the EU) for its whitening and opacifying properties. However, a risk of intestinal barrier disruption, including dysbiosis of the gut microbiota, is increasingly suspected because of the presence of a nano-sized fraction in this additive. We hypothesized that food-grade E171 and Aeroxyde P25 (identical to the NM-105 OECD reference nanomaterial in the European Union Joint Research Centre) interact with both commensal intestinal bacteria and transient food-borne bacteria under non-UV-irradiated conditions. Based on differences in their physicochemical properties, we expect a difference in their respective effects. To test these hypotheses, we chose a panel of eight Gram-positive/Gram-negative bacterial strains, isolated from different biotopes and belonging to the species Escherichia coli, Lactobacillus rhamnosus, Lactococcus lactis (subsp. lactis and cremoris), Streptococcus thermophilus, and Lactobacillus sakei. Bacterial cells were exposed to food-grade E171 vs. P25 in vitro and the interactions were explored with innovative (nano)imaging methods. The ability of bacteria to trap TiO2 was demonstrated using synchrotron UV fluorescence imaging with single cell resolution. Subsequent alterations in the growth profiles were shown, notably for the transient food-borne L. lactis and the commensal intestinal E. coli in contact with food-grade TiO2. However, for both species, the reduction in cell cultivability remained moderate, and the morphological and ultrastructural damages, observed with electron microscopy, were restricted to a small number of cells. E. coli exposed to food-grade TiO2 showed some internalization of TiO2 (7% of cells), observed with high-resolution nano-secondary ion mass spectrometry (Nano-SIMS) chemical imaging. Taken together, these data show that E171 may be trapped by commensal and transient food-borne bacteria within the gut. In return, it may induce some physiological alterations in the most sensitive species, with a putative impact on gut microbiota composition and functioning, especially after chronic exposure.
Collapse
Affiliation(s)
| | - Pauline Talbot
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jasper B J Kamphuis
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Véronique Robert
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Christel Cartier
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Isabelle Fourquaux
- Faculté de Médecine Rangueil, Centre de Microscopie Electronique Appliquée à la Biologie (CMEAB), Toulouse, France
| | - Esther Lentzen
- Luxembourg Institute of Science and Technology (LIST), Material Research and Technology Department (MRT), Belvaux, Luxembourg
| | - Jean-Nicolas Audinot
- Luxembourg Institute of Science and Technology (LIST), Material Research and Technology Department (MRT), Belvaux, Luxembourg
| | | | | | - Jacek K Bardowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Philippe Langella
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Magdalena Kowalczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Eric Houdeau
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Thomas
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| |
Collapse
|
114
|
Cha KH, Lee EH, Yoon HS, Lee JH, Kim JY, Kang K, Park JS, Jin JB, Ko G, Pan CH. Effects of fermented milk treatment on microbial population and metabolomic outcomes in a three-stage semi-continuous culture system. Food Chem 2018; 263:216-224. [PMID: 29784310 DOI: 10.1016/j.foodchem.2018.04.095] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/18/2018] [Accepted: 04/22/2018] [Indexed: 12/14/2022]
Abstract
We investigated the impact of a fermented milk product on gut microbiota and their metabolism in 3 different conditions of the colon with a systemic viewpoint. An in vitro semi-continuous anaerobic cultivation was used to assess the colon compartment-specific influence of fermented milk, followed by a multiomics approach combining 16S rDNA amplicon sequencing and nuclear magnetic resonance (NMR) spectroscopy. The microbiome profiling and metabolomic features were significantly different across three colon compartments and after fermented milk treatment. Integrative correlation analysis indicated that the alteration of butyrate-producing microbiota (Veillonella, Roseburia, Lachnospira, and Coprococcus) and some primary metabolites (butyrate, ethanol, lactate, and isobutyrate) in the treatment group had a strong association with the fermented milk microorganisms. Our findings suggested that fermented milk treatment significantly affected microbial population in an in vitro cultivation system as well as the colonic metabolome in different ways in each of colon compartment.
Collapse
Affiliation(s)
- Kwang Hyun Cha
- Systems Biotechnology Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea; Department of Environmental Health Sciences, School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Eun Ha Lee
- Systems Biotechnology Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea.
| | - Hyo Shin Yoon
- Department of Environmental Health Sciences, School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jae Ho Lee
- R&BD Center, Korea Yakult Co. Ltd., Yongin 17086, Republic of Korea.
| | - Joo Yun Kim
- R&BD Center, Korea Yakult Co. Ltd., Yongin 17086, Republic of Korea.
| | - Kyungsu Kang
- Systems Biotechnology Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea.
| | - Jin-Soo Park
- Natural Constituents Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea.
| | - Jong Beom Jin
- Systems Biotechnology Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea.
| | - GwangPyo Ko
- Department of Environmental Health Sciences, School of Public Health, Seoul National University, Seoul 08826, Republic of Korea; Center for Human and Environmental Microbiome, Seoul National University, Seoul 08826, Republic of Korea; KoBioLabs, Inc., 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea.
| | - Cheol-Ho Pan
- Systems Biotechnology Research Center, KIST Gangneung Institute of Natural Products, Gangneung 25451, Republic of Korea.
| |
Collapse
|
115
|
Hornung B, Martins Dos Santos VAP, Smidt H, Schaap PJ. Studying microbial functionality within the gut ecosystem by systems biology. GENES AND NUTRITION 2018; 13:5. [PMID: 29556373 PMCID: PMC5840735 DOI: 10.1186/s12263-018-0594-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 02/13/2018] [Indexed: 12/13/2022]
Abstract
Humans are not autonomous entities. We are all living in a complex environment, interacting not only with our peers, but as true holobionts; we are also very much in interaction with our coexisting microbial ecosystems living on and especially within us, in the intestine. Intestinal microorganisms, often collectively referred to as intestinal microbiota, contribute significantly to our daily energy uptake by breaking down complex carbohydrates into simple sugars, which are fermented to short-chain fatty acids and subsequently absorbed by human cells. They also have an impact on our immune system, by suppressing or enhancing the growth of malevolent and beneficial microbes. Our lifestyle can have a large influence on this ecosystem. What and how much we consume can tip the ecological balance in the intestine. A "western diet" containing mainly processed food will have a different effect on our health than a balanced diet fortified with pre- and probiotics. In recent years, new technologies have emerged, which made a more detailed understanding of microbial communities and ecosystems feasible. This includes progress in the sequencing of PCR-amplified phylogenetic marker genes as well as the collective microbial metagenome and metatranscriptome, allowing us to determine with an increasing level of detail, which microbial species are in the microbiota, understand what these microorganisms do and how they respond to changes in lifestyle and diet. These new technologies also include the use of synthetic and in vitro systems, which allow us to study the impact of substrates and addition of specific microbes to microbial communities at a high level of detail, and enable us to gather quantitative data for modelling purposes. Here, we will review the current state of microbiome research, summarizing the computational methodologies in this area and highlighting possible outcomes for personalized nutrition and medicine.
Collapse
Affiliation(s)
- Bastian Hornung
- 1Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Vitor A P Martins Dos Santos
- 1Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Hauke Smidt
- 2Laboratory of Microbiology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Peter J Schaap
- 1Laboratory of Systems and Synthetic Biology, Wageningen University and Research, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| |
Collapse
|
116
|
Cook DP, Gysemans C, Mathieu C. Lactococcus lactis As a Versatile Vehicle for Tolerogenic Immunotherapy. Front Immunol 2018; 8:1961. [PMID: 29387056 PMCID: PMC5776164 DOI: 10.3389/fimmu.2017.01961] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022] Open
Abstract
Genetically modified Lactococcus lactis bacteria have been engineered as a tool to deliver bioactive proteins to mucosal tissues as a means to exert both local and systemic effects. They have an excellent safety profile, the result of years of human consumption in the food industry, as well as a lack of toxicity and immunogenicity. Also, containment strategies have been developed to promote further application as clinical protein-based therapeutics. Here, we review technological advancements made to enhanced the potential of L. lactis as live biofactories and discuss some examples of tolerogenic immunotherapies mediated by mucosal drug delivery via L. lactis. Additionally, we highlight their use to induce mucosal tolerance by targeted autoantigen delivery to the intestine as an approach to reverse autoimmune type 1 diabetes.
Collapse
Affiliation(s)
- Dana P Cook
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Conny Gysemans
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| |
Collapse
|
117
|
Draft Genome Sequence of Lactobacillus fermentum NCDC 400, Isolated from a Traditional Indian Dairy Product. GENOME ANNOUNCEMENTS 2018; 6:6/2/e01492-17. [PMID: 29326227 PMCID: PMC5764951 DOI: 10.1128/genomea.01492-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We announce here the draft genome sequence of Lactobacillus fermentum NCDC 400, a potential probiotic strain isolated from a traditional Indian dairy product. The genome size of Lactobacillus fermentum NCDC 400 is 1.89 Mb, and the assembled sequence consists of 185 contigs joined into 138 scaffolds.
Collapse
|
118
|
Mercier-Bonin M, Chapot-Chartier MP. Surface Proteins of Lactococcus lactis: Bacterial Resources for Muco-adhesion in the Gastrointestinal Tract. Front Microbiol 2017; 8:2247. [PMID: 29218032 PMCID: PMC5703838 DOI: 10.3389/fmicb.2017.02247] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/31/2017] [Indexed: 01/13/2023] Open
Abstract
Food and probiotic bacteria, in particular lactic acid bacteria, are ingested in large amounts by humans and are part of the transient microbiota which is increasingly considered to be able to impact the resident microbiota and thus possibly the host health. The lactic acid bacterium Lactococcus lactis is extensively used in starter cultures to produce dairy fermented food. Also because of a generally recognized as safe status, L. lactis has been considered as a possible vehicle to deliver in vivo therapeutic molecules with anti-inflammatory properties in the gastrointestinal tract. One of the key factors that may favor health effects of beneficial bacteria to the host is their capacity to colonize transiently the gut, notably through close interactions with mucus, which covers and protects the intestinal epithelium. Several L. lactis strains have been shown to exhibit mucus-binding properties and bacterial surface proteins have been identified as key determinants of such capacity. In this review, we describe the different types of surface proteins found in L. lactis, with a special focus on mucus-binding proteins and pili. We also review the different approaches used to investigate the adhesion of L. lactis to mucus, and particularly to mucins, one of its major components, and we present how these approaches allowed revealing the role of surface proteins in muco-adhesion.
Collapse
Affiliation(s)
- Muriel Mercier-Bonin
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | |
Collapse
|
119
|
Fernandez MA, Panahi S, Daniel N, Tremblay A, Marette A. Yogurt and Cardiometabolic Diseases: A Critical Review of Potential Mechanisms. Adv Nutr 2017; 8:812-829. [PMID: 29141967 PMCID: PMC5682997 DOI: 10.3945/an.116.013946] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Associations between yogurt intake and risk of diet-related cardiometabolic diseases (CMDs) have been the subject of recent research in epidemiologic nutrition. A healthy dietary pattern has been identified as a pillar for the prevention of weight gain and CMDs. Epidemiologic studies suggest that yogurt consumption is linked to healthy dietary patterns, lifestyles, and reduced risk of CMDs, particularly type 2 diabetes. However, to our knowledge, few to no randomized controlled trials have investigated yogurt intake in relation to cardiometabolic clinical outcomes. Furthermore, there has been little attempt to clarify the mechanisms that underlie the potential beneficial effects of yogurt consumption on CMDs. Yogurt is a nutrient-dense dairy food and has been suggested to reduce weight gain and prevent CMDs by contributing to intakes of protein, calcium, bioactive lipids, and several other micronutrients. In addition, fermentation with bacterial strains generates bioactive peptides, resulting in a potentially greater beneficial effect of yogurt on metabolic health than nonfermented dairy products such as milk. To date, there is little concrete evidence that the mechanisms proposed in observational studies to explain positive results of yogurt on CMDs or parameters are valid. Many proposed mechanisms are based on assumptions that commercial yogurts contain strain-specific probiotics, that viable yogurt cultures are present in adequate quantities, and that yogurt provides a minimum threshold dose of nutrients or bioactive components capable of exerting a physiologic effect. Therefore, the primary objective of this review is to investigate the plausibility of potential mechanisms commonly cited in the literature in order to shed light on the inverse associations reported between yogurt intake and various cardiometabolic health parameters that are related to its nutrient profile, bacterial constituents, and food matrix. This article reviews current gaps and challenges in identifying such mechanisms and provides a perspective on the research agenda to validate the proposed role of yogurt in protecting against CMDs.
Collapse
Affiliation(s)
- Melissa Anne Fernandez
- Heart and Lung Institute of Quebec, Laval University, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
- School of Nutrition, Faculty of Agriculture and Food Sciences, Laval University, Quebec, Canada
| | - Shirin Panahi
- Department of Kinesiology, Laval University, Quebec, Canada
| | - Noémie Daniel
- Heart and Lung Institute of Quebec, Laval University, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
- School of Nutrition, Faculty of Agriculture and Food Sciences, Laval University, Quebec, Canada
| | - Angelo Tremblay
- Heart and Lung Institute of Quebec, Laval University, Quebec, Canada
- School of Nutrition, Faculty of Agriculture and Food Sciences, Laval University, Quebec, Canada
- Department of Kinesiology, Laval University, Quebec, Canada
| | - André Marette
- Heart and Lung Institute of Quebec, Laval University, Quebec, Canada
- Institute of Nutrition and Functional Foods, Laval University, Quebec, Canada
- Department of Medicine, Faculty of Medicine, Laval University, Quebec, Canada
| |
Collapse
|
120
|
Dubinkina VB, Tyakht AV, Odintsova VY, Yarygin KS, Kovarsky BA, Pavlenko AV, Ischenko DS, Popenko AS, Alexeev DG, Taraskina AY, Nasyrova RF, Krupitsky EM, Shalikiani NV, Bakulin IG, Shcherbakov PL, Skorodumova LO, Larin AK, Kostryukova ES, Abdulkhakov RA, Abdulkhakov SR, Malanin SY, Ismagilova RK, Grigoryeva TV, Ilina EN, Govorun VM. Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. MICROBIOME 2017; 5:141. [PMID: 29041989 PMCID: PMC5645934 DOI: 10.1186/s40168-017-0359-2] [Citation(s) in RCA: 295] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 10/02/2017] [Indexed: 05/21/2023]
Abstract
BACKGROUND Alcohol abuse has deleterious effects on human health by disrupting the functions of many organs and systems. Gut microbiota has been implicated in the pathogenesis of alcohol-related liver diseases, with its composition manifesting expressed dysbiosis in patients suffering from alcoholic dependence. Due to its inherent plasticity, gut microbiota is an important target for prevention and treatment of these diseases. Identification of the impact of alcohol abuse with associated psychiatric symptoms on the gut community structure is confounded by the liver dysfunction. In order to differentiate the effects of these two factors, we conducted a comparative "shotgun" metagenomic survey of 99 patients with the alcohol dependence syndrome represented by two cohorts-with and without liver cirrhosis. The taxonomic and functional composition of the gut microbiota was subjected to a multifactor analysis including comparison with the external control group. RESULTS Alcoholic dependence and liver cirrhosis were associated with profound shifts in gut community structures and metabolic potential across the patients. The specific effects on species-level community composition were remarkably different between cohorts with and without liver cirrhosis. In both cases, the commensal microbiota was found to be depleted. Alcoholic dependence was inversely associated with the levels of butyrate-producing species from the Clostridiales order, while the cirrhosis-with multiple members of the Bacteroidales order. The opportunist pathogens linked to alcoholic dependence included pro-inflammatory Enterobacteriaceae, while the hallmarks of cirrhosis included an increase of oral microbes in the gut and more frequent occurrence of abnormal community structures. Interestingly, each of the two factors was associated with the expressed enrichment in many Bifidobacterium and Lactobacillus-but the exact set of the species was different between alcoholic dependence and liver cirrhosis. At the level of functional potential, the patients showed different patterns of increase in functions related to alcohol metabolism and virulence factors, as well as pathways related to inflammation. CONCLUSIONS Multiple shifts in the community structure and metabolic potential suggest strong negative influence of alcohol dependence and associated liver dysfunction on gut microbiota. The identified differences in patterns of impact between these two factors are important for planning of personalized treatment and prevention of these pathologies via microbiota modulation. Particularly, the expansion of Bifidobacterium and Lactobacillus suggests that probiotic interventions for patients with alcohol-related disorders using representatives of the same taxa should be considered with caution. Taxonomic and functional analysis shows an increased propensity of the gut microbiota to synthesis of the toxic acetaldehyde, suggesting higher risk of colorectal cancer and other pathologies in alcoholics.
Collapse
Affiliation(s)
- Veronika B. Dubinkina
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 1304 W. Springfield Avenue Urbana, Champaign, IL 61801 USA
- Carl R. Woese Institute for Genomic Biology, 1206 West Gregory Drive, Urbana, IL 61801 USA
| | - Alexander V. Tyakht
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
- ITMO University, Kronverkskiy pr. 49, Saint-Petersburg, 197101 Russia
| | - Vera Y. Odintsova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Konstantin S. Yarygin
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Boris A. Kovarsky
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Alexander V. Pavlenko
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Dmitry S. Ischenko
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Anna S. Popenko
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Dmitry G. Alexeev
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Anastasiya Y. Taraskina
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Bekhtereva 3, Saint-Petersburg, 192019 Russia
| | - Regina F. Nasyrova
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Bekhtereva 3, Saint-Petersburg, 192019 Russia
| | - Evgeny M. Krupitsky
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Bekhtereva 3, Saint-Petersburg, 192019 Russia
| | - Nino V. Shalikiani
- Moscow Clinical Scientific Center, Shosse Entuziastov 86, Moscow, 111123 Russia
| | - Igor G. Bakulin
- Moscow Clinical Scientific Center, Shosse Entuziastov 86, Moscow, 111123 Russia
| | - Petr L. Shcherbakov
- Moscow Clinical Scientific Center, Shosse Entuziastov 86, Moscow, 111123 Russia
| | - Lyubov O. Skorodumova
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Andrei K. Larin
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Elena S. Kostryukova
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | | | - Sayar R. Abdulkhakov
- Kazan State Medical University, Butlerova 49, Kazan, 420012 Russia
- Kazan Federal University, Kremlyovskaya 18, Kazan, 420008 Russia
| | | | | | | | - Elena N. Ilina
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| | - Vadim M. Govorun
- Moscow Institute of Physics and Technology, Institutskiy per. 9, Dolgoprudny, Moscow Region, 141700 Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine, Malaya Pirogovskaya 1a, Moscow, 119435 Russia
| |
Collapse
|
121
|
Abstract
Bifidobacteria are members of the intestinal microbiota of mammals and other animals, and some strains are able to exert health-promoting effects. The genus Bifidobacterium belongs to the Actinobacteria phylum. Firmicutes, Bacteroidetes, and Actinobacteria constitute the most abundant phyla in the human intestinal microbiota, Firmicutes and Bacteroidetes being predominant in adults, and Actinobacteria in breast-fed infants, where bifidobacteria can reach levels higher than 90% of the total bacterial population. They are among the first microbial colonizers of the intestines of newborns, and play key roles in the development of their physiology, including maturation of the immune system and use of dietary components. Indeed, some nutrients, such as human milk oligosaccharides, are important drivers of bifidobacterial development. Some Bifidobacterium strains are considered probiotic microorganisms because of their beneficial effects, and they have been included as bioactive ingredients in functional foods, mainly dairy products, as well as in food supplements and pharma products, alone, or together with, other microbes or microbial substrates. Well-documented scientific evidence of their activities is currently available for bifidobacteria-containing preparations in some intestinal and extraintestinal pathologies. In this review, we focus on the role of bifidobacteria as members of the human intestinal microbiota and their use as probiotics in the prevention and treatment of disease.
Collapse
|
122
|
Tao L, Wang B, Zhong Y, Pow SH, Zeng X, Qin C, Zhang P, Chen S, He W, Tan Y, Liu H, Jiang Y, Chen W, Chen YZ. Database and Bioinformatics Studies of Probiotics. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7599-7606. [PMID: 28727425 DOI: 10.1021/acs.jafc.7b01815] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Probiotics have been widely explored for health benefits, animal cares, and agricultural applications. Recent advances in microbiome, microbiota, and microbial dark matter research have fueled greater interests in and paved ways for the study of the mechanisms of probiotics and the discovery of new probiotics from uncharacterized microbial sources. A probiotics database named PROBIO was developed to facilitate these efforts and the need for the information on the known probiotics, which provides the comprehensive information about the probiotic functions of 448 marketed, 167 clinical trial/field trial, and 382 research probiotics for use or being studied for use in humans, animals, and plants. The potential applications of the probiotics data are illustrated by several literature-reported investigations, which have used the relevant information for probing the function and mechanism of the probiotics and for discovering new probiotics. PROBIO can be accessed free of charge at http://bidd2.nus.edu.sg/probio/homepage.htm .
Collapse
Affiliation(s)
- Lin Tao
- School of Medicine, Hangzhou Normal University , Hangzhou, P. R. China 310012
| | - Bohua Wang
- College of Life and Environmental Sciences, Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Hunan University of Arts and Science , Changde, Hunan, P. R. China 415000
- Key Lab of Agricultural Products Processing and Quality Control of Nanchang City, Jiangxi Agricultural University , Nanchang, P. R. China 330045
| | - Yafen Zhong
- Key Lab of Agricultural Products Processing and Quality Control of Nanchang City, Jiangxi Agricultural University , Nanchang, P. R. China 330045
| | - Siok Hoon Pow
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Xian Zeng
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Chu Qin
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Peng Zhang
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Shangying Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Weidong He
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| | - Ying Tan
- The Key Laboratory of Chemical Biology, Guangdong Province, Graduate School at Shenzhen, Tsinghua University , Shenzhen, Guangdong, P. R. China 518055
| | - Hongxia Liu
- The Key Laboratory of Chemical Biology, Guangdong Province, Graduate School at Shenzhen, Tsinghua University , Shenzhen, Guangdong, P. R. China 518055
| | - Yuyang Jiang
- The Key Laboratory of Chemical Biology, Guangdong Province, Graduate School at Shenzhen, Tsinghua University , Shenzhen, Guangdong, P. R. China 518055
| | - Weiping Chen
- Key Lab of Agricultural Products Processing and Quality Control of Nanchang City, Jiangxi Agricultural University , Nanchang, P. R. China 330045
| | - Yu Zong Chen
- Bioinformatics and Drug Design Group, Department of Pharmacy, and Center for Computational Science and Engineering, National University of Singapore , Singapore 117543
| |
Collapse
|
123
|
Matsumoto M, Kitada Y, Shimomura Y, Naito Y. Bifidobacterium animalis subsp. lactis LKM512 reduces levels of intestinal trimethylamine produced by intestinal microbiota in healthy volunteers: A double-blind, placebo-controlled study. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
124
|
Corsello G, Carta M, Marinello R, Picca M, De Marco G, Micillo M, Ferrara D, Vigneri P, Cecere G, Ferri P, Roggero P, Bedogni G, Mosca F, Paparo L, Nocerino R, Berni Canani R. Preventive Effect of Cow's Milk Fermented with Lactobacillus paracasei CBA L74 on Common Infectious Diseases in Children: A Multicenter Randomized Controlled Trial. Nutrients 2017; 9:E669. [PMID: 28654019 PMCID: PMC5537784 DOI: 10.3390/nu9070669] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 12/13/2022] Open
Abstract
Background: Fermented foods have been proposed to prevent common infectious diseases (CIDs) in children attending day care or preschool. OBJECTIVES To investigate the efficacy of dietary supplementation with cow's skim milk fermented with the probiotic Lactobacillus paracasei CBA L74 in reducing CIDs in children attending day care or preschool. Methods: Multicenter, randomized, double-blind, placebo-controlled trial on healthy children (aged 12-48 months) consuming daily 7 grams of cow's skim milk fermented with L. paracasei CBA L74 (group A), or placebo (maltodextrins group B) attending day care or preschool during the winter season. The main outcome was the proportion of children who experienced ≥1 episode of CID during a 3-month follow-up. Fecal biomarkers of innate (α- and β-defensins, cathelicidin) and acquired immunity (secretory IgA) were also monitored. Results: A total of 126 children (71 males, 56%) with a mean (SD) age of 33 (9) months completed the study, 66 in group A and 60 in group B. At intention to treat analysis, the proportion of children presenting ≥1 CID was 60% in group A vs. 83% in group B, corresponding to an absolute risk difference (ARD) of -23% (95% CI: -37% to -9%, p < 0.01). At per-protocol-analysis (PPA), the proportion of children presenting ≥1 CID was 18% in group A vs. 40% in group B, corresponding to an absolute risk difference (ARD) of -22% (95% CI: -37% to -6%, p < 0.01). PPA showed that the proportion of children presenting ≥1 acute gastroenteritis (AGE) was significantly lower in group A (18% vs. 40%, p < 0.05). The ARD for the occurrence of ≥1 AGE was -22% (95% CI: -37% to -6%, p < 0.01) in group A. Similar findings were obtained at PPA regarding the proportion of children presenting ≥1 upper respiratory tract infection (URTI), which was significantly lower in group A (51% vs. 74%, p < 0.05), corresponding to an ARD of -23% (95% CI: -40% to -7%, p < 0.01). Significant changes in innate and acquired immunity biomarkers were observed only in subjects in group A. Conclusions: Dietary supplementation with cow's skim milk fermented with L. paracasei CBA L74 is an efficient strategy in preventing CIDs in children.
Collapse
Affiliation(s)
- Giovanni Corsello
- Operative Unit of Pediatrics and Neonatal Intensive Therapy, Mother and Child Department, University of Palermo, 90121 Palermo, Italy.
| | - Maurizio Carta
- Operative Unit of Pediatrics and Neonatal Intensive Therapy, Mother and Child Department, University of Palermo, 90121 Palermo, Italy.
| | - Roberto Marinello
- Federazione Italiana Medici Pediatri Lombardia, 46100 Mantova, Italy.
| | - Marina Picca
- Pediatric Society of Primary Health Care (SICuPP), 20135 Milan, Italy.
| | - Giulio De Marco
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Maria Micillo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Dante Ferrara
- Department of Sciences for Health Promotion and Mother and Child Care, University of Palermo, 90121 Palermo, Italy.
| | | | - Gaetano Cecere
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Pasqualina Ferri
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Paola Roggero
- Department of Clinical Science and Community Health, Neonatal Intensive Care Unit, Fondazione I.R.C.C.S. Cà Granda Ospedale Maggiore Policlinico, University of Milan, 20143 Milan, Italy.
| | - Giorgio Bedogni
- Clinical Epidemiology Unit, Liver Research Center, Basovizza, 34012 Trieste, Italy.
| | - Fabio Mosca
- Department of Clinical Science and Community Health, Neonatal Intensive Care Unit, Fondazione I.R.C.C.S. Cà Granda Ospedale Maggiore Policlinico, University of Milan, 20143 Milan, Italy.
| | - Lorella Paparo
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Rita Nocerino
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
| | - Roberto Berni Canani
- Department of Translational Medical Science-Pediatric Section, University of Naples "Federico II" Via S. Pansini, 5, 80131 Naples, Italy.
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples "Federico II", 80131 Naples, Italy.
- CEINGE Advanced Biotechnologies, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
125
|
Wen L, Duffy A. Factors Influencing the Gut Microbiota, Inflammation, and Type 2 Diabetes. J Nutr 2017; 147:1468S-1475S. [PMID: 28615382 DOI: 10.3945/jn.116.240754] [Citation(s) in RCA: 214] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/12/2016] [Accepted: 01/11/2017] [Indexed: 12/17/2022] Open
Abstract
The gut microbiota is a complex community of bacteria residing in the intestine. Animal models have demonstrated that several factors contribute to and can significantly alter the composition of the gut microbiota, including genetics; the mode of delivery at birth; the method of infant feeding; the use of medications, especially antibiotics; and the diet. There may exist a gut microbiota signature that promotes intestinal inflammation and subsequent systemic low-grade inflammation, which in turn promotes the development of type 2 diabetes. There are preliminary studies that suggest that the consumption of probiotic bacteria such as those found in yogurt and other fermented milk products can beneficially alter the composition of the gut microbiome, which in turn changes the host metabolism. Obesity, insulin resistance, fatty liver disease, and low-grade peripheral inflammation are more prevalent in patients with low α diversity in the gut microbiome than they are in patients with high α diversity. Fermented milk products, such as yogurt, deliver a large number of lactic acid bacteria to the gastrointestinal tract. They may modify the intestinal environment, including inhibiting lipopolysaccharide production and increasing the tight junctions of gut epithelia cells.
Collapse
Affiliation(s)
- Li Wen
- Section of Endocrinology and
| | - Andrew Duffy
- Department of Surgery, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
126
|
Probiotic yogurt and acidified milk similarly reduce postprandial inflammation and both alter the gut microbiota of healthy, young men. Br J Nutr 2017; 117:1312-1322. [PMID: 28558854 DOI: 10.1017/s0007114517000885] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Probiotic yogurt and milk supplemented with probiotics have been investigated for their role in 'low-grade' inflammation but evidence for their efficacy is inconclusive. This study explores the impact of probiotic yogurt on metabolic and inflammatory biomarkers, with a parallel study of gut microbiota dynamics. The randomised cross-over study was conducted in fourteen healthy, young men to test probiotic yogurt compared with milk acidified with 2 % d-(+)-glucono-δ-lactone during a 2-week intervention (400 g/d). Fasting assessments, a high-fat meal test (HFM) and microbiota analyses were used to assess the intervention effects. Baseline assessments for the HFM were carried out after a run-in during which normal milk was provided. No significant differences in the inflammatory response to the HFM were observed after probiotic yogurt compared with acidified milk intake; however, both products were associated with significant reductions in the inflammatory response to the HFM compared with the baseline tests (assessed by IL6, TNFα and chemokine ligand 5) (P<0·001). These observations were accompanied by significant changes in microbiota taxa, including decreased abundance of Bilophila wadsworthia after acidified milk (log 2-fold-change (FC)=-1·5, P adj=0·05) and probiotic yogurt intake (FC=-1·3, P adj=0·03), increased abundance of Bifidobacterium species after acidified milk intake (FC=1·4, P adj=0·04) and detection of Lactobacillus delbrueckii spp. bulgaricus (FC=7·0, P adj<0·01) and Streptococcus salivarius spp. thermophilus (FC=6·0, P adj<0·01) after probiotic yogurt intake. Probiotic yogurt and acidified milk similarly reduce postprandial inflammation that is associated with a HFM while inducing distinct changes in the gut microbiota of healthy men. These observations could be relevant for dietary treatments that target 'low-grade' inflammation.
Collapse
|
127
|
Dimidi E, Christodoulides S, Scott SM, Whelan K. Mechanisms of Action of Probiotics and the Gastrointestinal Microbiota on Gut Motility and Constipation. Adv Nutr 2017; 8:484-494. [PMID: 28507013 PMCID: PMC5421123 DOI: 10.3945/an.116.014407] [Citation(s) in RCA: 244] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Constipation is a common and burdensome gastrointestinal disorder that may result from altered gastrointestinal motility. The effect of probiotics on constipation has been increasingly investigated in both animal and human studies, showing promising results. However, there is still uncertainty regarding the mechanisms of action of probiotics on gut motility and constipation. Several factors are vital to normal gut motility, including immune and nervous system function, bile acid metabolism and mucus secretion, and the gastrointestinal microbiota and fermentation; an imbalance or dysfunction in any of these components may contribute to aberrant gut motility and, consequently, symptoms of constipation. For example, adults with functional constipation have significantly decreased numbers of bifidobacteria (with one study showing a mean difference of 1 log10/g) and lactobacilli (mean difference, 1.4 log10/g) in stool samples, as well as higher breath methane, compared with control subjects. Modifying the gut luminal environment with certain probiotic strains may affect motility and secretion in the gut and, hence, provide a benefit for patients with constipation. Therefore, this review explores the mechanisms through which probiotics may exert an effect on gut motility and constipation. Nevertheless, the majority of current evidence is derived from animal studies, and therefore, further human studies are needed to determine the mechanisms through specific probiotic strains that might be effective in constipation.
Collapse
Affiliation(s)
- Eirini Dimidi
- Faculty of Life Sciences and Medicine, Diabetes and Nutritional Sciences Division, King’s College London, London, United Kingdom; and,Centre for Neuroscience and Trauma, Neurogastroenterology Group and GI Physiology Unit, Queen Mary University of London, London, United Kingdom
| | - Stephanos Christodoulides
- Faculty of Life Sciences and Medicine, Diabetes and Nutritional Sciences Division, King’s College London, London, United Kingdom; and,Centre for Neuroscience and Trauma, Neurogastroenterology Group and GI Physiology Unit, Queen Mary University of London, London, United Kingdom
| | - S Mark Scott
- Centre for Neuroscience and Trauma, Neurogastroenterology Group and GI Physiology Unit, Queen Mary University of London, London, United Kingdom
| | - Kevin Whelan
- Faculty of Life Sciences and Medicine, Diabetes and Nutritional Sciences Division, King's College London, London, United Kingdom; and
| |
Collapse
|
128
|
Lim SM, Kim DH. Bifidobacterium adolescentis IM38 ameliorates high-fat diet-induced colitis in mice by inhibiting NF-κB activation and lipopolysaccharide production by gut microbiota. Nutr Res 2017; 41:86-96. [PMID: 28479226 DOI: 10.1016/j.nutres.2017.04.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/08/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023]
Abstract
Gut microbiota play essential roles in the regulation of human metabolism via symbiotic interactions with the host. Prolonged consumption of high-fat diet (HFD) elevates the Firmicutes to Bacteroidetes ratio and lipopolysaccharide (LPS) production by gut microbiota, thereby increasing the probability of developing metabolic and immune disorders such as obesity and colitis. The use of probiotics with anti-inflammatory properties has been suggested to counteract this effect. Here, we tested whether Bifidobacterium adolescentis IM38, which inhibited nuclear factor-kappa B (NF-κB) activation in Caco-2 cells and peritoneal macrophages and inhibited Escherichia coli LPS production, exerted an anticolitic effect in mice with HFD-induced obesity. Oral administration of IM38 (2×109CFU/mouse per day) for 6 weeks in mice with HFD-induced obesity inhibited whole-body and epididymal fat weight gain. IM38 also increased HFD-suppressed expression of interleukin (IL)-10 and tight junction proteins but significantly downregulated HFD-induced NF-κB activation and tumor necrosis factor expression in the colon. IM38 inhibited differentiation into helper T17 cells and reduced IL-17 levels in the colon of mice with HFD-induced obesity but increased HFD-suppressed differentiation into regulatory T cells and IL-10 levels. Furthermore, treatment with IM38 lowered the HFD-induced LPS levels in blood and colonic fluid, as well as the Proteobacteria to Bacteroidetes ratio in gut microbiota. Therefore, we suggest that IM38 can inhibit HFD-induced LPS production in gut microbiota through the regulation of Proteobacteria to Bacteroidetes ratio and NF-κB activation in the colon, which ultimately attenuates colitis. Thus, IM38 may be a suitable ingredient of functional foods designed for treating or preventing colitis.
Collapse
Affiliation(s)
- Su-Min Lim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dong-Hyun Kim
- Department of Life and Nanopharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
129
|
Gomez DE, Arroyo LG, Costa MC, Viel L, Weese JS. Characterization of the Fecal Bacterial Microbiota of Healthy and Diarrheic Dairy Calves. J Vet Intern Med 2017; 31:928-939. [PMID: 28390070 PMCID: PMC5435056 DOI: 10.1111/jvim.14695] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/13/2017] [Accepted: 02/21/2017] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Neonatal diarrhea accounts for more than 50% of total deaths in dairy calves. Few population-based studies of cattle have investigated how the microbiota is impacted during diarrhea. OBJECTIVES To characterize the fecal microbiota and predict the functional potential of the microbial communities in healthy and diarrheic calves. METHODS Fifteen diarrheic calves between the ages of 1 and 30 days and 15 age-matched healthy control calves were enrolled from 2 dairy farms. The Illumina MiSeq sequencer was used for high-throughput sequencing of the V4 region of the 16S rRNA gene (Illumina, San Diego, CA). RESULTS Significant differences in community membership and structure were identified among healthy calves from different farms. Differences in community membership and structure also were identified between healthy and diarrheic calves within each farm. Based on linear discriminant analysis effect size (LEfSe), the genera Bifidobacterium, Megamonas, and a genus of the family Bifidobacteriaceae were associated with health at farm 1, whereas Lachnospiraceae incertae sedis, Dietzia and an unclassified genus of the family Veillonellaceae were significantly associated with health at farm 2. The Phylogenetic Investigation of Communities Reconstruction of Unobserved States (PICRUSt) analysis indicated that diarrheic calves had decreased abundances of genes responsible for metabolism of various vitamins, amino acids, and carbohydrate. CLINICAL RELEVANCE The fecal microbiota of healthy dairy calves appeared to be farm specific as were the changes observed during diarrhea. The differences in microbiota structure and membership between healthy and diarrheic calves suggest that dysbiosis can occur in diarrheic calves and it is associated with changes in predictive metagenomic function.
Collapse
Affiliation(s)
- D E Gomez
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - L G Arroyo
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - M C Costa
- Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - L Viel
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - J S Weese
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
130
|
Guo J, Astrup A, Lovegrove JA, Gijsbers L, Givens DI, Soedamah-Muthu SS. Milk and dairy consumption and risk of cardiovascular diseases and all-cause mortality: dose-response meta-analysis of prospective cohort studies. Eur J Epidemiol 2017; 32:269-287. [PMID: 28374228 PMCID: PMC5437143 DOI: 10.1007/s10654-017-0243-1] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 03/27/2017] [Indexed: 02/07/2023]
Abstract
With a growing number of prospective cohort studies, an updated dose-response meta-analysis of milk and dairy products with all-cause mortality, coronary heart disease (CHD) or cardiovascular disease (CVD) have been conducted. PubMed, Embase and Scopus were searched for articles published up to September 2016. Random-effect meta-analyses with summarised dose-response data were performed for total (high-fat/low-fat) dairy, milk, fermented dairy, cheese and yogurt. Non-linear associations were investigated using the spine models and heterogeneity by subgroup analyses. A total of 29 cohort studies were available for meta-analysis, with 938,465 participants and 93,158 mortality, 28,419 CHD and 25,416 CVD cases. No associations were found for total (high-fat/low-fat) dairy, and milk with the health outcomes of mortality, CHD or CVD. Inverse associations were found between total fermented dairy (included sour milk products, cheese or yogurt; per 20 g/day) with mortality (RR 0.98, 95% CI 0.97-0.99; I2 = 94.4%) and CVD risk (RR 0.98, 95% CI 0.97-0.99; I2 = 87.5%). Further analyses of individual fermented dairy of cheese and yogurt showed cheese to have a 2% lower risk of CVD (RR 0.98, 95% CI 0.95-1.00; I2 = 82.6%) per 10 g/day, but not yogurt. All of these marginally inverse associations of totally fermented dairy and cheese were attenuated in sensitivity analyses by removing one large Swedish study. This meta-analysis combining data from 29 prospective cohort studies demonstrated neutral associations between dairy products and cardiovascular and all-cause mortality. For future studies it is important to investigate in more detail how dairy products can be replaced by other foods.
Collapse
Affiliation(s)
- Jing Guo
- Institute for Food, Nutrition and Health, University of Reading, Reading, RG6 6AR UK
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Julie A. Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Institute for Cardiovascular and Metabolic Research, University of Reading, Reading, RG6 6AP UK
| | - Lieke Gijsbers
- Division of Human Nutrition, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - David I. Givens
- Institute for Food, Nutrition and Health, University of Reading, Reading, RG6 6AR UK
| | - Sabita S. Soedamah-Muthu
- Division of Human Nutrition, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| |
Collapse
|
131
|
Walsh AM, Crispie F, Claesson MJ, Cotter PD. Translating Omics to Food Microbiology. Annu Rev Food Sci Technol 2017; 8:113-134. [DOI: 10.1146/annurev-food-030216-025729] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Aaron M. Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Marcus J. Claesson
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
132
|
Rethinking Diet to Aid Human–Microbe Symbiosis. Trends Microbiol 2017; 25:100-112. [DOI: 10.1016/j.tim.2016.09.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/19/2016] [Accepted: 09/26/2016] [Indexed: 01/06/2023]
|
133
|
In vitro toxicity assessment of oral nanocarriers. Adv Drug Deliv Rev 2016; 106:381-401. [PMID: 27544694 DOI: 10.1016/j.addr.2016.08.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 02/08/2023]
Abstract
The fascinating properties of nanomaterials opened new frontiers in medicine. Nanocarriers are useful systems in transporting drugs to site-specific targets. The unique physico-chemical characteristics making nanocarriers promising devices to treat diseases may also be responsible for potential adverse effects. In order to develop functional nano-based drug delivery systems, efficacy and safety should be carefully evaluated. To date, no common testing strategy to address nanomaterial toxicological challenges has been generated. Different cell culture models are currently used to evaluate nanocarrier safety using conventional in vitro assays, but overall they have generated a huge amount of conflicting data. In this review we describe state-of-the-art approaches for in vitro testing of orally administered nanocarriers, highlighting the importance of developing harmonized and validated standard operating procedures. These procedures should be applied in a safe-by-design context with the aim to reduce and/or eliminate the uncertainties and risks associated with nanomedicine development.
Collapse
|
134
|
Hsieh YH, Peterson CM, Raggio A, Keenan MJ, Martin RJ, Ravussin E, Marco ML. Impact of Different Fecal Processing Methods on Assessments of Bacterial Diversity in the Human Intestine. Front Microbiol 2016; 7:1643. [PMID: 27812352 PMCID: PMC5071325 DOI: 10.3389/fmicb.2016.01643] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 10/03/2016] [Indexed: 12/14/2022] Open
Abstract
The intestinal microbiota are integral to understanding the relationships between nutrition and health. Therefore, fecal sampling and processing protocols for metagenomic surveys should be sufficiently robust, accurate, and reliable to identify the microorganisms present. We investigated the use of different fecal preparation methods on the bacterial community structures identified in human stools. Complete stools were collected from six healthy individuals and processed according to the following methods: (i) randomly sampled fresh stool, (ii) fresh stool homogenized in a blender for 2 min, (iii) randomly sampled frozen stool, and (iv) frozen stool homogenized in a blender for 2 min, or (v) homogenized in a pneumatic mixer for either 10, 20, or 30 min. High-throughput DNA sequencing of the 16S rRNA V4 regions of bacterial community DNA extracted from the stools showed that the fecal microbiota remained distinct between individuals, independent of processing method. Moreover, the different stool preparation approaches did not alter intra-individual bacterial diversity. Distinctions were found at the level of individual taxa, however. Stools that were frozen and then homogenized tended to have higher proportions of Faecalibacterium, Streptococcus, and Bifidobacterium and decreased quantities of Oscillospira, Bacteroides, and Parabacteroides compared to stools that were collected in small quantities and not mixed prior to DNA extraction. These findings indicate that certain taxa are at particular risk for under or over sampling due to protocol differences. Importantly, homogenization by any method significantly reduced the intra-individual variation in bacteria detected per stool. Our results confirm the robustness of fecal homogenization for microbial analyses and underscore the value of collecting and mixing large stool sample quantities in human nutrition intervention studies.
Collapse
Affiliation(s)
- Yu-Hsin Hsieh
- Department of Food Science and Technology, University of California, Davis, DavisCA, USA; Agricultural Biotechnology Center, National Chung Hsing UniversityTaichung, Taiwan
| | | | - Anne Raggio
- Louisiana State University Agricultural Center, Baton Rouge LA, USA
| | - Michael J Keenan
- Louisiana State University Agricultural Center, Baton Rouge LA, USA
| | - Roy J Martin
- Western Human Nutrition Research Center, Davis CA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge LA, USA
| | - Maria L Marco
- Department of Food Science and Technology, University of California, Davis, Davis CA, USA
| |
Collapse
|
135
|
Sánchez B, Delgado S, Blanco-Míguez A, Lourenço A, Gueimonde M, Margolles A. Probiotics, gut microbiota, and their influence on host health and disease. Mol Nutr Food Res 2016; 61. [PMID: 27500859 DOI: 10.1002/mnfr.201600240] [Citation(s) in RCA: 573] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/29/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022]
Abstract
The gastrointestinal tract of mammals hosts a high and diverse number of different microorganisms, known as intestinal microbiota. Many probiotics were originally isolated from the gastrointestinal tract, and they were defined by the Food and Agriculture Organization of the United Nations (FAO)/WHO as "live microorganisms which when administered in adequate amounts confer a health benefit on the host." Probiotics exert their beneficial effects on the host through four main mechanisms: interference with potential pathogens, improvement of barrier function, immunomodulation and production of neurotransmitters, and their host targets vary from the resident microbiota to cellular components of the gut-brain axis. However, in spite of the wide array of beneficial mechanisms deployed by probiotic bacteria, relatively few effects have been supported by clinical data. In this regard, different probiotic strains have been effective in antibiotic-associated diarrhea or inflammatory bowel disease for instance. The aim of this review was to compile the molecular mechanisms underlying the beneficial effects of probiotics, mainly through their interaction with the intestinal microbiota and with the intestinal mucosa. The specific benefits discussed in this paper include among others those elicited directly through dietary modulation of the human gut microbiota.
Collapse
Affiliation(s)
- Borja Sánchez
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Villaviciosa, Asturias, Spain
| | - Susana Delgado
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Villaviciosa, Asturias, Spain
| | - Aitor Blanco-Míguez
- ESEI - Department of Computer Science, University of Vigo, Edificio Politécnico, Campus Universitario As Lagoas s/n 32004, Ourense, Spain
| | - Anália Lourenço
- ESEI - Department of Computer Science, University of Vigo, Edificio Politécnico, Campus Universitario As Lagoas s/n 32004, Ourense, Spain.,CEB - Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Villaviciosa, Asturias, Spain
| | - Abelardo Margolles
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA), Consejo Superior de Investigaciones Científicas (CSIC), Villaviciosa, Asturias, Spain
| |
Collapse
|
136
|
Stilling RM, van de Wouw M, Clarke G, Stanton C, Dinan TG, Cryan JF. The neuropharmacology of butyrate: The bread and butter of the microbiota-gut-brain axis? Neurochem Int 2016; 99:110-132. [DOI: 10.1016/j.neuint.2016.06.011] [Citation(s) in RCA: 331] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/30/2016] [Accepted: 06/21/2016] [Indexed: 02/07/2023]
|
137
|
Consumption of a Bifidobacterium bifidum Strain for 4 Weeks Modulates Dominant Intestinal Bacterial Taxa and Fecal Butyrate in Healthy Adults. Appl Environ Microbiol 2016; 82:5850-9. [PMID: 27451450 DOI: 10.1128/aem.01753-16] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/15/2016] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED Modulation of the intestinal microbial ecosystem (IME) is a useful target to establish probiotic efficacy in a healthy population. We conducted a randomized, double-blind, crossover, and placebo-controlled intervention study to determine the impact of Bifidobacterium bifidum strain Bb on the IME of adult healthy volunteers of both sexes. High-throughput 16S rRNA gene sequencing was used to characterize the fecal microbiota before and after 4 weeks of daily probiotic cell consumption. The intake of approximately one billion live B. bifidum cells affected the relative abundance of dominant taxa in the fecal microbiota and modulated fecal butyrate levels. Specifically, Prevotellaceae (P = 0.041) and Prevotella (P = 0.034) were significantly decreased, whereas Ruminococcaceae (P = 0.039) and Rikenellaceae (P = 0.010) were significantly increased. We also observed that the probiotic intervention modulated the fecal concentrations of butyrate in a manner dependent on the initial levels of short-chain fatty acids (SCFAs). In conclusion, our study demonstrates that a single daily administration of Bifidobacterium bifidum strain Bb can significantly modify the IME in healthy (not diseased) adults. These findings demonstrate the need to reassess the notion that probiotics do not influence the complex and stable IME of a healthy individual. IMPORTANCE Foods and supplements claimed to contain health-promoting probiotic microorganisms are everywhere these days and mainly intended for consumption by healthy people. However, it is still debated what actual effects probiotic products may have on the healthy population. In this study, we report the results of an intervention trial aimed at assessing the modifications induced in the intestinal microbial ecosystem of healthy adults from the consumption of a probiotic product. Our results demonstrate that the introduction of a probiotic product in the dietary habits of healthy people may significantly modify dominant taxa of the intestinal microbiota, resulting in the modulation of short-chain fatty acid concentrations in the gut. The overall changes witnessed in the probiotic intervention indicate a mechanism of microbiota modulation that could have potential effects on human health.
Collapse
|
138
|
Aizawa E, Tsuji H, Asahara T, Takahashi T, Teraishi T, Yoshida S, Ota M, Koga N, Hattori K, Kunugi H. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord 2016; 202:254-7. [PMID: 27288567 DOI: 10.1016/j.jad.2016.05.038] [Citation(s) in RCA: 382] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/21/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Bifidobacterium and Lactobacillus in the gut have been suggested to have a beneficial effect on stress response and depressive disorder. We examined whether these bacterial counts are reduced in patients with major depressive disorder (MDD) than in healthy controls. METHOD Bifidobacterium and Lactobacillus counts in fecal samples were estimated in 43 patients and 57 controls using bacterial rRNA-targeted reverse transcription-quantitative polymerase chain reaction RESULTS The patients had significantly lower Bifidobacterium counts (P=0.012) and tended to have lower Lactobacillus counts (P=0.067) than the controls. Individuals whose bacterial counts below the optimal cut-off point (9.53 and 6.49log10 cells/g for Bifidobacterium and Lactobacillus, respectively) were significantly more common in the patients than in the controls for both bacteria (Bifidobacterium: odds ratio 3.23, 95% confidence interval [CI] 1.38-7.54, P=0.010; Lactobacillus: 2.57, 95% CI 1.14-5.78, P=0.027). Using the same cut-off points, we observed an association between the bacterial counts and Irritable bowel syndrome. Frequency of fermented milk consumption was associated with higher Bifidobacterium counts in the patients. LIMITATIONS The findings should be interpreted with caution since effects of gender and diet were not fully taken into account in the analysis. CONCLUSION Our results provide direct evidence, for the first time, that individuals with lower Bifidobacterium and/or Lactobacillus counts are more common in patients with MDD compared to controls. Our findings provide new insight into the pathophysiology of MDD and will enhance future research on the use of pro- and prebiotics in the treatment of MDD.
Collapse
Affiliation(s)
- Emiko Aizawa
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | | | | | - Toshiya Teraishi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Sumiko Yoshida
- Department of Psychiatry, National Center of Neurology and Psychiatry Hospital, Tokyo, Japan
| | - Miho Ota
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Norie Koga
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kotaro Hattori
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.
| |
Collapse
|
139
|
Bogovič Matijašić B, Obermajer T, Lipoglavšek L, Sernel T, Locatelli I, Kos M, Šmid A, Rogelj I. Effects of synbiotic fermented milk containing Lactobacillus acidophilus La-5 and Bifidobacterium animalis ssp. lactis BB-12 on the fecal microbiota of adults with irritable bowel syndrome: A randomized double-blind, placebo-controlled trial. J Dairy Sci 2016; 99:5008-5021. [PMID: 27157575 DOI: 10.3168/jds.2015-10743] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/23/2016] [Indexed: 12/12/2022]
Abstract
We conducted a randomized double-blind, placebo-controlled multicentric study to investigate the influence of a synbiotic fermented milk on the fecal microbiota composition of 30 adults with irritable bowel syndrome (IBS). The synbiotic product contained Lactobacillus acidophilus La-5, Bifidobacterium animalis ssp. lactis BB-12, Streptococcus thermophilus, and dietary fiber (90% inulin, 10% oligofructose), and a heat-treated fermented milk without probiotic bacteria or dietary fiber served as placebo. Stool samples were collected after a run-in period, a 4-wk consumption period, and a 1-wk follow-up period, and were subjected to real-time PCR and 16S rDNA profiling by next-generation sequencing. After 4wk of synbiotic (11 subjects) or placebo (19 subjects) consumption, a greater increase in DNA specific for L. acidophilus La-5 and Bifidobacterium animalis ssp. lactis was detected in the feces of the synbiotic group compared with the placebo group by quantitative real-time PCR. After 1wk of follow-up, the content of L. acidophilus La-5 and B. animalis ssp. lactis decreased to levels close to initial levels. No significant changes with time or differences between the groups were observed for Lactobacillus, Enterobacteriaceae, Bifidobacterium, or all bacteria. The presence of viable BB-12- and La-5-like bacteria in the feces resulting from the intake of synbiotic product was confirmed by random amplification of polymorphic DNA (RAPD)-PCR. At the end of consumption period, the feces of all subjects assigned to the synbiotic group contained viable bacteria with a BB-12-like RAPD profile, and after 1wk of follow-up, BB-12-like bacteria remained in the feces of 87.5% of these subjects. The presence of La-5-like colonies was observed less frequently (37.5 and 25% of subjects, respectively). Next-generation sequencing of 16S rDNA amplicons revealed that only the percentage of sequences assigned to Strep. thermophilus was temporarily increased in both groups, whereas the global profile of the fecal microbiota of patients was not altered by consumption of the synbiotic or placebo. In conclusion, daily consumption of a synbiotic fermented milk had a short-term effect on the amount and proportion of La-5-like strains and B. animalis ssp. lactis in the fecal microbiome of IBS patients. Furthermore, both synbiotic and placebo products caused a temporary increase in fecal Strep. thermophilus.
Collapse
Affiliation(s)
- Bojana Bogovič Matijašić
- Institute of Dairy Science and Probiotics, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1230 Domžale, Slovenia.
| | - Tanja Obermajer
- Institute of Dairy Science and Probiotics, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1230 Domžale, Slovenia
| | - Luka Lipoglavšek
- Division of Microbiology and Microbial Biotechnology, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1230 Domžale, Slovenia
| | - Tjaša Sernel
- Institute of Dairy Science and Probiotics, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1230 Domžale, Slovenia
| | - Igor Locatelli
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Mitja Kos
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Alenka Šmid
- Faculty of Pharmacy, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Irena Rogelj
- Institute of Dairy Science and Probiotics, Department of Animal Science, Biotechnical Faculty, University of Ljubljana, SI-1230 Domžale, Slovenia
| |
Collapse
|
140
|
Zhang C, Derrien M, Levenez F, Brazeilles R, Ballal SA, Kim J, Degivry MC, Quéré G, Garault P, van Hylckama Vlieg JET, Garrett WS, Doré J, Veiga P. Ecological robustness of the gut microbiota in response to ingestion of transient food-borne microbes. ISME JOURNAL 2016; 10:2235-45. [PMID: 26953599 PMCID: PMC4989305 DOI: 10.1038/ismej.2016.13] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 12/18/2016] [Accepted: 01/08/2016] [Indexed: 01/01/2023]
Abstract
Resident gut microbes co-exist with transient bacteria to form the gut microbiota. Despite increasing evidence suggesting a role for transient microbes on gut microbiota function, the interplay between resident and transient members of this microbial community is poorly defined. We aimed to determine the extent to which a host's autochthonous gut microbiota influences niche permissivity to transient bacteria using a fermented milk product (FMP) as a vehicle for five food-borne bacterial strains. Using conventional and gnotobiotic rats and gut microbiome analyses (16S rRNA genes pyrosequencing and reverse transcription qPCR), we demonstrated that the clearance kinetics of one FMP bacterium, Lactococcus lactis CNCM I-1631, were dependent on the structure of the resident gut microbiota. Susceptibility of the resident gut microbiota to modulation by FMP intervention correlated with increased persistence of L. lactis. We also observed gut microbiome configurations that were associated with altered stability upon exposure to transient bacteria. Our study supports the concept that allochthonous bacteria have transient and subject-specific effects on the gut microbiome that can be leveraged to re-engineer the gut microbiome and improve dysbiosis-related diseases.
Collapse
Affiliation(s)
- Chenhong Zhang
- Metagenopolis, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Muriel Derrien
- Life Science, Danone Nutricia Research, Palaiseau, France
| | - Florence Levenez
- Metagenopolis, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | | | - Sonia A Ballal
- Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jason Kim
- Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | | | - Gaëlle Quéré
- Life Science, Danone Nutricia Research, Palaiseau, France
| | - Peggy Garault
- Life Science, Danone Nutricia Research, Palaiseau, France
| | | | | | - Joël Doré
- Metagenopolis, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Patrick Veiga
- Life Science, Danone Nutricia Research, Palaiseau, France.,Harvard T. H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
141
|
Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de Los Reyes-Gavilán CG, Salazar N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front Microbiol 2016; 7:185. [PMID: 26925050 PMCID: PMC4756104 DOI: 10.3389/fmicb.2016.00185] [Citation(s) in RCA: 1232] [Impact Index Per Article: 154.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/02/2016] [Indexed: 12/18/2022] Open
Abstract
The colon is inhabited by a dense population of microorganisms, the so-called “gut microbiota,” able to ferment carbohydrates and proteins that escape absorption in the small intestine during digestion. This microbiota produces a wide range of metabolites, including short chain fatty acids (SCFA). These compounds are absorbed in the large bowel and are defined as 1-6 carbon volatile fatty acids which can present straight or branched-chain conformation. Their production is influenced by the pattern of food intake and diet-mediated changes in the gut microbiota. SCFA have distinct physiological effects: they contribute to shaping the gut environment, influence the physiology of the colon, they can be used as energy sources by host cells and the intestinal microbiota and they also participate in different host-signaling mechanisms. We summarize the current knowledge about the production of SCFA, including bacterial cross-feedings interactions, and the biological properties of these metabolites with impact on the human health.
Collapse
Affiliation(s)
- David Ríos-Covián
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| | - Patricia Ruas-Madiedo
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| | - Abelardo Margolles
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| | - Miguel Gueimonde
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| | - Clara G de Los Reyes-Gavilán
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| | - Nuria Salazar
- Probiotics and Prebiotics Group, Department of Biochemistry and Microbiology of Dairy Products, Instituto de Productos Lácteos de Asturias, Consejo Superior de Investigaciones Científicas Villaviciosa, Spain
| |
Collapse
|
142
|
Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, de los Reyes-Gavilán CG, Salazar N. Intestinal Short Chain Fatty Acids and their Link with Diet and Human Health. Front Microbiol 2016; 7:185. [PMID: 26925050 PMCID: PMC4756104 DOI: 10.3389/fmicb.2016.00185 10.3389/fmicb.2016.00185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The colon is inhabited by a dense population of microorganisms, the so-called "gut microbiota," able to ferment carbohydrates and proteins that escape absorption in the small intestine during digestion. This microbiota produces a wide range of metabolites, including short chain fatty acids (SCFA). These compounds are absorbed in the large bowel and are defined as 1-6 carbon volatile fatty acids which can present straight or branched-chain conformation. Their production is influenced by the pattern of food intake and diet-mediated changes in the gut microbiota. SCFA have distinct physiological effects: they contribute to shaping the gut environment, influence the physiology of the colon, they can be used as energy sources by host cells and the intestinal microbiota and they also participate in different host-signaling mechanisms. We summarize the current knowledge about the production of SCFA, including bacterial cross-feedings interactions, and the biological properties of these metabolites with impact on the human health.
Collapse
|
143
|
Le Nevé B, Brazeilles R, Derrien M, Tap J, Guyonnet D, Ohman L, Törnblom H, Simrén M. Lactulose Challenge Determines Visceral Sensitivity and Severity of Symptoms in Patients With Irritable Bowel Syndrome. Clin Gastroenterol Hepatol 2016; 14:226-33.e1-3. [PMID: 26492847 DOI: 10.1016/j.cgh.2015.09.039] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/23/2015] [Accepted: 09/29/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Patients with irritable bowel syndrome (IBS) can be assigned to groups with different gastrointestinal (GI) symptoms based on results from a combined nutrient and lactulose challenge. We aimed to identify factors that predict outcomes to this challenge and to determine whether this can be used in noninvasive assessment of visceral sensitivity in patients with IBS. METHODS We performed a prospective study of 100 patients with IBS diagnosed according to Rome III criteria (all subtypes) and seen at a secondary or tertiary care center. After an overnight fast, subjects were given a liquid breakfast (400 mL; Nutridrink) that contained 25 g lactulose. Before the challenge, we assessed visceral sensitivity (via rectal barostat), oro-anal transit time, and fecal microbiota composition (via 16S ribosomal RNA pyrosequencing); we determined IBS severity using questionnaires. The intensity of 8 GI symptoms, the level of digestive comfort, and the amount of exhaled H2 and CH4 in breath were measured before and during a 4-hour period after the liquid breakfast. RESULTS Based on the intensity of 8 GI symptoms and level of digestive comfort during the challenge, patients were assigned to groups with high-intensity GI symptoms (HGS; n = 39) or low-intensity GI symptoms (LGS; n = 61); patients with HGS had more severe IBS (P < .0001), higher somatization (P < .01), and lower quality of life (P < .05-.01) than patients with LGS. Patients with HGS also had significantly higher rectal sensitivity to random phasic distensions (P < .05-.001, compared with patients with LGS). There were no significant differences between groups in fecal microbiota composition, exhaled gas in breath, or oro-anal transit time. CONCLUSIONS We found, in a prospective study, that results from a lactulose challenge test could be used to determine visceral sensitivity and severity of IBS. The intensity of patient symptoms did not correlate with the composition of the fecal microbiota. The lactulose challenge test may help better characterize patients with IBS and evaluate the efficacy of new treatments. ClinicalTrial.gov no: NCT01252550.
Collapse
Affiliation(s)
- Boris Le Nevé
- Danone Nutricia Research, Life Sciences Department, Palaiseau, France.
| | - Rémi Brazeilles
- Danone Nutricia Research, Life Sciences Department, Palaiseau, France
| | - Muriel Derrien
- Danone Nutricia Research, Life Sciences Department, Palaiseau, France
| | - Julien Tap
- Danone Nutricia Research, Life Sciences Department, Palaiseau, France; INRA (Institut National de la Recherche Agronomique) MetaGenoPolis, Jouy en Josas, France
| | - Denis Guyonnet
- Danone Nutricia Research, Life Sciences Department, Palaiseau, France
| | - Lena Ohman
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Hans Törnblom
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Simrén
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
144
|
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal (GI) disorder usually originated from gut dysfunction, and the mechanisms underlying IBS are not clear. IBS can seriously disrupt patient's normal routine, even though it is not life-threatening. With the development of high-throughput sequencing technology, a large number of studies have showed that intestinal flora imbalance does play an important role in the pathogenesis of IBS, especially Bifidobacterium. Bifidobacterium can resist the colonization and invasion of intestinal pathogenic bacteria, and enhance the intestinal epithelial barrier function. Besides, its metabolites also can improve the defense function of the intestinal tract. In the gut of patients with IBS, the number of Bifidobacteria is usually significantly reduced, suggesting that increasing the number of intestinal Bifidobacteria may play a positive role in the treatment of IBS. This paper summarizes the relationship between Bifidobacterium and IBS, and discusses the effect of Bifidobacterium in the adjuvant treatment of IBS.
Collapse
|
145
|
Nocerino R, Paparo L, Terrin G, Pezzella V, Amoroso A, Cosenza L, Cecere G, De Marco G, Micillo M, Albano F, Nugnes R, Ferri P, Ciccarelli G, Giaccio G, Spadaro R, Maddalena Y, Berni Canani F, Berni Canani R. Cow's milk and rice fermented with Lactobacillus paracasei CBA L74 prevent infectious diseases in children: A randomized controlled trial. Clin Nutr 2015; 36:118-125. [PMID: 26732025 DOI: 10.1016/j.clnu.2015.12.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/24/2015] [Accepted: 12/10/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND & AIM Fermented foods have been proposed for the prevention of infectious diseases. We evaluated the efficacy of fermented foods in reducing common infectious diseases (CIDs) in children attending daycare. METHODS Prospective randomized, double-blind, placebo-controlled trial (registered under Clinical Trials.gov identifier NCT01909128) on healthy children (aged 12-48 months) consuming daily cow's milk (group A) or rice (group B) fermented with Lactobacillus paracasei CBA L74, or placebo (group C) for three months during the winter season. The main study outcome was the proportion of children who experienced at least one CID. All CIDs were diagnosed by family pediatricians. Fecal concentrations of innate (α- and β-defensins and cathelicidin LL-37) and acquired immunity biomarkers (secretory IgA) were also evaluated. RESULTS 377 children (193 males, 51%) with a mean (SD) age of 32 (10) months completed the study: 137 in group A, 118 in group B and 122 in group C. Intention-to-treat analysis showed that the proportion of children who experienced at least one CID was lower in group A (51.8%) and B (65.9%) compared to group C (80.3%). Per-protocol analysis showed that the proportion of children presenting upper respiratory tract infections was lower in group A (48.2%) and group B (58.5%) compared with group C (70.5%). The proportion of children presenting acute gastroenteritis was also lower in group A (13.1%) and group B (19.5%) compared with group C (31.1%). A net increase of all fecal biomarkers of innate and acquired immunity was observed for groups A and B compared to group C. Moreover, there was a negative association between fecal biomarkers and the occurrence of CID. CONCLUSION Dietary supplementation with cow's milk or rice fermented with L. paracasei CBA L74 prevents CIDs in children attending daycare possibly by means of a stimulation of innate and acquired immunity.
Collapse
Affiliation(s)
- Rita Nocerino
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Gianluca Terrin
- Department of Gynecology-Obstetrics and Perinatal Medicine, University of Rome "La Sapienza", Rome, Italy
| | - Vincenza Pezzella
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Antonio Amoroso
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Linda Cosenza
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Gaetano Cecere
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Giulio De Marco
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Maria Micillo
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Fabio Albano
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Rosa Nugnes
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Pasqualina Ferri
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Ciccarelli
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Giuliana Giaccio
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Raffaella Spadaro
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | - Ylenia Maddalena
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy
| | | | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples "Federico II", Naples, Italy; European Laboratory for the Investigation of Food Induced Diseases (ELFID), University of Naples "Federico II", Naples, Italy; CEINGE - Advanced Biotechnologies, University of Naples "Federico II", Naples, Italy.
| |
Collapse
|
146
|
Probiotic Bifidobacterium longum alters gut luminal metabolism through modification of the gut microbial community. Sci Rep 2015; 5:13548. [PMID: 26315217 PMCID: PMC4552000 DOI: 10.1038/srep13548] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 06/09/2015] [Indexed: 12/26/2022] Open
Abstract
Probiotics are well known as health-promoting agents that modulate intestinal microbiota. However, the molecular mechanisms underlying this effect remain unclear. Using gnotobiotic mice harboring 15 strains of predominant human gut-derived microbiota (HGM), we investigated the effects of Bifidobacterium longum BB536 (BB536-HGM) supplementation on the gut luminal metabolism. Nuclear magnetic resonance (NMR)-based metabolomics showed significantly increased fecal levels of pimelate, a precursor of biotin, and butyrate in the BB536-HGM group. In addition, the bioassay revealed significantly elevated fecal levels of biotin in the BB536-HGM group. Metatranscriptomic analysis of fecal microbiota followed by an in vitro bioassay indicated that the elevated biotin level was due to an alteration in metabolism related to biotin synthesis by Bacteroides caccae in this mouse model. Furthermore, the proportion of Eubacterium rectale, a butyrate producer, was significantly higher in the BB536-HGM group than in the group without B. longum BB536 supplementation. Our findings help to elucidate the molecular basis underlying the effect of B. longum BB536 on the gut luminal metabolism through its interactions with the microbial community.
Collapse
|
147
|
Le Barz M, Anhê FF, Varin TV, Desjardins Y, Levy E, Roy D, Urdaci MC, Marette A. Probiotics as Complementary Treatment for Metabolic Disorders. Diabetes Metab J 2015; 39:291-303. [PMID: 26301190 PMCID: PMC4543192 DOI: 10.4093/dmj.2015.39.4.291] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Over the past decade, growing evidence has established the gut microbiota as one of the most important determinants of metabolic disorders such as obesity and type 2 diabetes. Indeed, obesogenic diet can drastically alter bacterial populations (i.e., dysbiosis) leading to activation of pro-inflammatory mechanisms and metabolic endotoxemia, therefore promoting insulin resistance and cardiometabolic disorders. To counteract these deleterious effects, probiotic strains have been developed with the aim of reshaping the microbiome to improve gut health. In this review, we focus on benefits of widely used probiotics describing their potential mechanisms of action, especially their ability to decrease metabolic endotoxemia by restoring the disrupted intestinal mucosal barrier. We also discuss the perspective of using new bacterial strains such as butyrate-producing bacteria and the mucolytic Akkermansia muciniphila, as well as the use of prebiotics to enhance the functionality of probiotics. Finally, this review introduces the notion of genetically engineered bacterial strains specifically developed to deliver anti-inflammatory molecules to the gut.
Collapse
Affiliation(s)
- Mélanie Le Barz
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Québec, QC, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
- University of Bordeaux, UMR 5248, CBMN, Bordeaux, France
| | - Fernando F. Anhê
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Québec, QC, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| | - Thibaut V. Varin
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| | - Yves Desjardins
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| | - Emile Levy
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
- Research Centre, Sainte-Justine Hospital, Montreal, QC, Canada
- Department of Nutrition, University of Montreal Faculty of Medicine, Montreal, QC, Canada
| | - Denis Roy
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| | | | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Québec, QC, Canada
- Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada
| |
Collapse
|
148
|
Hisada T, Endoh K, Kuriki K. Inter- and intra-individual variations in seasonal and daily stabilities of the human gut microbiota in Japanese. Arch Microbiol 2015; 197:919-34. [PMID: 26068535 PMCID: PMC4536265 DOI: 10.1007/s00203-015-1125-0] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 05/23/2015] [Accepted: 05/26/2015] [Indexed: 12/23/2022]
Abstract
Relationships between human gut microbiota, dietary habits, and health/diseases are the subject of epidemiological and clinical studies. However, the temporal stability and variability of the bacterial community in fecal samples remain unclear. In this study, middle-aged Japanese male and female volunteers (n = 5 each) without disease were recruited from the Sakura Diet Study. Fecal samples and lifestyle information were collected in every quarter and at each defecation for 7 continuous days. Next-generation sequencing of 16S rDNA and hierarchical clustering showed no time trend and intra-individual differences in both fecal sample sets. Significant inter-individual variations in seasonal and daily fecal sample sets were detected for 24 and 23 out of 39 selected dominant genera (>0.1% of the total human gut microbiota; occupation rate >85%), respectively. Intra- to inter-individual variance ratios in 26 and 35 genera were significantly <1.0 for seasonal and daily stabilities. Seasonal variation in fermented milk consumption might be associated with Bifidobacterium composition, but not with Lactobacillus. For most of the dominant genera in the human gut microbiota, inter-individual variations were significantly larger than intra-individual variations. Further studies are warranted to determine the contributions of human gut microbiota to nutritional metabolism, health promotion, and prevention/development of diseases.
Collapse
Affiliation(s)
- Takayoshi Hisada
- Laboratory of Public Health, Graduate School of Nutritional and Environmental Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | | | | |
Collapse
|
149
|
Host lysozyme-mediated lysis of Lactococcus lactis facilitates delivery of colitis-attenuating superoxide dismutase to inflamed colons. Proc Natl Acad Sci U S A 2015; 112:7803-8. [PMID: 26056274 DOI: 10.1073/pnas.1501897112] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Beneficial microbes that target molecules and pathways, such as oxidative stress, which can negatively affect both host and microbiota, may hold promise as an inflammatory bowel disease therapy. Prior work showed that a five-strain fermented milk product (FMP) improved colitis in T-bet(-/-) Rag2(-/-) mice. By varying the number of strains used in the FMP, we found that Lactococcus lactis I-1631 was sufficient to ameliorate colitis. Using comparative genomic analyses, we identified genes unique to L. lactis I-1631 involved in oxygen respiration. Respiration of oxygen results in reactive oxygen species (ROS) generation. Also, ROS are produced at high levels during intestinal inflammation and cause tissue damage. L. lactis I-1631 possesses genes encoding enzymes that detoxify ROS, such as superoxide dismutase (SodA). Thus, we hypothesized that lactococcal SodA played a role in attenuating colitis. Inactivation of the sodA gene abolished L. lactis I-1631's beneficial effect in the T-bet(-/-) Rag2(-/-) model. Similar effects were obtained in two additional colonic inflammation models, Il10(-/-) mice and dextran sulfate sodium-treated mice. Efforts to understand how a lipophobic superoxide anion (O2 (-)) can be detoxified by cytoplasmic lactoccocal SodA led to the finding that host antimicrobial-mediated lysis is a prerequisite for SodA release and SodA's extracytoplasmic O2 (-) scavenging. L. lactis I-1631 may represent a promising vehicle to deliver antioxidant, colitis-attenuating SodA to the inflamed intestinal mucosa, and host antimicrobials may play a critical role in mediating SodA's bioaccessibility.
Collapse
|
150
|
Derrien M, van Hylckama Vlieg JE. Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol 2015; 23:354-66. [DOI: 10.1016/j.tim.2015.03.002] [Citation(s) in RCA: 282] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/28/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023]
|