51
|
Dao MC, Sokolovska N, Brazeilles R, Affeldt S, Pelloux V, Prifti E, Chilloux J, Verger EO, Kayser BD, Aron-Wisnewsky J, Ichou F, Pujos-Guillot E, Hoyles L, Juste C, Doré J, Dumas ME, Rizkalla SW, Holmes BA, Zucker JD, Clément K. A Data Integration Multi-Omics Approach to Study Calorie Restriction-Induced Changes in Insulin Sensitivity. Front Physiol 2019; 9:1958. [PMID: 30804813 PMCID: PMC6371001 DOI: 10.3389/fphys.2018.01958] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/27/2018] [Indexed: 12/17/2022] Open
Abstract
Background: The mechanisms responsible for calorie restriction (CR)-induced improvement in insulin sensitivity (IS) have not been fully elucidated. Greater insight can be achieved through deep biological phenotyping of subjects undergoing CR, and integration of big data. Materials and Methods: An integrative approach was applied to investigate associations between change in IS and factors from host, microbiota, and lifestyle after a 6-week CR period in 27 overweight or obese adults (ClinicalTrials.gov: NCT01314690). Partial least squares regression was used to determine associations of change (week 6 - baseline) between IS markers and lifestyle factors (diet and physical activity), subcutaneous adipose tissue (sAT) gene expression, metabolomics of serum, urine and feces, and gut microbiota composition. ScaleNet, a network learning approach based on spectral consensus strategy (SCS, developed by us) was used for reconstruction of biological networks. Results: A spectrum of variables from lifestyle factors (10 nutrients), gut microbiota (10 metagenomics species), and host multi-omics (metabolic features: 84 from serum, 73 from urine, and 131 from feces; and 257 sAT gene probes) most associated with IS were identified. Biological network reconstruction using SCS, highlighted links between changes in IS, serum branched chain amino acids, sAT genes involved in endoplasmic reticulum stress and ubiquitination, and gut metagenomic species (MGS). Linear regression analysis to model how changes of select variables over the CR period contribute to changes in IS, showed greatest contributions from gut MGS and fiber intake. Conclusion: This work has enhanced previous knowledge on links between host glucose homeostasis, lifestyle factors and the gut microbiota, and has identified potential biomarkers that may be used in future studies to predict and improve individual response to weight-loss interventions. Furthermore, this is the first study showing integration of the wide range of data presented herein, identifying 115 variables of interest with respect to IS from the initial input, consisting of 9,986 variables. Clinical Trial Registration: clinicaltrials.gov (NCT01314690).
Collapse
Affiliation(s)
- Maria Carlota Dao
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Nataliya Sokolovska
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | | | - Séverine Affeldt
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Véronique Pelloux
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Edi Prifti
- Institute of Cardiometabolism and Nutrition, Integromics, ICAN, Paris, France
- Sorbonne University, IRD, UMMISCO, Bondy, France
| | - Julien Chilloux
- Section of Biomolecular Medicine, Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Eric O. Verger
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Brandon D. Kayser
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Judith Aron-Wisnewsky
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| | - Farid Ichou
- Institute of Cardiometabolism and Nutrition, ICANalytics, Paris, France
| | - Estelle Pujos-Guillot
- Institut National de la Recherche Agronomique, Unité de Nutrition Humaine, Plateforme d’Exploration du Métabolisme, MetaboHUB, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Lesley Hoyles
- Section of Biomolecular Medicine, Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Clifton Campus, Nottingham, United Kingdom
| | - Catherine Juste
- National Institute of Agricultural Research, Micalis Institute, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Joël Doré
- National Institute of Agricultural Research, Micalis Institute, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Marc-Emmanuel Dumas
- Section of Biomolecular Medicine, Division of Integrative Systems Medicine and Digestive Disease, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Salwa W. Rizkalla
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
| | | | - Jean-Daniel Zucker
- Institute of Cardiometabolism and Nutrition, Integromics, ICAN, Paris, France
- Sorbonne University, IRD, UMMISCO, Bondy, France
| | - Karine Clément
- Sorbonne University, French National Institute for Health and Medical Research, NutriOmics Unit, Institute of Cardiometabolism and Nutrition, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition Department, CRNH Ile-de-France, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
52
|
Vinberg M, Ottesen NM, Meluken I, Sørensen N, Pedersen O, Kessing LV, Miskowiak KW. Remitted affective disorders and high familial risk of affective disorders associate with aberrant intestinal microbiota. Acta Psychiatr Scand 2019; 139:174-184. [PMID: 30374951 DOI: 10.1111/acps.12976] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/08/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Affective disorders seem associated with aberrant intestinal microbiota but whether this pattern also occurs in individuals at increased heritable risk is unknown. We investigated associations between gut microbiota profiles and affective disorders by comparing monozygotic (MZ) twins concordant (affected twins with unipolar or bipolar disorder in remission) and discordant to affective disorders (high-risk) with MZ twins without affective disorders (low-risk). METHODS Stool samples were collected from 128 MZ twins and the microbiome was profiled using 16S rDNA sequencing of the V3-V4 region. RESULTS Affected twins had a lower diversity and an absence of a specific operational taxonomical unit (OTU) in comparison with low-risk twins. The high-risk twins exhibited the same pattern although the lower diversity was only at a trend level. The OTU belonged to the family Christensenellaceae. The findings were not explained by lifestyle factors (smoking, alcohol consumption, body mass index, or psychotropic medication). CONCLUSION Affected twins in remission and high-risk twins presented aberrant gut microbiota with depletion of a specific OTU. If replicated, this reduced relative sequence absence may together with the globally altered microbiota composition act as a vulnerability marker by accentuating the effect of gene-environment interactions in individuals genetically disposed for an affective disorder.
Collapse
Affiliation(s)
- M Vinberg
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen Affective Disorder Research Center (CADIC), University Hospital of Copenhagen, Copenhagen, Denmark
| | - N M Ottesen
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen Affective Disorder Research Center (CADIC), University Hospital of Copenhagen, Copenhagen, Denmark
| | - I Meluken
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen Affective Disorder Research Center (CADIC), University Hospital of Copenhagen, Copenhagen, Denmark
| | - N Sørensen
- Clinical Microbiomics, Copenhagen, Denmark
| | - O Pedersen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, University of Copenhagen, Copenhagen, Denmark
| | - L V Kessing
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen Affective Disorder Research Center (CADIC), University Hospital of Copenhagen, Copenhagen, Denmark
| | - K W Miskowiak
- Psychiatric Centre Copenhagen, Rigshospitalet, Copenhagen Affective Disorder Research Center (CADIC), University Hospital of Copenhagen, Copenhagen, Denmark.,Institute of Psychology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
53
|
Zacarías MF, Collado MC, Gómez-Gallego C, Flinck H, Aittoniemi J, Isolauri E, Salminen S. Pregestational overweight and obesity are associated with differences in gut microbiota composition and systemic inflammation in the third trimester. PLoS One 2018; 13:e0200305. [PMID: 30005082 PMCID: PMC6044541 DOI: 10.1371/journal.pone.0200305] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022] Open
Abstract
The obesity epidemic is a global challenge, and the velocity of propagation is high in the population at reproductive age. Overweight and obesity during pregnancy have been associated with high birth weight and an increased risk of childhood obesity, reinforcing the risk of other non-communicable diseases. Obesity involves chronic low-grade systemic inflammation. New biomarkers for early detection of obesity risk are urgently required. The aim of this study was to identify the connection between pregestational BMI (pre-BMI) status and inflammatory biomarkers during the third trimester of pregnancy and their association with intestinal microbiota composition. Fifty-four pregnant women were classified according to pre-pregnancy BMI as normoweight, overweight, or obese. Weight gain, inflammatory biomarkers (hs_CRP, haptoglobin, and suPAR), and microbiota composition were assessed during the third trimester. A significant lower weight gain for obese mothers and a positive correlation between pre-BMI and inflammatory biomarkers were detected (Spearman’s correlation). Haptoglobin levels were significantly higher in overweight and obese mothers. Higher Firmicutes levels and a higher ratio Firmicutes/Bacteroidetes were observed in the overweight and obese subjects. High hs_CRP and haptoglobin levels were also correlated with decreased microbiota diversity (Shannon index), whereas haptoglobin and hs_CRP values were correlated with several microbiota components, such as Ruminococcus gnavus and Faecalibacterium, and with specific phyla in the normoweight and overweight mothers; no significant associations with microbiota were found for suPAR. In conclusion, haptoglobin and hs_CRP reflected pregestational BMI status and related microbiota components, but haptoglobin was a better biomarker for microbiota associated with overweight. suPAR was associated with low grade inflammation dependent on pre-pregnancy BMI, but it was not related to deviated microbiota profiles.
Collapse
Affiliation(s)
| | - María Carmen Collado
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Department of Biotechnology, Valencia, Spain
| | - Carlos Gómez-Gallego
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| | - Heini Flinck
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - Janne Aittoniemi
- Department of Clinical Microbiology, Fimlab Laboratories, Tampere, Finland
| | - Erika Isolauri
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital, Turku, Finland
- Department of Clinical Sciences, Faculty of Medicine, University of Turku, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, Faculty of Medicine, University of Turku, Turku, Finland
| |
Collapse
|
54
|
Dalvi PS, Yang S, Swain N, Kim J, Saha S, Bourdon C, Zhang L, Chami R, Bandsma RHJ. Long-term metabolic effects of malnutrition: Liver steatosis and insulin resistance following early-life protein restriction. PLoS One 2018; 13:e0199916. [PMID: 29965973 PMCID: PMC6028108 DOI: 10.1371/journal.pone.0199916] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Early postnatal-life malnutrition remains prevalent globally, and about 45% of all child deaths are linked to malnutrition. It is not clear whether survivors of childhood malnutrition suffer from long-term metabolic effects, especially when they are later in life exposed to a fat and carbohydrate rich obesogenic diet. The lack of knowledge around this dietary “double burden” warrants studies to understand the long-term consequences of children previously exposed to malnutrition. We hypothesized that an early-life nutritional insult of low protein consumption in mice would lead to long-term metabolic disturbances that would exacerbate the development of diet-induced insulin resistance and non-alcoholic fatty liver disease (NAFLD). We investigated the effects of feeding a low protein diet (4% wt/wt) immediately after weaning for four weeks and subsequent feeding of a high carbohydrate high fat feeding for 16 weeks on metabolic function and development of NAFLD. Mice exposed to early-life protein restriction demonstrated a transient glucose intolerance upon recovery by regular chow diet feeding. However, protein restriction after weaning in mice did not exacerbate an obesogenic diet-induced insulin resistance or progression to NAFLD. These data suggest that transient protein restriction in early-life does not exacerbate an obesogenic diet-induced NAFLD and insulin resistance.
Collapse
Affiliation(s)
- Prasad S. Dalvi
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Morosky College of Health Professions and Sciences, Gannon University, Erie, PA, United States of America
| | - Steven Yang
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Nathan Swain
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Junsoo Kim
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Senjuti Saha
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Celine Bourdon
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Ling Zhang
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rose Chami
- Department of Laboratory Medicine and Pathology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, ON, Canada
| | - Robert H. J. Bandsma
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- * E-mail:
| |
Collapse
|
55
|
Bubnov RV, Babenko LP, Lazarenko LM, Mokrozub VV, Spivak MY. Specific properties of probiotic strains: relevance and benefits for the host. EPMA J 2018; 9:205-223. [PMID: 29896319 PMCID: PMC5972142 DOI: 10.1007/s13167-018-0132-z] [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] [Received: 02/13/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Probiotics have tremendous potential to develop healthy diets, treatment, and prevention. Investigation of in vitro cultural properties of health-promoting microorganisms like lactic acid bacteria (LAB) and bifidobacteria is crucial to select probiotic strains for treatments based on gut microbiota modulation to justify individualized and personalized approach for nutrition and prevention of variety of diseases. The aim was to study the biological properties of LAB and bifidobacteria probiotic strains, namely adhesive properties; resistance to antibiotics; and biological fluids (gastric juice, bile, pancreatic enzymes), and to overview the literature in the field. MATERIALS AND METHODS We studied six LAB strains (Lactobacillus acidophilus ІМV В-7279, L. casei ІМV В-7280, L. delbrueckii subsp. bulgaricus ІМV В-7281, L. rhamnosus LB-3 VK6, L. delbrueckii LE VK8, L. plantarum LM VK7), and two bifidobacteria strains (Bifidobacterium animalis VKL, B. animalis VKB). We characterized tinctorial, culturally morphological, physiological, and biochemical properties of probiotic strains of LAB and bifidobacteria by commonly used research methods. Determination of the resistance to antibiotics was carried out using disc-diffusion method. The effects of gastric juice, bile, and pancreatin on the viability of LAB and bifidobacteria were evaluated. Adhesive properties of LAB and bifidobacteria to epithelial cells were assessed calculating three indicators: average adhesion rate (AAR), participation rate of epithelial cells (PRE), and adhesiveness index of microorganisms (AIM). Electron microscopy of LAB and bifidobacteria cells was conducted. RESULTS The studied strains of LAB and bifidobacteria did not form spores, were positively stained by Gram, grow on medium in a wide range of pH (1.0-9.0, optimum pH 5.5-6.5), were sensitive to a wide range of antibiotics; and showed different resistance to gastric juice, bile, and pancreatic enzymes. The most resistant to antibiotics were L. rhamnosus LB-3 VK6 and L. delbrueckii LE VK8 strains. The most susceptible to gastric juice was L. plantarum LM VK7, which stopped its growth at 8% of gastric juice; L. acidophilus IMV B-7279, B. animalis VKL, and B. animalis VKB strains were resistant even in the 100% concentration. Strains L. acidophilus IMV В-7279, L. casei IMV В-7280, B. animalis VKL, B. animalis VKB, L. rhamnosus LB-3 VK6, L. delbrueckii LE VK8, and L. delbrueckii subsp. bulgaricus IMV В-7281 were resistant to pancreatic enzymes. Adhesive properties of the strains according to AIM index were high in L. casei IMV В-7280, B. animalis VKL, and B. animalis VKB; were moderate in L. delbrueckii subsp. bulgaricus IMV В-7281; and were low in L. acidophilus IMV В-7279, L. rhamnosus LB-3 VK6, L. delbrueckii LE VK8, and L. plantarum LM VK7. CONCLUSION We recognized strain-dependent properties of studied LAB and bifidobacteria probiotic strains (adhesive ability, resistance to antibiotics, and gut biological fluids) and discussed potential for most effective individualized treatment for gut and distant sites microbiome modulation.
Collapse
Affiliation(s)
- Rostyslav V. Bubnov
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Zabolotny str., 154, Kyiv, 03143 Ukraine
- Clinical Hospital ‘Pheophania’ of State Affairs Department, Zabolotny str., 21, Kyiv, 03143 Ukraine
| | - Lidiia P. Babenko
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Zabolotny str., 154, Kyiv, 03143 Ukraine
| | - Liudmyla M. Lazarenko
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Zabolotny str., 154, Kyiv, 03143 Ukraine
| | - Victoria V. Mokrozub
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Zabolotny str., 154, Kyiv, 03143 Ukraine
| | - Mykola Ya. Spivak
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Zabolotny str., 154, Kyiv, 03143 Ukraine
- PJSC «SPC Diaproph-Med», Svitlycky Str., 35, Kyiv, 04123 Ukraine
| |
Collapse
|
56
|
Abstract
Due to declining estrogen levels during menopause, NAFLD prevalence is higher in postmenopausal women compared to in premenopausal women or in men. Postmenopausal women are more susceptible to weight gain, fat redistribution and dyslipidemia, all major hallmarks of metabolic syndrome associated with increased NAFLD risk. Gut microbiota plays important roles in development of gastrointestinal tract, metabolism and immunity. Host-microbe interactions allows regulation of a wide range of pathways that affect healthy and diseased physiology. Recent advances in - omics technologies, such as microbiome, transcriptome and metabolome analysis, provided evidence that estrogens and intestinal microbiota (IM) can collectively influence obesity, inflammatory disease, diabetes, and cancers. By understanding underlying mechanisms of estrogens and microbiota crosstalk, we might design dietary and pharmacological interventions to alleviate the metabolic syndrome and NAFLD.
Collapse
Affiliation(s)
- Karen L Chen
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Zeynep Madak-Erdogan
- Division of Nutritional Sciences, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA; Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA.
| |
Collapse
|
57
|
Healey GR, Murphy R, Brough L, Butts CA, Coad J. Interindividual variability in gut microbiota and host response to dietary interventions. Nutr Rev 2018; 75:1059-1080. [PMID: 29190368 DOI: 10.1093/nutrit/nux062] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dysbiosis is linked to human disease; therefore, gut microbiota modulation strategies provide an attractive means of correcting microbial imbalance to enhance human health. Because diet has a major influence on the composition, diversity, and metabolic capacity of the gut microbiota, numerous dietary intervention studies have been conducted to manipulate the gut microbiota to improve host outcomes and reduce disease risk. Emerging evidence suggests that interindividual variability in gut microbiota and host responsiveness exists, making it difficult to predict gut microbiota and host response to a given dietary intervention. This may, in turn, have implications on the consistency of results among studies and the perceived success or true efficacy of a dietary intervention in eliciting beneficial changes to the gut microbiota and human health.
Collapse
Affiliation(s)
- Genelle R Healey
- Massey Institute of Food Science and Technology, School of Food and Nutrition, Massey University, Palmerston North, New Zealand
- Food, Nutrition & Health Group, New Zealand Institute for Plant & Food Research Limited, Palmerston North, New Zealand
| | - Rinki Murphy
- Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Louise Brough
- Massey Institute of Food Science and Technology, School of Food and Nutrition, Massey University, Palmerston North, New Zealand
| | - Christine A Butts
- Food, Nutrition & Health Group, New Zealand Institute for Plant & Food Research Limited, Palmerston North, New Zealand
| | - Jane Coad
- Massey Institute of Food Science and Technology, School of Food and Nutrition, Massey University, Palmerston North, New Zealand
| |
Collapse
|
58
|
Dietary Pattern and Macronutrients Profile on the Variation of Inflammatory Biomarkers: Scientific Update. Cardiol Res Pract 2018; 2018:4762575. [PMID: 29725543 PMCID: PMC5872610 DOI: 10.1155/2018/4762575] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/30/2017] [Indexed: 01/18/2023] Open
Abstract
It is known that the dietary pattern and macronutrients profile may influence the expression and secretion of inflammatory biomarkers, and the low-grade inflammation is associated with the manifestation of noncommunicable chronic diseases. Therefore, this review aimed to present and discuss the role of dietary patterns and macronutrients on the variation of inflammatory markers related to NCD risk. Scientific evidences within the last five years based on clinical trials, case-controls, cohorts, and cross-sectional studies indicate that normocaloric, carbohydrate-moderated, low-glycemic index, protein-moderated, monounsaturated and polyunsaturated fatty acid-rich, omega-3, and low-saturated fat diets display positive effects on the inflammatory state, both in healthy individuals and in those with cardiovascular risk, although the second group seems to benefit more from changes in the dietary profile.
Collapse
|
59
|
Fangmann D, Theismann EM, Türk K, Schulte DM, Relling I, Hartmann K, Keppler JK, Knipp JR, Rehman A, Heinsen FA, Franke A, Lenk L, Freitag-Wolf S, Appel E, Gorb S, Brenner C, Seegert D, Waetzig GH, Rosenstiel P, Schreiber S, Schwarz K, Laudes M. Targeted Microbiome Intervention by Microencapsulated Delayed-Release Niacin Beneficially Affects Insulin Sensitivity in Humans. Diabetes Care 2018; 41:398-405. [PMID: 29212824 DOI: 10.2337/dc17-1967] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/08/2017] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Gut microbiota represent a potential novel target for future prediabetes and type 2 diabetes therapies. In that respect, niacin has been shown to beneficially affect the host-microbiome interaction in rodent models. RESEARCH DESIGN AND METHODS We characterized more than 500 human subjects with different metabolic phenotypes regarding their niacin (nicotinic acid [NA] and nicotinamide [NAM]) status and their gut microbiome. In addition, NA and NAM delayed-release microcapsules were engineered and examined in vitro and in vivo in two human intervention studies (bioavailability study and proof-of-concept/safety study). RESULTS We found a reduced α-diversity and Bacteroidetes abundance in the microbiome of obese human subjects associated with a low dietary niacin intake. We therefore developed delayed-release microcapsules targeting the ileocolonic region to deliver increasing amounts of NA and NAM to the microbiome while preventing systemic resorption to avoid negative side effects (e.g., facial flushing). In vitro studies on these delayed-release microcapsules revealed stable conditions at pH 1.4, 4.5, and 6.8, followed by release of the compounds at pH 7.4, simulating the ileocolonic region. In humans in vivo, gut-targeted delayed-release NA but not NAM produced a significant increase in the abundance of Bacteroidetes. In the absence of systemic side effects, these favorable microbiome changes induced by microencapsulated delayed-release NA were associated with an improvement of biomarkers for systemic insulin sensitivity and metabolic inflammation. CONCLUSION Targeted microbiome intervention by delayed-release NA might represent a future therapeutic option for prediabetes and type 2 diabetes.
Collapse
Affiliation(s)
- Daniela Fangmann
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | | | - Kathrin Türk
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Dominik M Schulte
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Isabelle Relling
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | | | - Julia K Keppler
- Department of Food Technology, University of Kiel, Kiel, Germany
| | | | - Ateequr Rehman
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | | | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Lennart Lenk
- Institute for Experimental Cancer Research, University of Kiel, Kiel, Germany
| | - Sandra Freitag-Wolf
- Institute of Medical Informatics and Statistics, University of Kiel, Kiel, Germany
| | - Esther Appel
- Zoological Institute, University of Kiel, Kiel, Germany
| | | | - Charles Brenner
- Department of Biochemistry, University of Iowa, Iowa City, IA
| | | | | | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Stefan Schreiber
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany .,Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Karin Schwarz
- Department of Food Technology, University of Kiel, Kiel, Germany
| | - Matthias Laudes
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| |
Collapse
|
60
|
Dao MC, Clément K. Gut microbiota and obesity: Concepts relevant to clinical care. Eur J Intern Med 2018; 48:18-24. [PMID: 29110901 DOI: 10.1016/j.ejim.2017.10.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/04/2017] [Indexed: 02/07/2023]
Abstract
The composition and function of gut microbiota play a role in obesity and metabolic disease, yet the mechanisms have not been fully described. As new discoveries and advances in the field have occurred, the relevance of gut microbiota in clinical care has become more substantial. There is promising potential for manipulation of the gut microbiota as treatment of obesity and associated health complications, both as a standalone therapy and as part of interventions such as weight loss. In this review we have compiled knowledge and concepts that are important in the consideration of gut microbiota for clinical care.
Collapse
Affiliation(s)
- Maria Carlota Dao
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France; INSERM, UMR S U1166, Nutriomics Team, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France.
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France; INSERM, UMR S U1166, Nutriomics Team, Paris, France; Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France.
| |
Collapse
|
61
|
Role of microbiota-derived lipopolysaccharide in adipose tissue inflammation, adipocyte size and pyroptosis during obesity. Nutr Res Rev 2018; 31:153-163. [DOI: 10.1017/s0954422417000269] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractIt has been established that ingestion of a high-fat diet increases the blood levels of lipopolysaccharides (LPS) from Gram-negative bacteria in the gut. Obesity is characterised by low-grade systemic and adipose tissue inflammation. This is suggested to be implicated in the metabolic syndrome and obesity. In the present review, we hypothesise that LPS directly and indirectly participates in the inflammatory reaction in adipose tissue during obesity. The experimental evidence shows that LPS is involved in the transition of macrophages from the M2 to the M1 phenotype. In addition, LPS inside adipocytes may activate caspase-4/5/11. This may induce a highly inflammatory type of programmed cell death (i.e. pyroptosis), which also occurs after infection with intracellular pathogens. Lipoproteins with or without LPS are taken up by adipocytes. Large adipocytes are more metabolically active and potentially more exposed to LPS than small adipocytes are. Thus, LPS might be involved in defining the adipocyte death size and the formation of crown-like structures. The adipocyte death size is reached when the intracellular concentration of LPS initiates pyroptosis. The mechanistic details remain to be elucidated, but the observations indicate that adipocytes are stimulated to cell death by processes that involve LPS from the gut microbiota. There is a complex interplay between the composition of the diet and microbiota. This influences the amount of LPS that is translocated from the gut. In particular, the lipid content of a meal may correlate with the amount of LPS built in to chylomicrons.
Collapse
|
62
|
Moran-Ramos S, López-Contreras BE, Canizales-Quinteros S. Gut Microbiota in Obesity and Metabolic Abnormalities: A Matter of Composition or Functionality? Arch Med Res 2017; 48:735-753. [PMID: 29292011 DOI: 10.1016/j.arcmed.2017.11.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/15/2017] [Indexed: 12/18/2022]
Abstract
The obesity pandemic and the metabolic complications derived from it represent a major public health challenge worldwide. Although obesity is a multifactorial disease, research from the past decade suggests that the gut microbiota interacts with host genetics and diet, as well as with other environmental factors, and thus contributes to the development of obesity and related complications. Despite abundant research on animal models, substantial evidence from humans has only started to accumulate over the past few years. Thus, the aim of the present review is to discuss structural and functional characteristics of the gut microbiome in human obesity, challenges associated with multi-omic technologies, and advances in identifying microbial metabolites with a direct link to obesity and metabolic complications. To date, studies suggests that obesity is related to low microbial diversity and taxon depletion sometimes resulting from an interaction with host dietary habits and genotype. These findings support the idea that the depletion or absence of certain taxa leaves an empty niche, likely leading to compromised functionality and thus promoting dysbiosis. Although the role of altered gut microbiota as cause or consequence of obesity remains controversial, research on microbial genomes and metabolites points towards an increased extraction of energy from the diet in obesity and suggests that metabolites, such as trimethylamine-N-oxide or branched-chain amino acids, participate in metabolic complications. Future research should be focused on structural and functional levels to unravel the mechanism linking gut microbiota and obesity.
Collapse
Affiliation(s)
- Sofia Moran-Ramos
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Ciudad de México, México; Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica, Ciudad de México, México.
| | - Blanca E López-Contreras
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica, Ciudad de México, México
| | - Samuel Canizales-Quinteros
- Unidad de Genómica de Poblaciones Aplicada a la Salud, Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica, Ciudad de México, México.
| |
Collapse
|
63
|
Amaral WZ, Lubach GR, Proctor A, Lyte M, Phillips GJ, Coe CL. Social Influences on Prevotella and the Gut Microbiome of Young Monkeys. Psychosom Med 2017; 79:888-897. [PMID: 28178033 PMCID: PMC5547018 DOI: 10.1097/psy.0000000000000454] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Our aim was to evaluate the bacterial profiles of young monkeys as they were weaned into peer groups with a particular focus on Prevotella, an important taxon in both human and nonhuman primates. The weaning of infants and increased social contact with peers is a developmental stage that is likely to affect the gut microbiome. METHODS Gut bacteria were assessed in 63 rhesus monkeys living in social groups comprised of 4 to 7 individuals. Two groups were assessed prospectively on day 1 and 2 weeks after rehousing away from the mother and group formation. Ten additional groups were assessed at 2 weeks after group establishment. Fecal genomic DNA was extracted and 16S ribosomal RNA sequenced by Illumina MiSeq (5 social groups) and 454-amplicon pyrosequencing (7 social groups). RESULTS Combining weaned infants into small social groups led to a microbial convergence by 2 weeks (p < .001). Diversity analyses indicated more similar community structure within peer groups than across groups (p < .01). Prevotella was the predominant taxon, and its abundance differed markedly across individuals. Indices of richness, microbial profiles, and less abundant taxa were all associated with the Prevotella levels. Functional Kyoto Encyclopedia of Genes and Genomes analyses suggested corresponding shifts in metabolic pathways. CONCLUSIONS The formation of small groups of young rhesus monkeys was associated with significant shifts in the gut microbiota. The profiles were closely associated with the abundance of Prevotella, a predominant taxon in the rhesus monkey gut. Changes in the structure of the gut microbiome are likely to induce differences in metabolic and physiologic functioning.
Collapse
Affiliation(s)
- Wellington Z Amaral
- From the Harlow Center for Biological Psychology (Amaral, Lubach, Coe), University of Wisconsin, Madison, Wisconsin; and the Department of Veterinary Microbiology and Veterinary Medicine (Proctor, Lyte, Phillips), Iowa State University, Ames, Iowa
| | | | | | | | | | | |
Collapse
|
64
|
Dietary intake of fat and fibre according to reference values relates to higher gut microbiota richness in overweight pregnant women. Br J Nutr 2017; 118:343-352. [PMID: 28901891 DOI: 10.1017/s0007114517002100] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The diet-microbiota-metabolism relationships during pregnancy are mostly unknown. We explored the effect of the habitual diet and adherence to the dietary reference values on gut microbiota composition and diversity. Further, the association of gut microbiota with serum lipidomics and low-grade inflammation was evaluated. Overweight and obese women (BMI 30·7 (sd 4·4) kg/m2, n 100) were studied at early pregnancy (≤17 weeks). Intakes of nutrients were calculated from 3-d food diaries. Faecal microbiota composition was analysed using 16S rRNA gene sequencing. Fasting serum lipidomic profiles were determined by NMR. High-sensitivity C-reactive protein, glycoprotein acetylation (GlycA) and lipopolysaccharide activity were used as markers for low-grade inflammation. The recommended dietary intake of fibre and fat was related to higher gut microbiota richness and lower abundance of Bacteroidaceae. Correlations were observed between gut microbiota richness and GlycA and between a few microbiota genera and serum lipoprotein particles. As a conclusion, adherence to the dietary reference intake of fat and fibre was associated with beneficial gut microbiota composition, which again contributed to lipidomic profile. Higher gut microbiota richness and nutrient intakes were linked to a lower level of low-grade inflammation marker GlycA. This finding offers novel insights and opportunities for dietary modification during pregnancy with potential of improving the health of the mother and the child.
Collapse
|
65
|
Deehan EC, Duar RM, Armet AM, Perez-Muñoz ME, Jin M, Walter J. Modulation of the Gastrointestinal Microbiome with Nondigestible Fermentable Carbohydrates To Improve Human Health. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0019-2017. [PMID: 28936943 PMCID: PMC11687544 DOI: 10.1128/microbiolspec.bad-0019-2017] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Indexed: 02/07/2023] Open
Abstract
There is a clear association between the gastrointestinal (GI) microbiome and the development of chronic noncommunicable diseases, providing a rationale for the development of strategies that target the GI microbiota to improve human health. In this article, we discuss the potential of supplementing the human diet with nondigestible fermentable carbohydrates (NDFCs) to modulate the composition, structure, diversity, and metabolic potential of the GI microbiome in an attempt to prevent or treat human disease. The current concepts by which NDFCs can be administered to humans, including prebiotics, fermentable dietary fibers, and microbiota-accessible carbohydrates, as well as the mechanisms by which these carbohydrates exert their health benefits, are discussed. Epidemiological research presents compelling evidence for the health effects of NDFCs, with clinical studies providing further support for some of these benefits. However, rigorously designed human intervention studies with well-established clinical markers and microbial endpoints are still essential to establish (i) the clinical efficiency of specific NDFCs, (ii) the causal role of the GI microbiota in these effects, (iii) the underlying mechanisms involved, and (iv) the degree by which inter-individual differences between GI microbiomes influence these effects. Such studies would provide the mechanistic understanding needed for a systematic application of NDFCs to improve human health via GI microbiota modulation while also allowing the personalization of these dietary strategies.
Collapse
Affiliation(s)
- Edward C Deehan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Rebbeca M Duar
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Anissa M Armet
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Maria Elisa Perez-Muñoz
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| | - Mingliang Jin
- Department of Microbiology and Immunology, Northwestern Polytechnical University, Xi'an, Shaanxi, China 710065
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada T6G 2E1
| |
Collapse
|
66
|
MIRMIRAN P, AMIRHAMIDI Z, EJTAHED HS, BAHADORAN Z, AZIZI F. Relationship between Diet and Non-alcoholic Fatty Liver Disease: A Review Article. IRANIAN JOURNAL OF PUBLIC HEALTH 2017; 46:1007-1017. [PMID: 28894701 PMCID: PMC5575379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
BACKGROUND Diet plays a key role in the development of non-alcoholic fatty liver disease (NAFLD). The aim of this study was to review systematically observational studies available regarding the relationship between food intakes and NAFLD. METHODS We searched Scopus, PubMed, and Cochrane Library databases to identify English observational studies on food groups, dietary patterns, and NAFLD. Cross-sectional, case-control and cohort studies were selected and then duplication, topic, type of study, study population, variables examined and quality of data reporting of the articles were evaluated. RESULTS We identified 2128 studies in the initial search, of which 33 were reviewed in full text and 7 articles were included in this systematic review. Intakes of red meat, fats, and sweets were high whereas consumption of whole grains, fruits and vegetables were less in NAFLD patients. Moreover, there was a positive association between the Western dietary pattern and the risk of NAFLD, while adherence to the Mediterranean diet was significantly associated with the severity of hepatic steatosis. CONCLUSION Generally, different food group intakes and dietary patterns are associated with the progression of NAFLD and its risk factors. Because of the many limitations of available studies reviewed on this topic, more prospective studies are suggested.
Collapse
Affiliation(s)
- Parvin MIRMIRAN
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeynab AMIRHAMIDI
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh-Sadat EJTAHED
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra BAHADORAN
- Nutrition and Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran,Corresponding Author:
| | - Fereidoun AZIZI
- Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
67
|
Heianza Y, Ma W, Manson JE, Rexrode KM, Qi L. Gut Microbiota Metabolites and Risk of Major Adverse Cardiovascular Disease Events and Death: A Systematic Review and Meta-Analysis of Prospective Studies. J Am Heart Assoc 2017; 6:JAHA.116.004947. [PMID: 28663251 PMCID: PMC5586261 DOI: 10.1161/jaha.116.004947] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Gut microbial metabolites have been implicated as novel risk factors for cardiovascular events and premature death. The strength and consistency of associations between blood concentrations of the gut microbial metabolites, trimethylamine-N-oxide (TMAO) and its precursors, with major adverse cardiovascular events (MACE) or death have not been comprehensively assessed. We quantified associations of blood concentrations of TMAO and its precursors with risks of MACE and mortality. METHODS AND RESULTS PubMed and Embase databases were searched up, and a total of 19 prospective studies from 16 publications (n=19 256, including 3315 incident cases) with quantitative estimates of the associations of TMAO with the development of MACE or death were included in our main analysis. Multivariate-adjusted relative risks (RRs) were used when these were available. Elevated concentrations of TMAO were associated with a pooled RR of 1.62 (95% CI, 1.45, 1.80; Pheterogeneity=0.2; I2=23.5%) for MACE compared with low TMAO levels, and 1 study of black participants influenced the heterogeneity of the association. After excluding the data of blacks, the RRs were not different according to body mass index, prevalence of diabetes mellitus, history of cardiovascular diseases, and kidney dysfunction. Furthermore, elevated TMAO concentrations were associated with a pooled RR of 1.63 (1.36, 1.95) for all-cause mortality. Individuals with elevated concentrations of TMAO precursors (l-carnitine, choline, or betaine) had an approximately 1.3 to 1.4 times higher risk for MACE compared to those with low concentrations. CONCLUSIONS Elevated concentrations of TMAO and its precursors were associated with increased risks of MACE and all-cause mortality independently of traditional risk factors.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Wenjie Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Kathryn M Rexrode
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
68
|
Demine S, Tejerina S, Bihin B, Thiry M, Reddy N, Renard P, Raes M, Jadot M, Arnould T. Mild mitochondrial uncoupling induces HSL/ATGL-independent lipolysis relying on a form of autophagy in 3T3-L1 adipocytes. J Cell Physiol 2017; 233:1247-1265. [PMID: 28488768 DOI: 10.1002/jcp.25994] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
Obesity is characterized by an excessive triacylglycerol accumulation in white adipocytes. Various mechanisms allowing the tight regulation of triacylglycerol storage and mobilization by lipid droplet-associated proteins as well as lipolytic enzymes have been identified. Increasing energy expenditure by inducing a mild uncoupling of mitochondria in adipocytes might represent a putative interesting anti-obesity strategy as it reduces the adipose tissue triacylglycerol content (limiting alterations caused by cell hypertrophy) by stimulating lipolysis through yet unknown mechanisms, limiting the adverse effects of adipocyte hypertrophy. Herein, the molecular mechanisms involved in lipolysis induced by a mild uncoupling of mitochondria in white 3T3-L1 adipocytes were characterized. Mitochondrial uncoupling-induced lipolysis was found to be independent from canonical pathways that involve lipolytic enzymes such as HSL and ATGL. Finally, enhanced lipolysis in response to mitochondrial uncoupling relies on a form of autophagy as lipid droplets are captured by endolysosomal vesicles. This new mechanism of triacylglycerol breakdown in adipocytes exposed to mild uncoupling provides new insights on the biology of adipocytes dealing with mitochondria forced to dissipate energy.
Collapse
Affiliation(s)
- Stéphane Demine
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Silvia Tejerina
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Benoît Bihin
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Marc Thiry
- Laboratory of Cell Biology, GIGA-R, University of Liège, Liège, Belgium
| | - Nagabushana Reddy
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Patricia Renard
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Martine Raes
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Michel Jadot
- Laboratory of Molecular Physiology (URPhyM), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), Namur, Belgium
| |
Collapse
|
69
|
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: 252] [Impact Index Per Article: 31.5] [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
|
70
|
Hao L, Xia Z, Yang H, Wang J, Han M. Ionic liquid-based reagents improve the stability of midterm fecal sample storage. J Microbiol Methods 2017; 139:68-73. [PMID: 28506638 DOI: 10.1016/j.mimet.2017.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 11/17/2022]
Abstract
Fecal samples are widely used in metagenomic research, which aims to elucidate the relationship between human health and the intestinal microbiota. However, the best conditions for stable and reliable storage and transport of these samples at room temperature are still unknown, and whether samples stored at room temperature for several days will maintain their microbiota composition is still unknown. Here, we established and tested a preservation method using reagents containing imidazolium- or pyridinium-based ionic liquids. We stored human fecal samples in these reagents for up to 7 days at different temperatures. Subsequently, all samples were sequenced and compared with fresh samples and/or samples treated under other conditions. The 16S rRNA sequencing results suggested that ionic liquid-based reagents could stabilize the composition of the microbiota in fecal samples during a 7-day storage period, particularly when stored at room temperature. Thus, this method may have implications in the storage of fecal samples for metagenomic research.
Collapse
Affiliation(s)
- Lilan Hao
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China; BGI Research, BGI-Shenzhen, Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Zhongkui Xia
- BGI Research, BGI-Shenzhen, Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, China
| | - Huanming Yang
- BGI Research, BGI-Shenzhen, Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, China; James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Jian Wang
- BGI Research, BGI-Shenzhen, Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, China; James D. Watson Institute of Genome Sciences, Hangzhou, China
| | - Mo Han
- BGI Research, BGI-Shenzhen, Shenzhen, China; China National GeneBank-Shenzhen, BGI-Shenzhen, Shenzhen, China.
| |
Collapse
|
71
|
The Microbiota-Obesity Connection, Part 2. Holist Nurs Pract 2017; 31:204-209. [PMID: 28406874 DOI: 10.1097/hnp.0000000000000213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
72
|
Romieu I, Dossus L, Barquera S, Blottière HM, Franks PW, Gunter M, Hwalla N, Hursting SD, Leitzmann M, Margetts B, Nishida C, Potischman N, Seidell J, Stepien M, Wang Y, Westerterp K, Winichagoon P, Wiseman M, Willett WC. Energy balance and obesity: what are the main drivers? Cancer Causes Control 2017; 28:247-258. [PMID: 28210884 PMCID: PMC5325830 DOI: 10.1007/s10552-017-0869-z] [Citation(s) in RCA: 423] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/06/2017] [Indexed: 12/19/2022]
Abstract
PURPOSE The aim of this paper is to review the evidence of the association between energy balance and obesity. METHODS In December 2015, the International Agency for Research on Cancer (IARC), Lyon, France convened a Working Group of international experts to review the evidence regarding energy balance and obesity, with a focus on Low and Middle Income Countries (LMIC). RESULTS The global epidemic of obesity and the double burden, in LMICs, of malnutrition (coexistence of undernutrition and overnutrition) are both related to poor quality diet and unbalanced energy intake. Dietary patterns consistent with a traditional Mediterranean diet and other measures of diet quality can contribute to long-term weight control. Limiting consumption of sugar-sweetened beverages has a particularly important role in weight control. Genetic factors alone cannot explain the global epidemic of obesity. However, genetic, epigenetic factors and the microbiota could influence individual responses to diet and physical activity. CONCLUSION Energy intake that exceeds energy expenditure is the main driver of weight gain. The quality of the diet may exert its effect on energy balance through complex hormonal and neurological pathways that influence satiety and possibly through other mechanisms. The food environment, marketing of unhealthy foods and urbanization, and reduction in sedentary behaviors and physical activity play important roles. Most of the evidence comes from High Income Countries and more research is needed in LMICs.
Collapse
Affiliation(s)
- Isabelle Romieu
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France.
| | - Laure Dossus
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Simón Barquera
- Centro de Investigación en Nutrición y Salud, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Hervé M Blottière
- Micalis Institute, MGP MetagenoPolis, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, CRC, University hospital Malmö, Malmö, Sweden
| | - Marc Gunter
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Nahla Hwalla
- Faculty of Agricultural and Food Science, American University of Beirut, Beirut, Lebanon
| | - Stephen D Hursting
- Department of Nutrition and the Nutrition Research Institute, The University of North Carolina, Chapel Hill, USA
| | - Michael Leitzmann
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Barrie Margetts
- Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| | - Chizuru Nishida
- Nutrition Policy and Scientific Advice (NPU), Department of Nutrition for Health and Development (NHD), World Health Organization (WHO), Geneva, Switzerland
| | - Nancy Potischman
- Office of the Associate Director, Applied Research Program, Division of Cancer Control and Population Sciences, National Cancer Institute, Bethesda, USA
| | - Jacob Seidell
- Faculty of Earth and Life Sciences, Department of Health Sciences, University Amsterdam, Amsterdam, The Netherlands
| | - Magdalena Stepien
- Nutrition and Metabolism Section, International Agency for Research on Cancer, 150 cours Albert Thomas, 69372, Lyon Cedex 08, France
| | - Youfa Wang
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, Joint Appointments, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, USA
| | - Klaas Westerterp
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | | | | | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, USA
| |
Collapse
|
73
|
High-fat meal, systemic inflammation and glucose homeostasis in obese children and adolescents. Int J Obes (Lond) 2017; 41:986-989. [PMID: 28216642 DOI: 10.1038/ijo.2017.48] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 01/13/2017] [Accepted: 01/31/2017] [Indexed: 12/22/2022]
Abstract
We aimed to assess in obese youths the relationships between interleukin-6 (IL-6), fat meal-induced endotoxemia and glucose homeostasis. Twenty obese children/adolescents (9-17 years old, 11 boys) underwent a standard oral glucose tolerance test and, 7-14 days later, a 5-h fat meal test (fat=69% of energy, saturated/monounsaturated/polyunsaturated fatty acids=31.5%/35%/33.5%), with serial measures of IL-6 and two markers of lipopolysaccharide (LPS) exposure and translocation, LPS-binding protein (LBP) and soluble CD14 (sCD14). IL-6 correlated not only with basal (homeostatic model assessment-insulin resistance) but also with post-prandial (Matsuda index) insulin sensitivity (r=0.61 (0.24-0.82), P=0.005, r=-0.53 (0.12-0.78), P=0.03, respectively). IL-6 did not change after the meal whereas LBP and sCD14 decreased significantly, indicating LPS translocation. Neither basal sCD14 and LBP nor their incremental concentrations correlated with IL-6 or glucose homeostasis. In our sample, IL-6 was associated with insulin sensitivity but not with LPS exposure, suggesting that meals with a balanced content of saturated/monounsaturated/polyunsaturated fatty acids may not be associated with LPS-induced inflammation and metabolic impairment.
Collapse
|
74
|
Doré J, Multon MC, Béhier JM, Affagard H, Andremont A, Barthélémy P, Batista R, Bonneville M, Bonny C, Boyaval G, Chamaillard M, Chevalier MP, Cordaillat-Simmons M, Cournarie F, Diaz I, Guillaume E, Guyard C, Jouvin-Marche E, Martin FP, Petiteau D. Microbiote intestinal : qu’en attendre au plan physiologique et thérapeutique ? Therapie 2017; 72:1-19. [PMID: 28214070 DOI: 10.1016/j.therap.2017.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Joël Doré
- INRA, Metagenopolis, 78350 Jouy-en-Josas, France
| | | | | | | | | | - Antoine Andremont
- Hôpital Bichat, université Paris Diderot, AP-HP, 92240 Malakoff, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
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: 8.6] [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]
|
76
|
Klinder A, Shen Q, Heppel S, Lovegrove JA, Rowland I, Tuohy KM. Impact of increasing fruit and vegetables and flavonoid intake on the human gut microbiota. Food Funct 2017; 7:1788-96. [PMID: 26757793 DOI: 10.1039/c5fo01096a] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Epidemiological studies have shown protective effects of fruits and vegetables (F&V) in lowering the risk of developing cardiovascular diseases (CVD) and cancers. Plant-derived dietary fibre (non-digestible polysaccharides) and/or flavonoids may mediate the observed protective effects particularly through their interaction with the gut microbiota. The aim of this study was to assess the impact of fruit and vegetable (F&V) intake on gut microbiota, with an emphasis on the role of flavonoids, and further to explore relationships between microbiota and factors associated with CVD risk. In the study, a parallel design with 3 study groups, participants in the two intervention groups representing high-flavonoid (HF) and low flavonoid (LF) intakes were asked to increase their daily F&V intake by 2, 4 and 6 portions for a duration of 6 weeks each, while a third (control) group continued with their habitual diet. Faecal samples were collected at baseline and after each dose from 122 subjects. Faecal bacteria enumeration was performed by fluorescence in situ hybridisation (FISH). Correlations of dietary components, flavonoid intake and markers of CVD with bacterial numbers were also performed. A significant dose X treatment interaction was only found for Clostidium leptum-Ruminococcus bromii/flavefaciens with a significant increase after intake of 6 additional portions in the LF group. Correlation analysis of the data from all 122 subjects independent from dietary intervention indicated an inhibitory role of F&V intake, flavonoid content and sugars against the growth of potentially pathogenic clostridia. Additionally, we observed associations between certain bacterial populations and CVD risk factors including plasma TNF-α, plasma lipids and BMI/waist circumference.
Collapse
Affiliation(s)
- Annett Klinder
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK. and Clinic of Orthopaedics, University Medicine Rostock, Rostock, Germany
| | - Qing Shen
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK.
| | - Susanne Heppel
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK.
| | - Julie A Lovegrove
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK.
| | - Ian Rowland
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK.
| | - Kieran M Tuohy
- Department of Food and Nutritional Sciences, University of Reading, Reading, Berkshire, UK. and Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach, San Michele all'Adige, Trento, Italy
| |
Collapse
|
77
|
Doré J, Multon MC, Béhier JM. The human gut microbiome as source of innovation for health: Which physiological and therapeutic outcomes could we expect? Therapie 2017; 72:21-38. [PMID: 28131442 DOI: 10.1016/j.therap.2016.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
From the moment of birth, each human being builds a microbe-host symbiosis which is key for the preservation of its health and well-being. This personal symbiotic coexistence is the result of progressive enrichments in microorganism diversity through external supplies. This diversity is nowadays massively overthrown by drastic changes related to clinical practice in birth management, environmental exposure, nutrition and healthcare behaviors. The last two generations have been the frame of massive modifications in life and food habits, with people being more and more sedentary, overfed and permeated with drugs and pollutants. We are now able to measure the impact of these changes on the gut microbiota diversity. Concomitantly, these modifications of lifestyle were associated with a dramatic increase in incidence of immune-mediated diseases including metabolic, allergic and inflammatory diseases and most likely neurodegenerative and psychiatric disorders. Microbiota is becoming a hot topic in the scientific community and in the mainstream media. The number of scientific publications increased by up to a factor three over the last five years, with gastrointestinal and metabolic diseases being the most productive areas. In the intellectual property landscape, the patent families on microbiota have more than doubled in the meantime. In parallel, funding either from National Institutes (e.g. from NIH which funds research mainly in the field of allergies, infections, cancer and cardiovascular diseases, from the White House which launched the national microbiome initiative) or by pharmaceutical companies follow the same trend, showing a boost and a strong support in the research field on microbiota. All major health players are investing in microbiome research as shown by the number of deals signed and by funding during 2015. The Giens round table addressed how the medicine of tomorrow, considering human beings as a human-microbe symbiotic supraorganism, could leverage microbiome knowledge and tools. The rationale for our working group has been structured around four domains of innovation that could derive from ongoing efforts in deciphering the interactions between human cells and intestinal microbiome as a central component of human health, namely: (1) development of stratification and monitoring tools; (2) identification of new target and drug discovery, as a part of our supra-genome; (4) exploitation of microbiota as a therapeutic target that can be modulated; (4) and finally as a source of live biotherapeutics and adjuvants. These four streams will exemplify how microbiota has changed the way we consider a wide range of chronic and incurable diseases and the consequences of long-lasting dysbiosis. In-depth microbiota analysis is opening one of the broadest fields of investigation for improving human and animal health and will be a source of major therapeutic innovations for tackling today's medical unmet needs. We thus propose a range of recommendations for basic researchers, care givers as well as for health authorities to gain reliability in microbiome analysis and accelerate discovery processes and their translation into applications for the benefits of the people. Finally, les Ateliers de Giens round table on microbiota benefited from the richness of the French ecosystem. France represents a center of excellence in the microbiota research field, with French institutions as Institut national de la recherche agronomique (INRA [Metagenopolis, Micalis]), Centre national de la recherché scientifique (CNRS), Unité de recherche sur les maladies infectieuses et tropicales émergentes (URMITE), Institut of Cardiometabolism and Nutrition (ICAN), Institut des maladies métaboliques et cardiovasculaires (I2MC), Institut national de la santé et de la recherche médicale (Inserm), Pasteur Institute and Gustave-Roussy being top-players for the number of publications.
Collapse
Affiliation(s)
- Joël Doré
- Institut national de la recherche agronomique (INRA), Metagenopolis, 78350 Jouy-en-Josas, France
| | - Marie-Christine Multon
- Sanofi R&D, unité sciences translationnelles, 13, quai Jules-Guesde, 94403 Vitry sur Seine, France.
| | | | | |
Collapse
|
78
|
Franco-de-Moraes AC, de Almeida-Pititto B, da Rocha Fernandes G, Gomes EP, da Costa Pereira A, Ferreira SRG. Worse inflammatory profile in omnivores than in vegetarians associates with the gut microbiota composition. Diabetol Metab Syndr 2017; 9:62. [PMID: 28814977 PMCID: PMC5557559 DOI: 10.1186/s13098-017-0261-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/05/2017] [Indexed: 02/06/2023] Open
Abstract
AIMS To describe the abundance of major phyla and some genera in the gut microbiota of individuals according to dietary habits and examine their associations with inflammatory markers, insulin resistance, and cardiovascular risk profile. METHODS A total of 268 non-diabetic individuals were stratified into groups of dietary types (strict vegetarians, lacto-ovo-vegetarians, and omnivores). The taxonomic composition and phylogenetic structure of the microbiota were obtained through the analysis of the 16S rRNA gene. Samples were clustered into operational taxonomic units at 97% similarity using GreenGenes 13.5 database. Clinical, biochemical, and circulating inflammatory markers were compared by ANOVA or Kruskal-Wallis test. RESULTS The sample (54.2% women, mean age 49.5 years) was composed of 66 strict vegetarians, 102 lacto-ovo-vegetarians and 100 omnivores. Considering the entire sample, the greatest abundant phyla were Firmicutes (40.7 ± 15.9%) and Bacteroidetes (39.5 ± 19.9%), and no difference in abundances was found between individuals with normal and excess weight. Stratifying by dietary types, the proportion of Firmicutes was lower and of Bacteroidetes was higher in strict vegetarians when compared to lacto-ovo-vegetarians and omnivores. At the genus level, strict vegetarians had a higher Prevotella abundance and Prevotella/Bacteroides ratio than the other groups. They also had a lower proportion of Faecalibacterium than lacto-ovo-vegetarians, and both vegetarian groups had higher proportions than did omnivores. Succinivibrio and Halomonas from the Proteobacteria phylum were overrepresented in omnivores. The omnivorous group showed higher values of anthropometric data, insulin, HOMA-IR, and a worse lipid profile. Inflammatory markers exhibited a gradual and significant increase from the vegetarians and lacto-ovo-vegetarians to the omnivorous group. CONCLUSIONS There are differences in gut microbiota composition of individuals with distinct dietary habits, who differ according to their inflammatory and metabolic profiles. Based on the findings relative to bacteria abundances and on their recognized actions in the metabolism, we suggest that exposure to animal foods may favor an intestinal environment which could trigger systemic inflammation and insulin resistance-dependent metabolic disorders.
Collapse
Affiliation(s)
- Ana Carolina Franco-de-Moraes
- Department of Epidemiology, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Sao Paulo, SP Zip code 01246-904 Brazil
| | - Bianca de Almeida-Pititto
- Department of Preventive Medicine, Federal University of Sao Paulo, Rua Botucatu, 720, Sao Paulo, SP Zip code 04023-900 Brazil
| | - Gabriel da Rocha Fernandes
- Oswaldo Cruz Foundation, René Rachou Research Center, Av. Augusto de Lima, 1715, Belo Horizonte, MG Zip code 30190-002 Brazil
| | - Everton Padilha Gomes
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, Medical School, University of Sao Paulo, Av. Dr. Eneas de Carvalho Aguiar, 44, 10°. andar, Sao Paulo, SP Zip code 05403-000 Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, Medical School, University of Sao Paulo, Av. Dr. Eneas de Carvalho Aguiar, 44, 10°. andar, Sao Paulo, SP Zip code 05403-000 Brazil
| | - Sandra Roberta G. Ferreira
- Department of Epidemiology, School of Public Health, University of Sao Paulo, Av. Dr. Arnaldo, 715, Sao Paulo, SP Zip code 01246-904 Brazil
| |
Collapse
|
79
|
Blottière HM, Doré J. Impact des nouveaux outils de métagénomique sur notre connaissance du microbiote intestinal et de son rôle en santé humaine. Med Sci (Paris) 2016; 32:944-951. [DOI: 10.1051/medsci/20163211009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
80
|
Verger EO, Armstrong P, Nielsen T, Chakaroun R, Aron-Wisnewsky J, Gøbel RJ, Schütz T, Delaere F, Gausseres N, Clément K, Holmes BA. Dietary Assessment in the MetaCardis Study: Development and Relative Validity of an Online Food Frequency Questionnaire. J Acad Nutr Diet 2016; 117:878-888. [PMID: 28024800 DOI: 10.1016/j.jand.2016.10.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/26/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND The European study MetaCardis aims to investigate the role of the gut microbiota in health and cardiometabolic diseases in France, Germany, and Denmark. To evaluate long-term diet-disease relationships, a food frequency questionnaire (FFQ) was found to be the most relevant dietary assessment method for the MetaCardis study. OBJECTIVE The objectives of this study were to describe the development of three semiquantitative online FFQs used in the MetaCardis study-one FFQ per country-and to assess the relative validity of the French MetaCardis FFQ. DESIGN The layout and format of the MetaCardis FFQ was based on the European Prospective Investigation of Cancer (EPIC)-Norfolk FFQ and the content was based on relevant European FFQs. Portion size and nutrient composition were derived from national food consumption surveys and food composition databases. To assess the validity of the French MetaCardis FFQ, a cross-sectional study design was utilized. PARTICIPANTS/SETTING The validation study included 324 adults recruited between September 2013 and June 2015 from different hospitals in Paris, France. MAIN OUTCOME MEASURES Food intakes were measured with both the French MetaCardis FFQ and 3 consecutive self-administered web-based 24-hour dietary recalls (DRs). STATISTICAL ANALYSES PERFORMED Several measures of validity of the French MetaCardis FFQ were evaluated: estimations of food groups, energy, and nutrient intakes from the DRs and the FFQ, Spearman and Pearson correlations, cross-classification, and Bland-Altman analyses. RESULTS The French MetaCardis FFQ tended to report higher food, energy, and nutrient intakes compared with the DRs. Mean correlation coefficient was 0.429 for food, 0.460 for energy, 0.544 for macronutrients, 0.640 for alcohol, and 0.503 for micronutrient intakes. Almost half of participants (44.4%) were correctly classified within tertiles of consumption, whereas 12.9% were misclassified in the opposite tertile. Performance of the FFQ was relatively similar after stratification by sex. CONCLUSIONS The French MetaCardis FFQ was found to have an acceptable level of validity and may be a useful instrument to rank individuals based on their food and nutrient intakes.
Collapse
|
81
|
Bissonnette N, Jiang X, Matte J, Guay F, Talbot G, Bontempo V, Gong J, Wang Q, Lessard M. Effect of a post-weaning diet supplemented with functional feed additives on ileal transcriptome activity and serum cytokines in piglets challenged with lipopolysaccharide. Vet Immunol Immunopathol 2016; 182:136-149. [DOI: 10.1016/j.vetimm.2016.10.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 09/26/2016] [Accepted: 10/14/2016] [Indexed: 10/20/2022]
|
82
|
A systematic review of the effect of yogurt consumption on chronic diseases risk markers in adults. Eur J Nutr 2016; 56:1375-1392. [DOI: 10.1007/s00394-016-1341-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 10/23/2016] [Indexed: 01/11/2023]
|
83
|
High-fat diet modifies the PPAR-γ pathway leading to disruption of microbial and physiological ecosystem in murine small intestine. Proc Natl Acad Sci U S A 2016; 113:E5934-E5943. [PMID: 27638207 DOI: 10.1073/pnas.1612559113] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Diet is among the most important factors contributing to intestinal homeostasis, and basic functions performed by the small intestine need to be tightly preserved to maintain health. Little is known about the direct impact of high-fat (HF) diet on small-intestinal mucosal defenses and spatial distribution of the microbiota during the early phase of its administration. We observed that only 30 d after HF diet initiation, the intervillous zone of the ileum-which is usually described as free of bacteria-became occupied by a dense microbiota. In addition to affecting its spatial distribution, HF diet also drastically affected microbiota composition with a profile characterized by the expansion of Firmicutes (appearance of Erysipelotrichi), Proteobacteria (Desulfovibrionales) and Verrucomicrobia, and decrease of Bacteroidetes (family S24-7) and Candidatus arthromitus A decrease in antimicrobial peptide expression was predominantly observed in the ileum where bacterial density appeared highest. In addition, HF diet increased intestinal permeability and decreased cystic fibrosis transmembrane conductance regulator (Cftr) and the Na-K-2Cl cotransporter 1 (Nkcc1) gene and protein expressions, leading to a decrease in ileal secretion of chloride, likely responsible for massive alteration in mucus phenotype. This complex phenotype triggered by HF diet at the interface between the microbiota and the mucosal surface was reversed when the diet was switched back to standard composition or when mice were treated for 1 wk with rosiglitazone, a specific agonist of peroxisome proliferator-activated receptor-γ (PPAR-γ). Moreover, weaker expression of antimicrobial peptide-encoding genes and intervillous bacterial colonization were observed in Ppar-γ-deficient mice, highlighting the major role of lipids in modulation of mucosal immune defenses.
Collapse
|
84
|
Poledne R, Kralova Lesna I, Kralova A, Fronek J, Cejkova S. The relationship between non-HDL cholesterol and macrophage phenotypes in human adipose tissue. J Lipid Res 2016; 57:1899-1905. [PMID: 27481939 PMCID: PMC5036370 DOI: 10.1194/jlr.p068015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Indexed: 12/12/2022] Open
Abstract
Data from experimental animal models and in vitro studies suggest that both hyperlipoproteinemia and obesity predispose to development of proinflammatory pathways of macrophages within adipose tissue. The aim of this study was to analyze whether non-HDL cholesterol concentration in healthy living kidney donors (LKDs) is related to the number and phenotype of proinflammatory macrophages in visceral and subcutaneous adipose tissue. Adipose tissue samples were collected by cleansing the kidney grafts of LKDs obtained peroperatively. The stromal vascular fractions of these tissues were analyzed by flow cytometry. Proinflammatory macrophages were defined as CD14+ cells coexpressing CD16+ and high-expression CD36 as well (CD14+CD16+CD36+++), while CD16 negativity and CD163 positivity identified alternatively stimulated, anti-inflammatory macrophages. Non-HDL cholesterol concentration positively correlated to proinflammatory macrophages within visceral adipose tissue, with increased strength with more precise phenotype determination. On the contrary, the proportion of alternatively stimulated macrophages correlated negatively with non-HDL cholesterol. The present study suggests a relationship of non-HDL cholesterol concentration to the number and phenotype proportion of macrophages in visceral adipose tissue of healthy humans.
Collapse
Affiliation(s)
- Rudolf Poledne
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | - Ivana Kralova Lesna
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Anna Kralova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jiri Fronek
- Transplant Surgery Department, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Sona Cejkova
- Laboratory for Atherosclerosis Research, Centre for Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| |
Collapse
|
85
|
Bourke CD, Berkley JA, Prendergast AJ. Immune Dysfunction as a Cause and Consequence of Malnutrition. Trends Immunol 2016; 37:386-398. [PMID: 27237815 PMCID: PMC4889773 DOI: 10.1016/j.it.2016.04.003] [Citation(s) in RCA: 403] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/31/2016] [Accepted: 04/06/2016] [Indexed: 12/13/2022]
Abstract
Malnutrition, which encompasses under- and overnutrition, is responsible for an enormous morbidity and mortality burden globally. Malnutrition results from disordered nutrient assimilation but is also characterized by recurrent infections and chronic inflammation, implying an underlying immune defect. Defects emerge before birth via modifications in the immunoepigenome of malnourished parents, and these may contribute to intergenerational cycles of malnutrition. This review summarizes key recent studies from experimental animals, in vitro models, and human cohorts, and proposes that immune dysfunction is both a cause and a consequence of malnutrition. Focusing on childhood undernutrition, we highlight gaps in current understanding of immune dysfunction in malnutrition, with a view to therapeutically targeting immune pathways as a novel means to reduce morbidity and mortality. Undernourished children principally die of common infections, and immune defects are consistently demonstrated in under- and overnutrition. Parental malnutrition leads to epigenetic modifications of infant immune and metabolic genes. Healthy gut development relies on sensing of dietary nutrients, commensal, and pathogenic microbes via immune receptors. Recurrent infections, chronic inflammation, and enteropathy compound clinical malnutrition by altering gut structure and function. Immune cell activation and systemic proinflammatory mediator levels are increased in malnutrition. Malnutrition impairs immune priming by DC and monocytes, and impairs effector memory T cell function. Immune dysfunction can directly drive pathological processes in malnutrition, including malabsorption, increased metabolic demand, dysregulation of the growth hormone and HPA axes, and greater susceptibility to infection.
Collapse
Affiliation(s)
- Claire D Bourke
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK.
| | - James A Berkley
- Kenya Medical Research Institute (KEMRI)-Wellcome Trust Collaborative Research Programme, Centre for Geographic Medicine Research, Kifili, Kenya; Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK
| | - Andrew J Prendergast
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, London, UK; Zvitambo Institute for Maternal and Child Health Research, Harare, Zimbabwe
| |
Collapse
|
86
|
Dao MC, Everard A, Clément K, Cani PD. Losing weight for a better health: Role for the gut microbiota. CLINICAL NUTRITION EXPERIMENTAL 2016; 6:39-58. [PMID: 33094147 PMCID: PMC7567023 DOI: 10.1016/j.yclnex.2015.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 12/12/2015] [Indexed: 01/07/2023]
Abstract
In recent years, there have been several reviews on gut microbiota, obesity and cardiometabolism summarizing interventions that may impact the gut microbiota and have beneficial effects on the host (some examples include [1–3]). In this review we discuss how the gut microbiota changes with weight loss (WL) interventions in relation to clinical and dietary parameters. We also evaluate available evidence on the heterogeneity of response to these interventions. Two important questions were generated in this regard: 1) Can response to an intervention be predicted? 2) Could pre-intervention modifications to the gut microbiota optimize WL and metabolic improvement? Finally, we have delineated some recommendations for future research, such as the importance of assessment of diet and other environmental exposures in WL intervention studies, and the need to shift to more integrative approaches of data analysis.
Collapse
Affiliation(s)
- Maria Carlota Dao
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, UMR S U1166, Nutriomics Team, Paris, France
- Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| | - Amandine Everard
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition Research Group, Av. E. Mounier, 73 Box B1.73.11, B-1200 Brussels, Belgium
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, France
- INSERM, UMR S U1166, Nutriomics Team, Paris, France
- Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
- Corresponding authors.
| | - Patrice D. Cani
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition Research Group, Av. E. Mounier, 73 Box B1.73.11, B-1200 Brussels, Belgium
- Corresponding authors.
| |
Collapse
|
87
|
Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Verger EO, Kayser BD, Levenez F, Chilloux J, Hoyles L, Dumas ME, Rizkalla SW, Doré J, Cani PD, Clément K. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 2016; 65:426-36. [PMID: 26100928 DOI: 10.1136/gutjnl-2014-308778] [Citation(s) in RCA: 1296] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 05/01/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Individuals with obesity and type 2 diabetes differ from lean and healthy individuals in their abundance of certain gut microbial species and microbial gene richness. Abundance of Akkermansia muciniphila, a mucin-degrading bacterium, has been inversely associated with body fat mass and glucose intolerance in mice, but more evidence is needed in humans. The impact of diet and weight loss on this bacterial species is unknown. Our objective was to evaluate the association between faecal A. muciniphila abundance, faecal microbiome gene richness, diet, host characteristics, and their changes after calorie restriction (CR). DESIGN The intervention consisted of a 6-week CR period followed by a 6-week weight stabilisation diet in overweight and obese adults (N=49, including 41 women). Faecal A. muciniphila abundance, faecal microbial gene richness, diet and bioclinical parameters were measured at baseline and after CR and weight stabilisation. RESULTS At baseline A. muciniphila was inversely related to fasting glucose, waist-to-hip ratio and subcutaneous adipocyte diameter. Subjects with higher gene richness and A. muciniphila abundance exhibited the healthiest metabolic status, particularly in fasting plasma glucose, plasma triglycerides and body fat distribution. Individuals with higher baseline A. muciniphila displayed greater improvement in insulin sensitivity markers and other clinical parameters after CR. These participants also experienced a reduction in A. muciniphila abundance, but it remained significantly higher than in individuals with lower baseline abundance. A. muciniphila was associated with microbial species known to be related to health. CONCLUSIONS A. muciniphila is associated with a healthier metabolic status and better clinical outcomes after CR in overweight/obese adults. The interaction between gut microbiota ecology and A. muciniphila warrants further investigation. TRIAL REGISTRATION NUMBER NCT01314690.
Collapse
Affiliation(s)
- Maria Carlota Dao
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France INSERM, UMR S U1166, Nutriomics Team, Paris, France Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| | - Amandine Everard
- Université Catholique de Louvain, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life Sciences and BIOtechnology), Brussels, Belgium
| | - Judith Aron-Wisnewsky
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France INSERM, UMR S U1166, Nutriomics Team, Paris, France Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| | - Nataliya Sokolovska
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France INSERM, UMR S U1166, Nutriomics Team, Paris, France Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| | - Edi Prifti
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France
| | - Eric O Verger
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France INSERM, UMR S U1166, Nutriomics Team, Paris, France Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| | - Brandon D Kayser
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France
| | - Florence Levenez
- INRA, US1367 MetaGenoPolis, Jouy-en-Josas, France AgroParisTech, UMR1319 MICALIS, Jouy-en-Josas, France
| | - Julien Chilloux
- Imperial College London, Section of Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, London, UK
| | - Lesley Hoyles
- Imperial College London, Section of Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, London, UK
| | | | - Marc-Emmanuel Dumas
- Imperial College London, Section of Biomolecular Medicine, Division of Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine, London, UK
| | - Salwa W Rizkalla
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France
| | - Joel Doré
- INRA, US1367 MetaGenoPolis, Jouy-en-Josas, France AgroParisTech, UMR1319 MICALIS, Jouy-en-Josas, France
| | - Patrice D Cani
- Université Catholique de Louvain, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life Sciences and BIOtechnology), Brussels, Belgium
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière hospital, Paris, France INSERM, UMR S U1166, Nutriomics Team, Paris, France Sorbonne Universités, UPMC University Paris 06, UMR_S 1166 I, Nutriomics Team, Paris, France
| |
Collapse
|
88
|
Patterson E, Ryan PM, Cryan JF, Dinan TG, Ross RP, Fitzgerald GF, Stanton C. Gut microbiota, obesity and diabetes. Postgrad Med J 2016; 92:286-300. [PMID: 26912499 DOI: 10.1136/postgradmedj-2015-133285] [Citation(s) in RCA: 360] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/28/2016] [Indexed: 02/06/2023]
Abstract
The central role of the intestinal microbiota in the progression and, equally, prevention of metabolic dysfunction is becoming abundantly apparent. The symbiotic relationship between intestinal microbiota and host ensures appropriate development of the metabolic system in humans. However, disturbances in composition and, in turn, functionality of the intestinal microbiota can disrupt gut barrier function, a trip switch for metabolic endotoxemia. This low-grade chronic inflammation, brought about by the influx of inflammatory bacterial fragments into circulation through a malfunctioning gut barrier, has considerable knock-on effects for host adiposity and insulin resistance. Conversely, recent evidence suggests that there are certain bacterial species that may interact with host metabolism through metabolite-mediated stimulation of enteric hormones and other systems outside of the gastrointestinal tract, such as the endocannabinoid system. When the abundance of these keystone species begins to decline, we see a collapse of the symbiosis, reflected in a deterioration of host metabolic health. This review will investigate the intricate axis between the microbiota and host metabolism, while also addressing the promising and novel field of probiotics as metabolic therapies.
Collapse
Affiliation(s)
- Elaine Patterson
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| | - Paul M Ryan
- Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Co. Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Department of Psychiatry and Neurobehavioural Science, University College Cork, Co. Cork, Ireland
| | - R Paul Ross
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland College of Science, Engineering and Food Science, University College Cork, Co. Cork, Ireland
| | - Gerald F Fitzgerald
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland School of Microbiology, University College Cork, Co. Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Institute, University College Cork, Co. Cork, Ireland Food Biosciences Department, Teagasc Food Research Centre, Fermoy, Co. Cork, Ireland
| |
Collapse
|
89
|
Kobyliak N, Conte C, Cammarota G, Haley AP, Styriak I, Gaspar L, Fusek J, Rodrigo L, Kruzliak P. Probiotics in prevention and treatment of obesity: a critical view. Nutr Metab (Lond) 2016; 13:14. [PMID: 26900391 PMCID: PMC4761174 DOI: 10.1186/s12986-016-0067-0] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022] Open
Abstract
The worldwide prevalence of obesity more than doubled between 1980 and 2014. The obesity pandemic is tightly linked to an increase in energy availability, sedentariness and greater control of ambient temperature that have paralleled the socioeconomic development of the past decades. The most frequent cause which leads to the obesity development is a dysbalance between energy intake and energy expenditure. The gut microbiota as an environmental factor which influence whole-body metabolism by affecting energy balance but also inflammation and gut barrier function, integrate peripheral and central food intake regulatory signals and thereby increase body weight. Probiotics have physiologic functions that contribute to the health of gut microbiota, can affect food intake and appetite, body weight and composition and metabolic functions through gastrointestinal pathways and modulation of the gut bacterial community.
Collapse
Affiliation(s)
- Nazarii Kobyliak
- Division of Endocrinology and Metabolic Diseases, Catholic University of Sacred Heart, A. Gemelli Medical School, Rome, Italy
| | - Caterina Conte
- Division of Endocrinology and Metabolic Diseases, Catholic University of Sacred Heart, A. Gemelli Medical School, Rome, Italy
| | - Giovanni Cammarota
- Division of Internal Medicine and Gastroenterology, Catholic University of Sacred Heart, A. Gemelli Medical School, Rome, Italy
| | - Andreana P Haley
- Department of Psychology, The University of Texas at Austin, Austin, TX USA ; University of Texas Imaging Research Center, Austin, TX USA
| | - Igor Styriak
- Institute of Geotechnics, Department of Biotechnology, Slovak Academy of Sciences, Kosice, Slovak Republic
| | - Ludovit Gaspar
- 2nd Department of Internal Medicine, Comenius University and University Hospital, Mickiewiczova 13, 813 69 Bratislava, Slovak Republic
| | - Jozef Fusek
- Faculty of Health Studies, University of Pardubice, Pardubice, Czech Republic
| | - Luis Rodrigo
- Department of Gastroenterology, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- Department of Gastroenterology, Central University Hospital of Asturias (HUCA), Oviedo, Spain ; 2nd Department of Internal Medicine, Faculty of Medicine, Masaryk University, Pekarska 53, 656 91 Brno, Czech Republic ; Laboratory of Structural Biology and Proteomics, Central Laboratories, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
90
|
Hutkins RW, Krumbeck JA, Bindels LB, Cani PD, Fahey G, Goh YJ, Hamaker B, Martens EC, Mills DA, Rastal RA, Vaughan E, Sanders ME. Prebiotics: why definitions matter. Curr Opin Biotechnol 2016; 37:1-7. [PMID: 26431716 PMCID: PMC4744122 DOI: 10.1016/j.copbio.2015.09.001] [Citation(s) in RCA: 264] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 08/21/2015] [Accepted: 09/02/2015] [Indexed: 01/03/2023]
Abstract
The prebiotic concept was introduced twenty years ago, and despite several revisions to the original definition, the scientific community has continued to debate what it means to be a prebiotic. How prebiotics are defined is important not only for the scientific community, but also for regulatory agencies, the food industry, consumers and healthcare professionals. Recent developments in community-wide sequencing and glycomics have revealed that more complex interactions occur between putative prebiotic substrates and the gut microbiota than previously considered. A consensus among scientists on the most appropriate definition of a prebiotic is necessary to enable continued use of the term.
Collapse
Affiliation(s)
- Robert W Hutkins
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68583-0919, USA.
| | - Janina A Krumbeck
- Department of Food Science and Technology, University of Nebraska, Lincoln, NE 68583-0919, USA; School of Biological Sciences, University of Nebraska, Lincoln, NE 68583-0919, USA
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- Université Catholique de Louvain, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Av. E. Mounier, 73 B1.73.11, B-1200 Brussels, Belgium
| | - George Fahey
- Department of Animal Science, University of Illinois, Urbana, IL 61801, USA
| | - Yong Jun Goh
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | - Bruce Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Eric C Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - David A Mills
- Department of Food Science & Technology, University of California, Davis, CA 95616, USA
| | - Robert A Rastal
- Department of Food and Nutritional Sciences, The University of Reading, PO Box 226, Whiteknights, Reading RG6 6AP, United Kingdom
| | - Elaine Vaughan
- Sensus BV (Royal Cosun), Borchwerf 3, 4704RG Roosendaal, The Netherlands
| | | |
Collapse
|
91
|
Haghighatdoost F, Azadbakht L, Keshteli AH, Feinle-Bisset C, Daghaghzadeh H, Afshar H, Feizi A, Esmaillzadeh A, Adibi P. Glycemic index, glycemic load, and common psychological disorders. Am J Clin Nutr 2016; 103:201-209. [PMID: 26607943 DOI: 10.3945/ajcn.114.105445] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 10/09/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Potential associations between dietary glycemic index (GI) and glycemic load (GL) with psychological disorders remain uncertain. OBJECTIVE We investigated the relations of dietary GI and GL with psychological distress, anxiety, and depression. DESIGN A total of 3363 nonacademic members of the staff of Isfahan University of Medical Sciences were included in this cross-sectional study. GI and GL were assessed by using a validated, self-administered, dish-based, semiquantitative food-frequency questionnaire. Validated Iranian versions of the Hospital Anxiety and Depression Scale and General Health Questionnaire-12 were used to assess anxiety, depression, and psychological distress. RESULTS After control for potential confounders, individuals in the top tertile of GI had greater odds of depression (OR: 1.44; 95% CI: 1.03, 2.02; P-trend = 0.03) and a trend for greater odds of anxiety (OR: 1.52; 95% CI: 0.97, 2.38; P trend = 0.06) compared with those in the first tertile. Higher GL values were linked to lower odds for mental disorders (OR: 0.66; 95% CI: 0.49, 0.90; P-trend = 0.009), depression (OR: 0.69; 95% CI: 0.51, 0.93; P-trend = 0.02), and psychological distress (OR: 0.67; 95% CI: 0.48, 0.92; P-trend = 0.01). Significant interactions were observed between GI and sex for depression (P = 0.01) and psychological distress (P = 0.046) in the crude model. In stratified analyses by sex, after control for potential confounders, a greater GI was linked to a higher odds of depression (OR: 1.52; 95% CI: 1.20, 1.94; P-trend = 0.001) and psychological distress (OR: 1.66; 95% CI: 1.28, 2.14; P-trend = 0.001) in women but not in men. CONCLUSION Our findings support a direct link between the odds of depression and dietary GI but inverse associations between GL and mental disorders, depression, and psychological distress. This trial was registered at clinicaltrials.gov as NCT02362113.
Collapse
Affiliation(s)
- Fahimeh Haghighatdoost
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science
| | - Leila Azadbakht
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran;
| | | | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council Centre of Research Excellence of Translating Nutritional Science to Good Health, Adelaide, Australia
| | | | - Hamid Afshar
- Integrative Functional Gastroenterology Research Center, and
| | - Awat Feizi
- Biostatistics and Epidemiology Department, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Peyman Adibi
- Integrative Functional Gastroenterology Research Center, and
| |
Collapse
|
92
|
Heinsen FA, Fangmann D, Müller N, Schulte DM, Rühlemann MC, Türk K, Settgast U, Lieb W, Baines JF, Schreiber S, Franke A, Laudes M. Beneficial Effects of a Dietary Weight Loss Intervention on Human Gut Microbiome Diversity and Metabolism Are Not Sustained during Weight Maintenance. Obes Facts 2016; 9:379-391. [PMID: 27898428 PMCID: PMC5644845 DOI: 10.1159/000449506] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 08/29/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE In the present study, we examined the effect of a very low-calorie diet(VLCD)-based obesity program on human gut microbiome diversity and metabolism during weight loss and weight maintenance. METHODS Obese subjects underwent 3 months of VLCD followed by 3 months of weight maintenance. A lean and an obese control group were included. The microbiome was characterized by performing high-throughput dual-indexed 16S rDNA amplicon sequencing. RESULTS At baseline, a significant difference in the Firmicutes/Bacteroidetes ratio between the lean and obese individuals was observed (p = 0.047). The VLCD resulted in significant alterations in gut microbiome diversity from baseline to 3 months (p = 0.0053). Acinetobacter represented an indicator species for the observed effect (indicator value = 0.998, p = 0.006). Metabolic analyses revealed alterations of the bacterial riboflavin pathway from baseline to 3 months (pnom = 0.0078). These changes in diversity and bacterial metabolism induced by VLCD diminished during the weight maintenance phase, despite sustained reductions in body weight and sustained improvements of insulin sensitivity. CONCLUSION The present data show that a VLCD is able to beneficially alter both gut microbiome diversity and metabolism in obese humans, but that these changes are not sustained during weight maintenance. This finding might suggest that the microbiome should be targeted during obesity programs.
Collapse
Affiliation(s)
| | - Daniela Fangmann
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Nike Müller
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | | | - Malte C. Rühlemann
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Kathrin Türk
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Ute Settgast
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Wolfgang Lieb
- Institute for Epidemiology, University of Kiel, Kiel, Germany
| | - John F. Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University of Kiel, Kiel, Germany
| | - Matthias Laudes
- Department of Internal Medicine 1, University of Kiel, Kiel, Germany
- *Prof. Dr. Matthias Laudes, Klinik 1 für Innere Medizin, Universitätsklinikum Schleswig-Holstein, Arnold Heller Straße 3, 24105 Kiel, Germany,
| |
Collapse
|
93
|
Backer V, Baines KJ, Powell H, Porsbjerg C, Gibson PG. Increased asthma and adipose tissue inflammatory gene expression with obesity and Inuit migration to a western country. Respir Med 2015; 111:8-15. [PMID: 26764119 DOI: 10.1016/j.rmed.2015.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/12/2015] [Accepted: 12/14/2015] [Indexed: 01/15/2023]
Abstract
BACKGROUND An overlap between obesity and asthma exists, and inflammatory cells in adipose tissue could drive the development of asthma. Comparison of adipose tissue gene expression among Inuit living in Greenland to those in Denmark provides an opportunity to assess how changes in adipose tissue inflammation can be modified by migration and diet. OBJECTIVE To examine mast cell and inflammatory markers in adipose tissue and the association with asthma. METHODS Two Inuit populations were recruited, one living in Greenland and another in Denmark. All underwent adipose subcutaneous biopsy, followed by clinical assessment of asthma, and measurement of AHR. Adipose tissue biopsies were homogenised, RNA extracted, and PCR was performed to determine the relative gene expression of mast cell (tryptase, chymase, CPA3) and inflammatory markers (IL-6, IL-1β, and CD163). RESULTS Of the 1059 Greenlandic Inuit participants, 556 were living in Greenland and 6.4% had asthma. Asthma was increased in Denmark (9%) compared to Greenland (3.6%, p < 0.0001) and associated with increased adipose tissue IL-6 gene expression and increased BMI. There was no association between asthma and adipose tissue mast cell gene expression. Pro-inflammatory gene expression (IL-6, IL-1β) was higher in those living in Denmark, and with increasing BMI and dietary changes. The anti-inflammatory (M2) macrophage marker, CD163, was higher in Greenland-dwelling Inuit (p < 0.01). CONCLUSIONS No association was found between gene expression of mast cell markers in adipose tissue and asthma. Among Greenlandic Inuit, adipose tissue inflammation is also increased in those who migrate to Denmark, possibly as a result of dietary changes.
Collapse
Affiliation(s)
- Vibeke Backer
- Department of Respiratory Medicine L, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Katherine J Baines
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW 2305 Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, NSW 2305 Australia
| | - Heather Powell
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW 2305 Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, NSW 2305 Australia
| | - Celeste Porsbjerg
- Department of Respiratory Medicine L, Bispebjerg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter G Gibson
- Priority Research Centre for Asthma and Respiratory Diseases, Hunter Medical Research Institute, The University of Newcastle, New Lambton Heights, NSW 2305 Australia; Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, NSW 2305 Australia
| |
Collapse
|
94
|
Aron-Wisnewsky J, Clément K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nat Rev Nephrol 2015; 12:169-81. [PMID: 26616538 DOI: 10.1038/nrneph.2015.191] [Citation(s) in RCA: 223] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiometabolic diseases (CMDs) have been associated with changes in the composition of the gut microbiota, with links between the host environment and microbiota identified in preclinical models. High-throughput sequencing technology has facilitated in-depth studies of the gut microbiota, bacterial-derived metabolites, and their association with CMDs. Such strategies have shown that patients with CMDs frequently exhibit enrichment or depletion of certain bacterial groups in their resident microbiota compared to healthy individuals. Furthermore, the ability to transfer resident gut microbiota from mice or humans into germ-free mouse models, or between human patients, has enabled researchers to characterize the causative role of the gut microbiota in CMDs. These approaches have helped identify that dietary intake of choline, which is metabolized by the gut microbiota, is associated with cardiovascular outcomes in mice and humans. Trimethylamine N-oxide (TMAO) - a metabolite derived from the gut microbiota - is also associated with poor cardiovascular outcomes in patients with cardiovascular disease and is elevated in patients with chronic kidney disease (CKD). TMAO might represent a biomarker that links the environment and microbiota with CKD. This Review summarizes data suggesting a link between the gut microbiota and derived metabolites with food intake patterns, metabolic alterations, and chronic CMDs.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, INSERM, Sorbonne Université, Paris 6, Pitié-Salpêtrière hospital, F-75013 Paris, France
| | - Karine Clément
- Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, INSERM, Sorbonne Université, Paris 6, Pitié-Salpêtrière hospital, F-75013 Paris, France
| |
Collapse
|
95
|
Ciubotaru I, Green SJ, Kukreja S, Barengolts E. Significant differences in fecal microbiota are associated with various stages of glucose tolerance in African American male veterans. Transl Res 2015; 166. [PMID: 26209747 PMCID: PMC4916963 DOI: 10.1016/j.trsl.2015.06.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The importance of gut microbiota in pathogenesis of diabetes remains unknown. This study investigated the relationship between microbiota and metabolic markers in African American men (AAM) with prediabetes and hypovitaminosis D. The study was ancillary to a randomized trial of vitamin D supplementation with weekly ergocalciferol (50,000 IU) conducted in AAM veterans over 12 months (D Intervention in Veterans Affairs). Glycemic groups (Gr) were characterized based on changes in oral glucose tolerance between baseline and exit. Subjects with stable normal glucose tolerance were assigned to Gr-1 and those with stable prediabetes (impaired glucose tolerance and impaired fasting glucose) to Gr-2. Microbiota composition was analyzed in stool collected at the exit (n = 115) and compared between Gr-1 and Gr-2, as well as between the lowest and highest quartiles of dietary intake of energy and fat, hemoglobin A1c, and serum 25-hydroxyvitamin D (25[OH]D) level. Differences between Gr-1 and Gr-2 included the Bacteroidetes/Firmicutes and Bacteroidales/Clostridia ratios and differences in genera such as Ruminococcus and Dialister. Changes in specific taxa associated with the lowest and highest quartiles of 25(OH)D (eg, Ruminococcus, Roseburia, Blautia, Dorea) were clearly distinct from those of dietary intake (eg, Bacteroides, Bacteroides/Prevotella ratio) or A1c (eg, Faecalibacterium, Catenibacterium, Streptococcus). These findings suggest a novel interaction between microbiota and vitamin D and a role for microbiota in early stages of diabetes development. Although results suggest that specific taxa are associated with glycemic stability over time, a causative relationship between microbiota makeup and dysglycemia is still to be demonstrated.
Collapse
Affiliation(s)
- Irina Ciubotaru
- Department of Medicine, University of Illinois Medical Center, Chicago, Ill
| | - Stefan J Green
- Department of Medicine, University of Illinois Medical Center, Chicago, Ill
| | - Subhash Kukreja
- Department of Medicine, University of Illinois Medical Center, Chicago, Ill
| | - Elena Barengolts
- Department of Medicine, University of Illinois Medical Center, Chicago, Ill; Department of Medicine, Jesse Brown VA Medical Center, Chicago, Ill.
| |
Collapse
|
96
|
Natural environments, ancestral diets, and microbial ecology: is there a modern "paleo-deficit disorder"? Part II. J Physiol Anthropol 2015; 34:9. [PMID: 25889196 PMCID: PMC4353476 DOI: 10.1186/s40101-014-0040-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 12/29/2014] [Indexed: 12/19/2022] Open
Abstract
Famed microbiologist René J. Dubos (1901–1982) was an early pioneer in the developmental origins of health and disease (DOHaD) construct. In the 1960s, he conducted groundbreaking research concerning the ways in which early-life experience with nutrition, microbiota, stress, and other environmental variables could influence later-life health outcomes. He recognized the co-evolutionary relationship between microbiota and the human host. Almost 2 decades before the hygiene hypothesis, he suggested that children in developed nations were becoming too sanitized (vs. our ancestral past) and that scientists should determine whether the childhood environment should be “dirtied up in a controlled manner.” He also argued that oft-celebrated growth chart increases via changes in the global food supply and dietary patterns should not be equated to quality of life and mental health. Here in the second part of our review, we reflect the words of Dubos off contemporary research findings in the areas of diet, the gut-brain-axis (microbiota and anxiety and depression) and microbial ecology. Finally, we argue, as Dubos did 40 years ago, that researchers should more closely examine the relevancy of silo-sequestered, reductionist findings in the larger picture of human quality of life. In the context of global climate change and the epidemiological transition, an allergy epidemic and psychosocial stress, our review suggests that discussions of natural environments, urbanization, biodiversity, microbiota, nutrition, and mental health, are often one in the same.
Collapse
|
97
|
Doré J, Blottière H. The influence of diet on the gut microbiota and its consequences for health. Curr Opin Biotechnol 2015; 32:195-199. [PMID: 25615931 DOI: 10.1016/j.copbio.2015.01.002] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 12/29/2014] [Accepted: 01/03/2015] [Indexed: 02/07/2023]
Abstract
Man is an intimate symbiosis between 10 trillion human cells and some 100 trillion bacteria, most of which inhabit the intestine where they constitute an extremely dense and diverse microbiota. This symbiotic balance that has to be established within each newborn is key to the maintenance of health and well being. Its development is markedly influenced by microbial exposure encountered very early in life. Mode of infant feeding, and the post-weaning transition to habitual diet will further shape the microbiota. Recent studies support the concept that diet should be viewed as a means to prevent potentially durable alterations of symbiosis observed in immune-mediated metabolic and inflammatory diseases. Non-digestible dietary fiber will play a major role in this context.
Collapse
Affiliation(s)
- Joël Doré
- INRA, Micalis & MetaGenoPolis, Jouy-en-Josas, France.
| | | |
Collapse
|