51
|
Degradation of the Incretin Hormone Glucagon-Like Peptide-1 (GLP-1) by Enterococcus faecalis Metalloprotease GelE. mSphere 2020; 5:5/1/e00585-19. [PMID: 32051237 PMCID: PMC7021470 DOI: 10.1128/msphere.00585-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Humans have a complex and interconnected relationship with their gastrointestinal microbiomes, yet our interest in the microbiome tends to focus on overt pathogenic or probiotic activities, leaving the roles that commensal species may have on host physiology and metabolic processes largely unexplored. Commensal organisms in the microbiome produce and secrete many factors that have an opportunity to interact with the gastrointestinal tract and host biology. Here, we show that a secreted protease from E. faecalis, GelE, is able to degrade the gastrointestinal hormone GLP-1, which is responsible for regulating glucose homeostasis and appetite in the body. The disruption of natural GLP-1 signaling by GelE may have significant consequences for maintaining healthy blood glucose levels and in the development of metabolic disease. Furthermore, this work deepens our understanding of specific host-microbiome interactions. Metabolic diseases, including type 2 diabetes and obesity, have become increasingly prevalent global health concerns. Studies over the past decade have established connections between the gastrointestinal microbiota and host metabolism, but the mechanisms behind these connections are only beginning to be understood. We were interested in identifying microbes that have the ability to modulate the levels of the incretin hormone glucagon-like peptide-1 (GLP-1). Using a human-derived cell line that is capable of secreting GLP-1 in response to stimulatory ligands (NCI-H716), we identified supernatants from several bacterial isolates that were capable of decreasing GLP-1 levels, including several strains of Enterococcus faecalis. We further identified the secreted protease GelE, an established virulence factor from E. faecalis, as being responsible for GLP-1 inhibition via direct cleavage of GLP-1 by GelE. Finally, we demonstrated that E. faecalis supernatants can disrupt a colonic epithelial monolayer and cleave GLP-1 in a gelE-dependent manner. This work suggests that a secreted factor from an intestinal microbe can traverse the epithelial barrier and impact levels of an important intestinal hormone. IMPORTANCE Humans have a complex and interconnected relationship with their gastrointestinal microbiomes, yet our interest in the microbiome tends to focus on overt pathogenic or probiotic activities, leaving the roles that commensal species may have on host physiology and metabolic processes largely unexplored. Commensal organisms in the microbiome produce and secrete many factors that have an opportunity to interact with the gastrointestinal tract and host biology. Here, we show that a secreted protease from E. faecalis, GelE, is able to degrade the gastrointestinal hormone GLP-1, which is responsible for regulating glucose homeostasis and appetite in the body. The disruption of natural GLP-1 signaling by GelE may have significant consequences for maintaining healthy blood glucose levels and in the development of metabolic disease. Furthermore, this work deepens our understanding of specific host-microbiome interactions.
Collapse
|
52
|
Liu Y, Chen J, Tan Q, Deng X, Tsai PJ, Chen PH, Ye M, Guo J, Su Z. Nondigestible Oligosaccharides with Anti-Obesity Effects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:4-16. [PMID: 31829005 DOI: 10.1021/acs.jafc.9b06079] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Obesity has an important influence on health conditions, causing a multitude of complications and comorbidities, and drug therapy is considered to be one of the treatment strategies. Nowadays, there is increasing interest in the study of intestinal microbiota regulation of obesity; also, an increasing number of agricultural and sideline products have been found to have anti-obesity potential. In the present review, we summarize an overview of current known and potential anti-obesity oligosaccharides and their molecular structures. We describe their anti-obesity potential activity and the molecular structure associated with this activity, the regulation of intestinal microbiota composition and its mechanism of action, including regulation of the short-chain fatty acid (SCFA) pathway and altering bile acid (BA) pathway. This review will provide new ideas for us to develop new anti-obesity functional foods.
Collapse
Affiliation(s)
- Yongjian Liu
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Jiajia Chen
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Qiuhua Tan
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Xiaoyi Deng
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Ping-Ju Tsai
- King-Prebiotics Biotechnology (TW) CO., LTD. , Linkou District, New Taipei City 24446 , Taiwan China
| | - Pei-Hsuan Chen
- King-Prebiotics Biotechnology (TW) CO., LTD. , Linkou District, New Taipei City 24446 , Taiwan China
| | - Manxiang Ye
- New Francisco (Yunfu City) Biotechnology CO., LTD. , Swan-kan-chiau Industrial District, Kaofong Village Yunfu City 527343 , Guangdong , China
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Centre of Integrated Chinese and Western Medicine , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Zhengquan Su
- Guangdong Engineering Research Center of Natural Products and New Drugs, Guangdong Provincial University Engineering Technology Research Center of Natural Products and Drugs , Guangdong Pharmaceutical University , Guangzhou 510006 , China
| |
Collapse
|
53
|
Li S, Wang L, Liu B, He N. Unsaturated alginate oligosaccharides attenuated obesity-related metabolic abnormalities by modulating gut microbiota in high-fat-diet mice. Food Funct 2020; 11:4773-4784. [DOI: 10.1039/c9fo02857a] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A new potent prebiotic oligosaccharide for obesity and related metabolic abnormalities by modulating gut microbiota.
Collapse
Affiliation(s)
- Shangyong Li
- College of Basic Medicine
- Qingdao University
- Qingdao 266071
- China
| | - Linna Wang
- Yellow Sea Fisheries Research Institute
- Chinese Academy of Fishery Sciences
- Key Laboratory for Sustainable Development of Marine Fisheries
- Ministry of Agriculture
- Qingdao 266071
| | - Bo Liu
- School of Pharmacy
- Qingdao University
- Qingdao 266071
- China
| | - Ningning He
- College of Basic Medicine
- Qingdao University
- Qingdao 266071
- China
| |
Collapse
|
54
|
Yu Y, Raka F, Adeli K. The Role of the Gut Microbiota in Lipid and Lipoprotein Metabolism. J Clin Med 2019; 8:jcm8122227. [PMID: 31861086 PMCID: PMC6947520 DOI: 10.3390/jcm8122227] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/06/2019] [Accepted: 12/08/2019] [Indexed: 12/12/2022] Open
Abstract
Both environmental and genetic factors contribute to relative species abundance and metabolic characteristics of the intestinal microbiota. The intestinal microbiota and accompanying microbial metabolites differ substantially in those who are obese or have other metabolic disorders. Accumulating evidence from germ-free mice and antibiotic-treated animal models suggests that altered intestinal gut microbiota contributes significantly to metabolic disorders involving impaired glucose and lipid metabolism. This review will summarize recent findings on potential mechanisms by which the microbiota affects intestinal lipid and lipoprotein metabolism including microbiota dependent changes in bile acid metabolism which affects bile acid signaling by bile acid receptors FXR and TGR5. Microbiota changes also involve altered short chain fatty acid signaling and influence enteroendocrine cell function including GLP-1/GLP-2-producing L-cells which regulate postprandial lipid metabolism.
Collapse
Affiliation(s)
- Yijing Yu
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
| | - Fitore Raka
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; (Y.Y.); (F.R.)
- Departments of Laboratory Medicine & Pathobiology and Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +416-813-8682
| |
Collapse
|
55
|
Abstract
The regulation of glycemia is under a tight neuronal detection of glucose levels performed by the gut-brain axis and an efficient efferent neuronal message sent to the peripheral organs, as the pancreas to induce insulin and inhibit glucagon secretions. The neuronal detection of glucose levels is performed by the autonomic nervous system including the enteric nervous system and the vagus nerve innervating the gastro-intestinal tractus, from the mouth to the anus. A dysregulation of this detection leads to the one of the most important current health issue around the world i.e. diabetes mellitus. Furthemore, the consequences of diabetes mellitus on neuronal homeostasis and activities participate to the aggravation of the disease establishing a viscious circle. Prokaryotic cells as bacteria, reside in our gut. The strong relationship between prokaryotic cells and our eukaryotic cells has been established long ago, and prokaryotic and eukaryotic cells in our body have evolved synbiotically. For the last decades, studies demonstrated the critical role of the gut microbiota on the metabolic control and how its shift can induce diseases such as diabetes. Despite an important increase of knowledge, few is known about 1) how the gut microbiota influences the neuronal detection of glucose and 2) how the diabetes mellitus-induced gut microbiota shift observed participates to the alterations of autonomic nervous system and the gut-brain axis activity.
Collapse
Affiliation(s)
- Estelle Grasset
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, 41345, Gothenburg, Sweden.
| | - Remy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2 : 'Intestinal Risk Factors, Diabetes, Université Paul Sabatier (UPS), Dyslipidemia', F-31432, Toulouse, Cedex 4, France
| |
Collapse
|
56
|
Inulin Supplementation Reduces Systolic Blood Pressure in Women with Breast Cancer Undergoing Neoadjuvant Chemotherapy. Cardiovasc Ther 2019; 2019:5707150. [PMID: 31772611 PMCID: PMC6739761 DOI: 10.1155/2019/5707150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/05/2019] [Accepted: 05/12/2019] [Indexed: 01/14/2023] Open
Abstract
Introduction Breast cancer is the most frequently diagnosed malignancy in women, and comorbidities like hypertension and obesity diminish their quality of life and negatively affect their response to chemotherapy. Furthermore, inulin supplementation is associated with the reduction of cardiovascular diseases (CVD) risk. Objective To determine whether inulin supplementation prevents the elevation of blood pressure in women with breast cancer undergoing neoadjuvant therapy with cyclophosphamide and doxorubicin. Methods This was a randomized, double-blind placebo controlled trial which included women with early-stage breast cancer undergoing neoadjuvant therapy (n=38). Patients were randomly assigned to participate in two different groups to receive either 15 g of inulin or 15 g of placebo (maltodextrin) for 21 days. Body composition and blood pressure were evaluated before and after the supplementation period. Results Women in the inulin group showed a lower systolic blood pressure (SBP) after the supplementation (-4.21 mmHg, p<0.001). However, SBP increased in the placebo supplemented group. Diastolic blood pressure (DBP) nonsignificantly decreased in the inulin group. Inulin supplementation also increased BMI (p<0.001) but reduced BFP (p=0.288). Furthermore, confounding variables, such as BMI, baseline fasting glucose, age, menopause status, vomiting, constipation, and chronic medication did not have a statistical influence over the inulin effect on SBP. Conclusion Inulin supplementation reduces SBP and prevents increases in DBP in women with breast cancer. This could be an innovative nutraceutical approach to prevent hypertension present in women with this type of cancer at an early stage and may improve the quality of life of the patients and their prognostic development through chemotherapy. Trial Registration Number This trial is registered with ACTRN12616001532493.
Collapse
|
57
|
Rastelli M, Cani PD, Knauf C. The Gut Microbiome Influences Host Endocrine Functions. Endocr Rev 2019; 40:1271-1284. [PMID: 31081896 DOI: 10.1210/er.2018-00280] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
The gut microbiome is considered an organ contributing to the regulation of host metabolism. Since the relationship between the gut microbiome and specific diseases was elucidated, numerous studies have deciphered molecular mechanisms explaining how gut bacteria interact with host cells and eventually shape metabolism. Both metagenomic and metabolomic analyses have contributed to the discovery of bacterial-derived metabolites acting on host cells. In this review, we examine the molecular mechanisms by which bacterial metabolites act as paracrine or endocrine factors, thereby regulating host metabolism. We highlight the impact of specific short-chain fatty acids on the secretion of gut peptides (i.e., glucagon-like peptide-1, peptide YY) and other metabolites produced from different amino acids and regulating inflammation, glucose metabolism, or energy homeostasis. We also discuss the role of gut microbes on the regulation of bioactive lipids that belong to the endocannabinoid system and specific neurotransmitters (e.g., γ-aminobutyric acid, serotonin, nitric oxide). Finally, we review the role of specific bacterial components (i.e., ClpB, Amuc_1100) also acting as endocrine factors and eventually controlling host metabolism. In conclusion, this review summarizes the recent state of the art, aiming at providing evidence that the gut microbiome influences host endocrine functions via several bacteria-derived metabolites.
Collapse
Affiliation(s)
- Marialetizia Rastelli
- Université Catholique de Louvain, UCLouvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Brussels, Belgium.,NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium
| | - Patrice D Cani
- Université Catholique de Louvain, UCLouvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Brussels, Belgium.,NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium.,Institut de Recherche en Santé Digestive et Nutrition (IRSD), Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier (UPS), Toulouse Cedex 3, France
| |
Collapse
|
58
|
Zhan J, Liang Y, Liu D, Ma X, Li P, Zhai W, Zhou Z, Wang P. Pectin reduces environmental pollutant-induced obesity in mice through regulating gut microbiota: A case study of p,p'-DDE. ENVIRONMENT INTERNATIONAL 2019; 130:104861. [PMID: 31195221 DOI: 10.1016/j.envint.2019.05.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/18/2019] [Accepted: 05/21/2019] [Indexed: 05/25/2023]
Abstract
BACKGROUND The prevalence of obesity has raised global concerns. Environmental pollutants are one of the main causes of obesity. Many studies have demonstrated that dietary fiber could reduce obesity induced by high-fat diets, but whether environmental pollutant-induced obesity can be reversed is still unknown. OBJECTIVES This study aimed to investigate the effects of pectin on obesity induced by a typical environmental pollutant p,p'-dichlorodiphenyldichloroethylene (p,p'-DDE) and explore the underlying mechanism by which pectin reversed p,p'-DDE-induced obesity. METHODS p,p'-DDE was used to induce obesity in C57BL/6J mice and pectin was supplied during and after cessation of p,p'-DDE exposure. Body and fat weight gain, plasma lipid profile and insulin resistance of mice were assessed. Gut microbiota composition and the levels of short-chain fatty acids (SCFAs) as well as the receptor proteins and hormones in the SCFAs-related signaling pathway were analyzed. Moreover, p,p'-DDE levels in various tissues of mice were detected. RESULTS Pectin supplementation reversed body and fat weight gain, dyslipidemia, hyperglycemia and insulin resistance in p,p'-DDE-exposed mice. Furthermore, pectin apparently altered the p,p'-DDE-induced microbial composition and then promoted the levels of SCFAs in colonic feces as well as the expression of G-protein coupled receptors and the concentration of hormone peptide YY (PYY) and glucagon like peptide-1 (GLP-1). Pectin treatment also significantly reduced p,p'-DDE accumulation in mice tissues during p,p'-DDE exposure but did not change p,p'-DDE metabolism after termination of p,p'-DDE exposure. CONCLUSIONS Pectin had a good effect on reducing p,p'-DDE-induced obesity through regulating gut microbiota and provided a potential strategy for the treatment of environmental pollutant-caused health problems.
Collapse
Affiliation(s)
- Jing Zhan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Yiran Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Donghui Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Xiaoran Ma
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Peize Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Wangjing Zhai
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Zhiqiang Zhou
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China
| | - Peng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Science, China Agricultural University, No. 2, West Yuanmingyuan Road, Beijing 100193, PR China.
| |
Collapse
|
59
|
Le Dréan G, Pocheron AL, Billard H, Grit I, Pagniez A, Parnet P, Chappuis E, Rolli-Derkinderen M, Michel C. Neonatal Consumption of Oligosaccharides Greatly Increases L-Cell Density without Significant Consequence for Adult Eating Behavior. Nutrients 2019; 11:nu11091967. [PMID: 31438620 PMCID: PMC6769936 DOI: 10.3390/nu11091967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
Oligosaccharides (OS) are commonly added to infant formulas, however, their physiological impact, particularly on adult health programming, is poorly described. In adult animals, OS modify microbiota and stimulate colonic fermentation and enteroendocrine cell (EEC) activity. Since neonatal changes in microbiota and/or EEC density could be long-lasting and EEC-derived peptides do regulate short-term food intake, we hypothesized that neonatal OS consumption could modulate early EECs, with possible consequences for adult eating behavior. Suckling rats were supplemented with fructo-oligosaccharides (FOS), beta-galacto-oligosaccharides/inulin (GOS/In) mix, alpha-galacto-oligosaccharides (αGOS) at 3.2 g/kg, or a control solution (CTL) between postnatal day (PND) 5 and 14/15. Pups were either sacrificed at PND14/15 or weaned at PND21 onto standard chow. The effects on both microbiota and EEC were characterized at PND14/15, and eating behavior at adulthood. Very early OS supplementation drastically impacted the intestinal environment, endocrine lineage proliferation/differentiation particularly in the ileum, and the density of GLP-1 cells and production of satiety-related peptides (GLP-1 and PYY) in the neonatal period. However, it failed to induce any significant lasting changes on intestinal microbiota, enteropeptide secretion or eating behavior later in life. Overall, the results did not demonstrate any OS programming effect on satiety peptides secreted by L-cells or on food consumption, an observation which is a reassuring outlook from a human perspective.
Collapse
Affiliation(s)
- Gwenola Le Dréan
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France.
- IMAD, F-44000 Nantes, France.
- CRNH-Ouest, F-44000 Nantes, France.
| | - Anne-Lise Pocheron
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| | - Hélène Billard
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| | - Isabelle Grit
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| | - Anthony Pagniez
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| | - Patricia Parnet
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| | - Eric Chappuis
- Olygose, parc Technologique des Rives de l'Oise, F 60280 Venette, France
| | - Malvyne Rolli-Derkinderen
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
- Nantes Université, INSERM, UMR 1235, TENS, F-44000 Nantes, France
| | - Catherine Michel
- Nantes Université, INRA, UMR1280, PhAN, F-44000 Nantes, France
- IMAD, F-44000 Nantes, France
- CRNH-Ouest, F-44000 Nantes, France
| |
Collapse
|
60
|
Connection between gut microbiome and the development of obesity. Eur J Clin Microbiol Infect Dis 2019; 38:1987-1998. [PMID: 31367997 DOI: 10.1007/s10096-019-03623-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023]
Abstract
The potential role of the gut microbiota in various human diseases has attracted considerable attention worldwide. Here, we discuss the vital role of the intestinal microbiota in the development of obesity. First, we describe how the gut microbiota promotes fat accumulation. Additionally, a high-fat diet leads to structural instability among in the gut microbiota, further leading to an increase in endotoxins, which aggravates obesity. We then discuss how gut microbiota metabolites, including short-chain fatty acids and lipopolysaccharides, affect the host. Finally, we review several strategies for regulating the intestinal flora.
Collapse
|
61
|
Byrne CS, Chambers ES, Preston T, Tedford C, Brignardello J, Garcia-Perez I, Holmes E, Wallis GA, Morrison DJ, Frost GS. Effects of Inulin Propionate Ester Incorporated into Palatable Food Products on Appetite and Resting Energy Expenditure: A Randomised Crossover Study. Nutrients 2019; 11:nu11040861. [PMID: 30995824 PMCID: PMC6520886 DOI: 10.3390/nu11040861] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
Supplementation with inulin-propionate ester (IPE), which delivers propionate to the colon, suppresses ad libitum energy intake and stimulates the release of satiety hormones acutely in humans, and prevents weight gain. In order to determine whether IPE remains effective when incorporated into food products (FP), IPE needs to be added to a widely accepted food system. A bread roll and fruit smoothie were produced. Twenty-one healthy overweight and obese humans participated. Participants attended an acclimatisation visit and a control visit where they consumed un-supplemented food products (FP). Participants then consumed supplemented-FP, containing 10 g/d inulin or IPE for six days followed by a post-supplementation visit in a randomised crossover design. On study visits, supplemented-FP were consumed for the seventh time and ad libitum energy intake was assessed 420 min later. Blood samples were collected to assess hormones and metabolites. Resting energy expenditure (REE) was measured using indirect calorimetry. Taste and appearance ratings were similar between FP. Ad libitum energy intake was significantly different between treatments, due to a decreased intake following IPE-FP. These observations were not related to changes in blood hormones and metabolites. There was an increase in REE following IPE-FP. However, this effect was lost after correcting for changes in fat free mass. Our results suggest that IPE suppresses appetite and may alter REE following its incorporation into palatable food products.
Collapse
Affiliation(s)
- Claire S Byrne
- Section for Nutrition Research, Division of Diabetes, Endocrinology and Metabolism, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| | - Edward S Chambers
- Section for Nutrition Research, Division of Diabetes, Endocrinology and Metabolism, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| | - Tom Preston
- Stable Isotope Biochemistry Laboratory, Scottish Universities Environmental Research Centre, University of Glasgow, East Kilbride, Glasgow G75 0QF, Scotland.
| | - Catriona Tedford
- School of Computing, Engineering and Physical Sciences, University of the West of Scotland, Paisley Campus, Paisley PA1 2BE, Scotland.
| | - Jerusa Brignardello
- Department of Surgery and Cancer, Computational and Systems Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Isabel Garcia-Perez
- Department of Surgery and Cancer, Computational and Systems Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Elaine Holmes
- Department of Surgery and Cancer, Computational and Systems Medicine, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
- Institute of Health Futures, Murdoch University, South Street, Western Australia 6150, Australia.
| | - Gareth A Wallis
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Douglas J Morrison
- Stable Isotope Biochemistry Laboratory, Scottish Universities Environmental Research Centre, University of Glasgow, East Kilbride, Glasgow G75 0QF, Scotland.
| | - Gary S Frost
- Section for Nutrition Research, Division of Diabetes, Endocrinology and Metabolism, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London W12 0NN, UK.
| |
Collapse
|
62
|
Santos-Marcos JA, Perez-Jimenez F, Camargo A. The role of diet and intestinal microbiota in the development of metabolic syndrome. J Nutr Biochem 2019; 70:1-27. [PMID: 31082615 DOI: 10.1016/j.jnutbio.2019.03.017] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/28/2019] [Accepted: 03/25/2019] [Indexed: 02/09/2023]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic factors that increase the risk of cardiovascular disease and type 2 diabetes mellitus (T2DM), which is in itself a major cardiovascular disease risk factor. The aim of this review is to summarize the data related to the influence of the gut microbiota on the development of obesity and the MetS, highlighting the role of diet in controlling the MetS by modifying the gut microbiota. The main alterations in the gut microbiota of individuals with MetS consist of an increased Firmicutes/Bacteriodetes ratio and a reduced capacity to degrade carbohydrates to short-chain fatty acids, which in turn is related with the metabolic dysfunction of the host organism rather than with obesity itself. In addition to a low-fat, high-carbohydrate diet, with its high fiber intake, a diet with 30% fat content but with a high content in fruit and vegetables, such as the Mediterranean diet, is beneficial and partially restores the dysbiosis found in individuals with MetS. Overall, the shaping of the gut microbiota through the administration of prebiotics or probiotics increases the short-chain fatty acid production and is therefore a valid alternative in MetS treatment.
Collapse
Affiliation(s)
- Jose A Santos-Marcos
- Lipids and Atherosclerosis Research Unit, GC9 Nutrigenomic-Metabolic Syndrome, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Cordoba, Spain
| | - Francisco Perez-Jimenez
- Lipids and Atherosclerosis Research Unit, GC9 Nutrigenomic-Metabolic Syndrome, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Cordoba, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Research Unit, GC9 Nutrigenomic-Metabolic Syndrome, Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain; CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Cordoba, Spain.
| |
Collapse
|
63
|
Kyriachenko Y, Falalyeyeva T, Korotkyi O, Molochek N, Kobyliak N. Crosstalk between gut microbiota and antidiabetic drug action. World J Diabetes 2019; 10:154-168. [PMID: 30891151 PMCID: PMC6422856 DOI: 10.4239/wjd.v10.i3.154] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2D) is a disorder characterized by chronic inflated blood glucose levels (hyperglycemia), at first due to insulin resistance and unregulated insulin secretion but with tendency towards global spreading. The gut microbiota is recognized to have an influence on T2D, although surveys have not formed a clear overview to date. Because of the interactions between gut microbiota and host homeostasis, intestinal bacteria are believed to play a large role in various diseases, including metabolic syndrome, obesity and associated disease. In this review, we highlight the animal and human studies which have elucidated the roles of metformin, α-glucosidase inhibitors, glucagon-like peptide-1 agonists, peroxisome proliferator-activated receptors γ agonists, inhibitors of dipeptidyl peptidase-4, sodium/glucose cotransporter inhibitors, and other less studied medications on gut microbiota. This review is dedicated to one of the most widespread diseases, T2D, and the currently used antidiabetic drugs and most promising new findings. In general, the gut microbiota has been shown to have an influence on host metabolism, food consumption, satiety, glucose homoeostasis, and weight gain. Altered intestinal microbiota composition has been noticed in cardiovascular diseases, colon cancer, rheumatoid arthritis, T2D, and obesity. Therefore, the main effect of antidiabetic drugs is on the microbiome composition, basically increasing the short-chain fatty acids-producing bacteria, responsible for losing weight and suppressing inflammation.
Collapse
Affiliation(s)
- Yevheniia Kyriachenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Tetyana Falalyeyeva
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Oleksandr Korotkyi
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Nataliia Molochek
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, Kyiv 01601, Ukraine
| |
Collapse
|
64
|
Delzenne NM, Olivares M, Neyrinck AM, Beaumont M, Kjølbæk L, Larsen TM, Benítez-Páez A, Romaní-Pérez M, Garcia-Campayo V, Bosscher D, Sanz Y, van der Kamp JW. Nutritional interest of dietary fiber and prebiotics in obesity: Lessons from the MyNewGut consortium. Clin Nutr 2019; 39:414-424. [PMID: 30904186 DOI: 10.1016/j.clnu.2019.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 02/24/2019] [Accepted: 03/04/2019] [Indexed: 01/31/2023]
Abstract
The aim of EU project MyNewGut is to contribute to future public health-related recommendations supported by new insight in gut microbiome and nutrition-host relationship. In this Opinion Paper, we first revisit the concept of dietary fiber, taking into account their interaction with the gut microbiota. This paper also summarizes the main effects of dietary fibers with prebiotic properties in intervention studies in humans, with a particular emphasis on the effects of arabinoxylans and arabinoxylo-oligosaccharides on metabolic alterations associated with obesity. Based on the existing state of the art and future development, we elaborate the steps required to propose dietary guidelines related to dietary fibers, taking into account their interaction with the gut microbiota.
Collapse
Affiliation(s)
- Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium.
| | - Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Martin Beaumont
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Louise Kjølbæk
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 30, 1958, Frederiksberg C, Denmark
| | - Thomas Meinert Larsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Rolighedsvej 30, 1958, Frederiksberg C, Denmark
| | - Alfonso Benítez-Páez
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Marina Romaní-Pérez
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | | | | | - Yolanda Sanz
- Microbial Ecology, Nutrition & Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | | |
Collapse
|
65
|
Xing YW, Lei GT, Wu QH, Jiang Y, Huang MX. Procyanidin B2 protects against diet-induced obesity and non-alcoholic fatty liver disease via the modulation of the gut microbiota in rabbits. World J Gastroenterol 2019; 25:955-966. [PMID: 30833801 PMCID: PMC6397725 DOI: 10.3748/wjg.v25.i8.955] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Procyanidins have beneficial effects on metabolic syndrome and antimicrobial activity, but the mechanisms underlying these effects are unclear.
AIM To investigate the effects of procyanidin B2 (PB2) on non-alcoholic fatty liver disease and to explore the possible mechanism.
METHODS Thirty male New Zealand white rabbits were randomized into three groups. All of them were fed either a high-fat-cholesterol diet (HCD) or chow diet. HCD-fed rabbits were treated with vehicle or PB2 daily for 12 wk. Body weight and food intake were evaluated once a week. Serum biomarkers, such as total cholesterols, triglycerides, and aspartate transaminase, were detected. All rabbits were sacrificed and histological parameters of liver were assessed by hematoxylin and eosin-stained sections. Moreover, several lipogenic genes and gut microbiota (by 16S rRNA sequencing) were investigated to explore the possible mechanism.
RESULTS The HCD group had higher body weight, liver index, serum lipid profile, insulin resistance, serum glucose, and hepatic steatosis compared to the CHOW group. PB2 treatment prevented HCD-induced increases in body weight and hypertriglyceridemia in association with triglyceride accumulation in the liver. PB2 also ameliorated low-grade inflammation, which was reflected by serum lipopolysaccharides and improved insulin resistance. In rabbit liver, PB2 prevented the upregulation of steroid response element binding protein 1c and fatty acid synthase and the downregulation of carnitine palmitoyltransferase, compared to the HCD group. Moreover, HCD led to a decrease of Bacteroidetes in gut microbiota. PB2 significantly improved the proportions of Bacteroidetes at the phylum level and Akkermansia at the genus level.
CONCLUSION Our results indicate the possible mechanism of PB2 to improve HCD-induced features of metabolic syndrome and provide a new dietary supplement.
Collapse
Affiliation(s)
- Ya-Wei Xing
- Department of Gastroenterology, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Guang-Tao Lei
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Qing-Hua Wu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Yu Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Man-Xiang Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
| |
Collapse
|
66
|
Zhang Q, Xiao X, Zheng J, Li M, Yu M, Ping F, Wang T, Wang X. Influence of Maternal Inulin-Type Prebiotic Intervention on Glucose Metabolism and Gut Microbiota in the Offspring of C57BL Mice. Front Endocrinol (Lausanne) 2019; 10:675. [PMID: 31632351 PMCID: PMC6779716 DOI: 10.3389/fendo.2019.00675] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 09/17/2019] [Indexed: 01/14/2023] Open
Abstract
Scope: Maternal obesity leads to glucose intolerance in the offspring. Changes in the gut microbiota are being increasingly implicated in the pathogenesis of diabetes. We hypothesized that inulin intervention during gestation and lactation improves glucose metabolism disorders in mouse offspring from high-fat diet (HD)-fed dams. Procedures: Female C57BL mice were fed a control diet or HD for 4 weeks before mating. After mating, pregnant mice were randomly divided into three groups through gestation and lactation: control diet (CD) group, HD group, and HD treated with inulin (HD-inulin) group. At weaning, glucose metabolic status was assessed. Gut microbial DNA from offspring cecal contents was isolated and processed for metagenomic shotgun sequencing, and taxonomic and functional profiling were performed. Results: Offspring from dams in the HD-inulin groups demonstrated reduced fasting blood glucose, decreased blood glucose area under the curve during the oral glucose tolerance test, and reduced fasting serum insulin and HOMA-IR compared to offspring from dams in the HD group. Nineteen differentially abundant bacterial species were identified between the HD-inulin and HD groups. The HD-inulin group displayed significantly greater abundances of Bacteroides_acidifaciens, Eubacterium_sp_CAG_786, Clostridium_sp_CAG_343, and Bifidobacterium_breve species and lower abundances of Oscillibacter_sp_1_3, Ruminococcus_gnavus_CAG_126, and Bacteroides_massiliensis species. Differentially abundant bacterial species among the three groups were involved in 38 metabolic pathways, including several glucose and lipid metabolism pathways. Conclusion: Our results show that early inulin intervention in HD-fed mouse dams moderates offspring glucose metabolism and gut dysbiosis.
Collapse
|
67
|
Cani PD, Van Hul M, Lefort C, Depommier C, Rastelli M, Everard A. Microbial regulation of organismal energy homeostasis. Nat Metab 2019; 1:34-46. [PMID: 32694818 DOI: 10.1038/s42255-018-0017-4] [Citation(s) in RCA: 316] [Impact Index Per Article: 63.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/15/2018] [Indexed: 12/13/2022]
Abstract
The gut microbiome has emerged as a key regulator of host metabolism. Here we review the various mechanisms through which the gut microbiome influences the energy metabolism of its host, highlighting the complex interactions between gut microbes, their metabolites and host cells. Among the most important bacterial metabolites are short-chain fatty acids, which serve as a direct energy source for host cells, stimulate the production of gut hormones and act in the brain to regulate food intake. Other microbial metabolites affect systemic energy expenditure by influencing thermogenesis and adipose tissue browning. Both direct and indirect mechanisms of action are known for specific metabolites, such as bile acids, branched chain amino acids, indole propionic acid and endocannabinoids. We also discuss the roles of specific bacteria in the production of specific metabolites and explore how external factors, such as antibiotics and exercise, affect the microbiome and thereby energy homeostasis. Collectively, we present a large body of evidence supporting the concept that gut microbiota-based therapies can be used to modulate host metabolism, and we expect to see such approaches moving from bench to bedside in the near future.
Collapse
Affiliation(s)
- Patrice D Cani
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium.
| | - Matthias Van Hul
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Charlotte Lefort
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Clara Depommier
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Marialetizia Rastelli
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, WELBIO-Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
68
|
Covasa M, Stephens RW, Toderean R, Cobuz C. Intestinal Sensing by Gut Microbiota: Targeting Gut Peptides. Front Endocrinol (Lausanne) 2019; 10:82. [PMID: 30837951 PMCID: PMC6390476 DOI: 10.3389/fendo.2019.00082] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022] Open
Abstract
There are more than 2 billion overweight and obese individuals worldwide, surpassing for the first time, the number of people affected by undernutrition. Obesity and its comorbidities inflict a heavy burden on the global economies and have become a serious threat to individuals' wellbeing with no immediate cure available. The causes of obesity are manifold, involving several factors including physiological, metabolic, neural, psychosocial, economic, genetics and the environment, among others. Recent advances in genome sequencing and metagenomic profiling have added another dimension to this complexity by implicating the gut microbiota as an important player in energy regulation and the development of obesity. As such, accumulating evidence demonstrate the impact of the gut microbiota on body weight, adiposity, glucose, lipid metabolism, and metabolic syndrome. This also includes the role of microbiota as a modulatory signal either directly or through its bioactive metabolites on intestinal lumen by releasing chemosensing factors known to have a major role in controlling food intake and regulating body weight. The importance of gut signaling by microbiota signaling is further highlighted by the presence of taste and nutrient receptors on the intestinal epithelium activated by the microbial degradation products as well as their role in release of peptides hormones controlling appetite and energy homeostasis. This review present evidence on how gut microbiota interacts with intestinal chemosensing and modulates the release and activity of gut peptides, particularly GLP-1 and PYY.
Collapse
Affiliation(s)
- Mihai Covasa
- Department of Health and Human Development, University of Suceava, Suceava, Romania
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
- *Correspondence: Mihai Covasa
| | - Richard W. Stephens
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Roxana Toderean
- Department of Health and Human Development, University of Suceava, Suceava, Romania
| | - Claudiu Cobuz
- Department of Health and Human Development, University of Suceava, Suceava, Romania
| |
Collapse
|
69
|
Jangra S, K. RS, Sharma RK, Pothuraju R, Mohanty AK. Ameliorative effect of fermentable fibres on adiposity and insulin resistance in C57BL/6 mice fed a high-fat and sucrose diet. Food Funct 2019; 10:3696-3705. [DOI: 10.1039/c8fo02578a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The consumption of diets rich in fat and refined sugars is recognized to be one of the causes of lifestyle disorders, and dietary fibres are being advocated to ameliorate the complications associated with these disorders.
Collapse
Affiliation(s)
- Surender Jangra
- Division of Animal Biochemistry
- ICAR-National Dairy Research Institute
- Karnal
- India
| | - Raja Shekar K.
- Division of Animal Biochemistry
- ICAR-National Dairy Research Institute
- Karnal
- India
| | - Raj Kumar Sharma
- Division of Animal Biochemistry
- ICAR-National Dairy Research Institute
- Karnal
- India
| | - Ramesh Pothuraju
- Division of Animal Biochemistry
- ICAR-National Dairy Research Institute
- Karnal
- India
| | - A. K. Mohanty
- Animal Biotechnology Centre
- ICAR-National Dairy Research Institute
- Karnal
- India
| |
Collapse
|
70
|
Vallianou NG, Stratigou T, Tsagarakis S. Microbiome and diabetes: Where are we now? Diabetes Res Clin Pract 2018; 146:111-118. [PMID: 30342053 DOI: 10.1016/j.diabres.2018.10.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/23/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023]
Abstract
Alterations in the diversity or structure of gut microbiota known as dysbiosis, may affect metabolic activities, resulting in metabolic disorders, such as obesity and diabetes. The development of more sophisticated methods, such as metagenomics sequencing, PCR-denaturing gradient gel electrophoresis, microarrays and fluorescence in situ hybridization, has expanded our knowledge on gut microbiome. Dysbiosis has been related to increased plasma concentrations of gut microbiota-derived lipopolysaccharide (LPS), which triggers the production of a variety of cytokines and the recruitment of inflammatory cells. Metabolomics have demonstrated that butyrate and propionate suppress weight gain in mice with high fat diet-induced obesity, and acetate has been proven to reduce food intake in healthy mice. The role of prebiotics, probiotics, genetically modified bacteria and fecal microbiota transplantation, as potential therapeutic challenges for type 2 diabetes will be discussed in this review.
Collapse
Affiliation(s)
- Natalia G Vallianou
- Evangelismos General Hospital, Department of Endocrinology, Diabetes and Metabolism, Athens, Greece.
| | - Theodora Stratigou
- Evangelismos General Hospital, Department of Endocrinology, Diabetes and Metabolism, Athens, Greece
| | - Stylianos Tsagarakis
- Evangelismos General Hospital, Department of Endocrinology, Diabetes and Metabolism, Athens, Greece
| |
Collapse
|
71
|
Urrutia-Piñones J, Illanes-González J, López-Aguilera A, Julio-Pieper M, Bravo JA. Do Obese Bacteria Make us “Want them”? Intestinal Microbiota, Mesocorticolimbic Circuit and Non-Homeostatic Feeding. Curr Behav Neurosci Rep 2018. [DOI: 10.1007/s40473-018-0161-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
72
|
Cani PD, Jordan BF. Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer. Nat Rev Gastroenterol Hepatol 2018; 15:671-682. [PMID: 29844585 DOI: 10.1038/s41575-018-0025-6] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overweight and obesity are associated with increased risk of developing metabolic disorders such as diabetes and cardiovascular diseases. However, besides these metabolic diseases, excess body weight is also associated with different cancers, including gastrointestinal cancers, such as liver, pancreatic and colon cancers. Inflammation is a common feature of both obesity and cancer; however, the origin of this inflammation has been largely debated. Over the past decade, growing evidence has shown that the composition of the gut microbiota and its activity might be associated not only with the onset of inflammation but also with metabolic disorders and cancer. Here, we review the links between the gut microbiota, gut barrier function and the onset of low-grade inflammation in the development of gastrointestinal cancer. We also describe the mechanisms by which specific microorganism-associated molecular patterns crosstalk with the immune system and how the metabolic activity of bacteria induces specific signalling pathways beyond the gut that eventually trigger carcinogenesis.
Collapse
Affiliation(s)
- Patrice D Cani
- Université catholique de Louvain, Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life sciences and BIOtechnology), Metabolism and Nutrition Research Group, Brussels, Belgium.
| | - Benedicte F Jordan
- Université catholique de Louvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Brussels, Belgium
| |
Collapse
|
73
|
Honoré SM, Grande MV, Gomez Rojas J, Sánchez SS. Smallanthus sonchifolius (Yacon) Flour Improves Visceral Adiposity and Metabolic Parameters in High-Fat-Diet-Fed Rats. J Obes 2018; 2018:5341384. [PMID: 30510798 PMCID: PMC6230400 DOI: 10.1155/2018/5341384] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/31/2018] [Accepted: 09/26/2018] [Indexed: 11/29/2022] Open
Abstract
Smallanthus sonchifolius (yacon), a native plant of South America, was observed to improve lipid profile in rodents and humans. This study aimed to investigate the antiobesity properties of yacon roots in a high-fat-diet (HFD) model and the underlying mechanisms. A total of 30 Wistar male rats were divided into five groups (n=6): the standard chow diet (SD) group was fed a SD; the HFD group was fed a HFD; and the HFD Y340 and HFD Y680 groups were fed a HFD plus yacon flour (340 and 680 mg FOS/kg b. w./day, respectively). HFD Y340 and HFD Y680 rats exhibited marked attenuation of weight gain, a decrease in visceral fat pad weight, a restoration of the serum lipid profile and atherogenic index in a dose-dependent manner, being the higher dose more effective (p < 0.05). In addition, we found that HFD Y680 rats showed lower glucose and insulin levels, improved glucose tolerance, and insulin sensitivity (p < 0.5). A downregulation of several adipocyte specific-transcription factors, including peroxisome proliferator-activated receptor gamma2 (PPAR-γ2), CCAAT/enhancer binding protein a (C/EBP-a) and activating protein (aP2) mRNA levels, was determined in the visceral adipose tissue of HFD Y680 rats (p < 0.05). An improvement of adipokine profile in HFD Y680 rats and decreased serum proinflammatory cytokine levels (p < 0.05) were determined by ELISA. Decreased macrophage infiltration and F4/80 and MCP-1 expression in the visceral adipose tissue of HFD Y680 rats (p < 0.5), together with a higher pAkt/Akt expression (p < 0.05) were also observed by immunofluorescence and immunoblotting. A significant increase in glucagon (Gcg) and PYY mRNA levels in distal ileum of HFD Y680 rats (p < 0.05) were also detected. In the second approach, we determined that yacon supplementation potentiates the effects of the HFD reversion to a standard diet. In conclusion, yacon showed antiobesity properties by inhibiting adipogenesis and improving the visceral adipose tissue function.
Collapse
Affiliation(s)
- Stella Maris Honoré
- Instituto Superior de Investigaciones Biológicas (INSIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad Nacional de Tucumán (CONICET-UNT), Chacabuco 461, T4000ILI San Miguel de Tucumán, Argentina
| | - Maria Virginia Grande
- Instituto Superior de Investigaciones Biológicas (INSIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad Nacional de Tucumán (CONICET-UNT), Chacabuco 461, T4000ILI San Miguel de Tucumán, Argentina
| | - Jorge Gomez Rojas
- Instituto Superior de Investigaciones Biológicas (INSIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad Nacional de Tucumán (CONICET-UNT), Chacabuco 461, T4000ILI San Miguel de Tucumán, Argentina
| | - Sara Serafina Sánchez
- Instituto Superior de Investigaciones Biológicas (INSIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas-Universidad Nacional de Tucumán (CONICET-UNT), Chacabuco 461, T4000ILI San Miguel de Tucumán, Argentina
| |
Collapse
|
74
|
Chianese R, Coccurello R, Viggiano A, Scafuro M, Fiore M, Coppola G, Operto FF, Fasano S, Laye S, Pierantoni R, Meccariello R. Impact of Dietary Fats on Brain Functions. Curr Neuropharmacol 2018; 16:1059-1085. [PMID: 29046155 PMCID: PMC6120115 DOI: 10.2174/1570159x15666171017102547] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 08/24/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Adequate dietary intake and nutritional status have important effects on brain functions and on brain health. Energy intake and specific nutrients excess or deficiency from diet differently affect cognitive processes, emotions, behaviour, neuroendocrine functions and synaptic plasticity with possible protective or detrimental effects on neuronal physiology. Lipids, in particular, play structural and functional roles in neurons. Here the importance of dietary fats and the need to understand the brain mechanisms activated by peripheral and central metabolic sensors. Thus, the manipulation of lifestyle factors such as dietary interventions may represent a successful therapeutic approach to maintain and preserve brain health along lifespan. METHODS This review aims at summarizing the impact of dietary fats on brain functions. RESULTS Starting from fat consumption, nutrient sensing and food-related reward, the impact of gut-brain communications will be discussed in brain health and disease. A specific focus will be on the impact of fats on the molecular pathways within the hypothalamus involved in the control of reproduction via the expression and the release of Gonadotropin-Releasing Hormone. Lastly, the effects of specific lipid classes such as polyunsaturated fatty acids and of the "fattest" of all diets, commonly known as "ketogenic diets", on brain functions will also be discussed. CONCLUSION Despite the knowledge of the molecular mechanisms is still a work in progress, the clinical relevance of the manipulation of dietary fats is well acknowledged and such manipulations are in fact currently in use for the treatment of brain diseases.
Collapse
Affiliation(s)
- Rosanna Chianese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Roberto Coccurello
- Institute of Cell Biology and Neurobiology, National Research Council (C.N.R.), Rome, Italy.,Fondazione S. Lucia (FSL) IRCCS, Roma, Italy
| | - Andrea Viggiano
- Department of Medicine, Surgery and Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy
| | - Marika Scafuro
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marco Fiore
- Institute of Cell Biology and Neurobiology, National Research Council (C.N.R.), Rome, Italy.,Fondazione S. Lucia (FSL) IRCCS, Roma, Italy
| | - Giangennaro Coppola
- Department of Medicine, Surgery and Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy.,UO Child and Adolescent Neuropsychiatry, Medical School, University of Salerno, Salerno, Italy
| | | | - Silvia Fasano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Sophie Laye
- INRA, Bordeaux University, Nutrition and Integrative Neurobiology, UMR, Bordeaux, France
| | - Riccardo Pierantoni
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosaria Meccariello
- Department of Movement and Wellness Sciences, Parthenope University of Naples, Naples, Italy
| |
Collapse
|
75
|
Vannan DT, Bomhof MR, Reimer RA. Comparison of Glucose and Satiety Hormone Response to Oral Glucose vs. Two Mixed-Nutrient Meals in Rats. Front Nutr 2018; 5:89. [PMID: 30320120 PMCID: PMC6168634 DOI: 10.3389/fnut.2018.00089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/07/2018] [Indexed: 11/30/2022] Open
Abstract
The obesity epidemic is driving interest in identifying strategies that enhance appetite control by altering the secretion of hormones that regulate satiety and food intake. An appropriate nutrient stimulus, such as a meal or oral nutrient solution, is needed to elicit the secretion of satiety hormones in order to evaluate the impact of dietary and other interventions. Our objective was to compare the effects of oral glucose vs. mixed nutrients on plasma concentrations of glucose and appetite-regulating hormones to determine the most appropriate oral nutrient challenge to trigger robust hormone secretion. A 120 min oral glucose tolerance test (OGTT) was compared with two meal tolerance tests (MTT) of differing formulation to evaluate glucose and satiety hormone responses. Following overnight feed deprivation, male Sprague-Dawley rats were given one of three oral gavages with equal carbohydrate content (2 g CHO/kg) in the form of: (1) Dextrose, (2) Ensure®, or (3) Mixed Meal. A fourth group was given saline as a control. Blood was collected via tail snip and analyzed for glucose, insulin, GLP-1, GIP, PYY, amylin, leptin, and ghrelin. Dextrose resulted in the highest blood glucose at T15 (P = 0.014), while the mixed meal was significantly higher than saline from T30-T120 (P < 0.05). Insulin was higher at T15 with dextrose compared to saline (P = 0.031) and Ensure® (P = 0.033). GLP-1 tAUC was significantly higher with dextrose compared to mixed meal (P = 0.04) while GIP tAUC was higher with dextrose and mixed meal compared to saline (P < 0.05). Changes in tAUC for insulin, amylin, leptin, ghrelin, and PYY did not reach significance. Based on these findings, dextrose appears to provide a robust acute glycemic and hormone response and is therefore likely an appropriate oral solution to reproducibly test the impact of various dietary, surgical, or pharmacological interventions on glucose and satiety hormone response.
Collapse
Affiliation(s)
| | - Marc R Bomhof
- Department of Kinesiology and Physical Education, University of Lethbridge, Lethbridge, AB, Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
76
|
Belobrajdic DP, Jenkins CLD, Christophersen CT, Bird AR. Cereal fructan extracts alter intestinal fermentation to reduce adiposity and increase mineral retention compared to oligofructose. Eur J Nutr 2018; 58:2811-2821. [PMID: 30284066 DOI: 10.1007/s00394-018-1830-y] [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: 06/04/2018] [Accepted: 09/19/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE Intestinal fermentation of inulin-type fructans, including oligofructose, can modulate adiposity, improve energy regulation, and increase mineral absorption. We aimed to determine whether cereal fructans had greater effects on reducing adiposity and improving mineral absorption compared with oligofructose. METHODS Thirty-two male Sprague-Dawley rats were randomly assigned to one of four dietary treatments that contained 0% fructan (control), or 5% fructan provided by oligofructose (OF), a barley grain fraction (BGF), or a wheat stem fraction (WSF). After 1 week on the diets, mineral absorption and retention was assessed. At 4 weeks, blood samples were collected for gut hormone analysis, adipose depots were removed and weighed, and caecal digesta was analyzed for pH and short-chain fatty acids (SCFA). RESULTS The BGF and WSF, but not OF, had lower total visceral fat weights than the Control (p < 0.05). The fructan diets all lowered caecal pH and raised caecal digesta weight and total SCFA content, in comparison to the Control. Caecal propionate levels for OF were similar to the Control and higher for WSF (p < 0.05). Plasma peptide YY and glucagon-like peptide-1 levels were elevated for all fructan groups when compared to Control (p < 0.001) and gastric inhibitory peptide was lower for the WSF compared to the other groups (p < 0.05). The fructan diets improved calcium and magnesium retention, which was highest for WSF (p < 0.05). BGF and WSF in comparison to OF showed differential effects on fermentation, gut hormone levels, and adiposity. CONCLUSIONS Cereal fructan sources have favorable metabolic effects that suggest greater improvements in energy regulation and mineral status to those reported for oligofructose.
Collapse
Affiliation(s)
| | | | | | - Anthony R Bird
- CSIRO Health and Biosecurity, PO Box 10041, Adelaide, BC, 5000, Australia
| |
Collapse
|
77
|
Cussotto S, Sandhu KV, Dinan TG, Cryan JF. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Front Neuroendocrinol 2018; 51:80-101. [PMID: 29753796 DOI: 10.1016/j.yfrne.2018.04.002] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
The human gut harbours trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. These intestinal microbes are also key components of the gut-brain axis, the bidirectional communication pathway between the gut and the central nervous system (CNS). In addition, the CNS is closely interconnected with the endocrine system to regulate many physiological processes. An expanding body of evidence is supporting the notion that gut microbiota modifications and/or manipulations may also play a crucial role in the manifestation of specific behavioural responses regulated by neuroendocrine pathways. In this review, we will focus on how the intestinal microorganisms interact with elements of the host neuroendocrine system to modify behaviours relevant to stress, eating behaviour, sexual behaviour, social behaviour, cognition and addiction.
Collapse
Affiliation(s)
- Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kiran V Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
78
|
Drapkina OM, Korneeva ON. [Gut microbiota and obesity: Pathogenetic relationships and ways to normalize the intestinal microflora]. TERAPEVT ARKH 2018. [PMID: 28635818 DOI: 10.17116/terarkh2016889135-142] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The review demonstrates mechanisms in the relationship of obesity to gut microbiota, as well as possible therapeutic measures to normalize the intestinal microflora. There is evidence that the latter makes a great contribution to the pathogenesis of obesity and related diseases. Investigations have shown the role of the nature of consumed foods (fatty foods) in reducing the amount of bifidobacteria and lactobacilli, as well as the effects of bacterial lipopolysaccharides and metabolites from the intestinal microflora (trimethylamine-N-oxide, bile acids, etc.). The use of prebiotics, probiotics and ursodeoxycholic acid preparations and fecal transplantation are promising in correcting the microflora and in providing their positive effect on metabolic disturbances. Certain probiotic strains are effective in treating dyslipidemia, diabetes mellitus, obesity, and metabolic syndrome. Gut microbiota is impaired in obesity and contributes to the development of cardiovascular diseases. The control of the gut microbiota and the use of drugs altering the composition of the microflora may become a novel approach to reducing the risk of cardiovascular diseases.
Collapse
Affiliation(s)
- O M Drapkina
- National Research Center for Preventive Medicine, Ministry of Health of Russia, Moscow, Russia
| | - O N Korneeva
- Art-Med Therapeutic and Diagnostic Center, Moscow, Russia
| |
Collapse
|
79
|
Olivares M, Schüppel V, Hassan AM, Beaumont M, Neyrinck AM, Bindels LB, Benítez-Páez A, Sanz Y, Haller D, Holzer P, Delzenne NM. The Potential Role of the Dipeptidyl Peptidase-4-Like Activity From the Gut Microbiota on the Host Health. Front Microbiol 2018; 9:1900. [PMID: 30186247 PMCID: PMC6113382 DOI: 10.3389/fmicb.2018.01900] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/27/2018] [Indexed: 12/16/2022] Open
Abstract
The Dipeptidyl peptidase-4 (DPP-4) activity influences metabolic, behavioral and intestinal disorders through the cleavage of key hormones and peptides. Some studies describe the existence of human DPP-4 homologs in commensal bacteria, for instance in Prevotella or Lactobacillus. However, the role of the gut microbiota as a source of DPP-4-like activity has never been investigated. Through the comparison of the DPP-4 activity in the cecal content of germ-free mice (GFM) and gnotobiotic mice colonized with the gut microbiota of a healthy subject, we bring the proof of concept that a significant DPP-4-like activity occurs in the microbiota. By analyzing the existing literature, we propose that DPP-4-like activity encoded by the intestinal microbiome could constitute a novel mechanism to modulate protein digestion as well as host metabolism and behavior.
Collapse
Affiliation(s)
- Marta Olivares
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Valentina Schüppel
- ZIEL Institute for Food and Health, Technical University of Munich, Freising-Weihenstephan, Germany.,Chair of Nutrition and Immunology, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Ahmed M Hassan
- Research Unit of Translational Neurogastroenterology, Pharmacology Section, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Martin Beaumont
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Alfonso Benítez-Páez
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Dirk Haller
- ZIEL Institute for Food and Health, Technical University of Munich, Freising-Weihenstephan, Germany.,Chair of Nutrition and Immunology, Technical University of Munich, Freising-Weihenstephan, Germany
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Pharmacology Section, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
80
|
Diet, gut microbiota composition and feeding behavior. Physiol Behav 2018; 192:177-181. [DOI: 10.1016/j.physbeh.2018.03.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 03/23/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
|
81
|
Bidu C, Escoula Q, Bellenger S, Spor A, Galan M, Geissler A, Bouchot A, Dardevet D, Morio B, Cani PD, Lagrost L, Narce M, Bellenger J. The Transplantation of ω3 PUFA-Altered Gut Microbiota of fat-1 Mice to Wild-Type Littermates Prevents Obesity and Associated Metabolic Disorders. Diabetes 2018; 67:1512-1523. [PMID: 29793999 DOI: 10.2337/db17-1488] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 04/21/2018] [Indexed: 11/13/2022]
Abstract
Altering the gut microbiome may be beneficial to the host and recently arose as a promising strategy to manage obesity. Here, we investigated the relative contribution of ω3 polyunsaturated fatty acid (PUFA)-mediated alterations in the microbiota to metabolic parameter changes in mice. Four groups were compared: male fat-1 transgenic mice (with constitutive production of ω3 PUFAs) and male wild-type (WT) littermates fed an obesogenic (high fat/high sucrose [HFHS]) or a control diet. Unlike WT mice, HFHS-fed fat-1 mice were protected against obesity, glucose intolerance, and hepatic steatosis. Unlike WT mice, fat-1 mice maintained a normal barrier function, resulting in a significantly lower metabolic endotoxemia. The fat-1 mice displayed greater phylogenic diversity in the cecum, and fecal microbiota transplantation from fat-1 to WT mice was able to reverse weight gain and to normalize glucose tolerance and intestinal permeability. We concluded that the ω3 PUFA-mediated alteration of gut microbiota contributed to the prevention of metabolic syndrome in fat-1 mice. It occurred independently of changes in the PUFA content of host tissues and may represent a promising strategy to prevent metabolic disease and preserve a lean phenotype.
Collapse
Affiliation(s)
- Célia Bidu
- University of Bourgogne Franche-Comté, L'Unité de Formation Sciences de la Vie, de la Terre et de l'Environnement, Lipides Nutrition Cancer UMR1231, Dijon, France
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Quentin Escoula
- University of Bourgogne Franche-Comté, L'Unité de Formation Sciences de la Vie, de la Terre et de l'Environnement, Lipides Nutrition Cancer UMR1231, Dijon, France
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Sandrine Bellenger
- University of Bourgogne Franche-Comté, L'Unité de Formation Sciences de la Vie, de la Terre et de l'Environnement, Lipides Nutrition Cancer UMR1231, Dijon, France
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Aymé Spor
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1347, Agroécologie, Dijon, France
| | - Maxime Galan
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1062 Centre de Biologie pour la Gestion des Populations (Institut National de la Recherche Agronomique, L'Institut de Recherche pour le Développement, Centre de coopération Internationale en Recherche Agronomique pour le Développement, Montpellier SupAgro), Montferrier-sur-Lez, France
| | - Audrey Geissler
- CellImap-Cellular Imaging Platform, Faculté de Médecine et Pharmacie, Dijon, France
| | - André Bouchot
- CellImap-Cellular Imaging Platform, Faculté de Médecine et Pharmacie, Dijon, France
| | - Dominique Dardevet
- Clermont Université, Université d'Auvergne, Unité de Nutrition Humaine, Clermont-Ferrand, France
- Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1019, Unité de Nutrition Humaine, Centre de Recherche en Nutrition Humaine Auvergne, Clermont-Ferrand, France
| | - Béatrice Morio
- Institut National de la Recherche Agronomique , Unité Mixte de Recherche 1397, CarMeN Laboratory, Lyon 1 University, INSERM U1060, Institut National des Sciences Appliquées of Lyon, Rockefeller and Charles Merieux Lyon-Sud Medical Universities, Lyon, France
| | - Patrice D Cani
- Université Catholique de Louvain, Welbio (Walloon Excellence in Life Sciences and BIOtechnology), Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Brussels, Belgium
| | - Laurent Lagrost
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
- L'Unité de Formation Médecine, Université de Bourgogne, Dijon, France
| | - Michel Narce
- University of Bourgogne Franche-Comté, L'Unité de Formation Sciences de la Vie, de la Terre et de l'Environnement, Lipides Nutrition Cancer UMR1231, Dijon, France
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| | - Jérôme Bellenger
- University of Bourgogne Franche-Comté, L'Unité de Formation Sciences de la Vie, de la Terre et de l'Environnement, Lipides Nutrition Cancer UMR1231, Dijon, France
- INSERM, Lipides Nutrition Cancer UMR1231, Dijon, France
- LipSTIC LabEx, Fondation de Coopération Scientifique Bourgogne-Franche Comté, Dijon, France
| |
Collapse
|
82
|
Bliss ES, Whiteside E. The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol 2018; 9:900. [PMID: 30050464 PMCID: PMC6052131 DOI: 10.3389/fphys.2018.00900] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic, placing socioeconomic strain on public healthcare systems, especially within the so-called Western countries, such as Australia, United States, United Kingdom, and Canada. Obesity results from an imbalance between energy intake and energy expenditure, where energy intake exceeds expenditure. Current non-invasive treatments lack efficacy in combating obesity, suggesting that obesity is a multi-faceted and more complex disease than previously thought. This has led to an increase in research exploring energy homeostasis and the discovery of a complex bidirectional communication axis referred to as the gut-brain axis. The gut-brain axis is comprised of various neurohumoral components that allow the gut and brain to communicate with each other. Communication occurs within the axis via local, paracrine and/or endocrine mechanisms involving a variety of gut-derived peptides produced from enteroendocrine cells (EECs), including glucagon-like peptide 1 (GLP1), cholecystokinin (CCK), peptide YY3-36 (PYY), pancreatic polypeptide (PP), and oxyntomodulin. Neural networks, such as the enteric nervous system (ENS) and vagus nerve also convey information within the gut-brain axis. Emerging evidence suggests the human gut microbiota, a complex ecosystem residing in the gastrointestinal tract (GIT), may influence weight-gain through several inter-dependent pathways including energy harvesting, short-chain fatty-acids (SCFA) signalling, behaviour modifications, controlling satiety and modulating inflammatory responses within the host. Hence, the gut-brain axis, the microbiota and the link between these elements and the role each plays in either promoting or regulating energy and thereby contributing to obesity will be explored in this review.
Collapse
Affiliation(s)
- Edward S. Bliss
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | | |
Collapse
|
83
|
Li Z, Yi CX, Katiraei S, Kooijman S, Zhou E, Chung CK, Gao Y, van den Heuvel JK, Meijer OC, Berbée JFP, Heijink M, Giera M, Willems van Dijk K, Groen AK, Rensen PCN, Wang Y. Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit. Gut 2018; 67:1269-1279. [PMID: 29101261 DOI: 10.1136/gutjnl-2017-314050] [Citation(s) in RCA: 384] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Butyrate exerts metabolic benefits in mice and humans, the underlying mechanisms being still unclear. We aimed to investigate the effect of butyrate on appetite and energy expenditure, and to what extent these two components contribute to the beneficial metabolic effects of butyrate. DESIGN Acute effects of butyrate on appetite and its method of action were investigated in mice following an intragastric gavage or intravenous injection of butyrate. To study the contribution of satiety to the metabolic benefits of butyrate, mice were fed a high-fat diet with butyrate, and an additional pair-fed group was included. Mechanistic involvement of the gut-brain neural circuit was investigated in vagotomised mice. RESULTS Acute oral, but not intravenous, butyrate administration decreased food intake, suppressed the activity of orexigenic neurons that express neuropeptide Y in the hypothalamus, and decreased neuronal activity within the nucleus tractus solitarius and dorsal vagal complex in the brainstem. Chronic butyrate supplementation prevented diet-induced obesity, hyperinsulinaemia, hypertriglyceridaemia and hepatic steatosis, largely attributed to a reduction in food intake. Butyrate also modestly promoted fat oxidation and activated brown adipose tissue (BAT), evident from increased utilisation of plasma triglyceride-derived fatty acids. This effect was not due to the reduced food intake, but explained by an increased sympathetic outflow to BAT. Subdiaphragmatic vagotomy abolished the effects of butyrate on food intake as well as the stimulation of metabolic activity in BAT. CONCLUSION Butyrate acts on the gut-brain neural circuit to improve energy metabolism via reducing energy intake and enhancing fat oxidation by activating BAT.
Collapse
Affiliation(s)
- Zhuang Li
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Saeed Katiraei
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander Kooijman
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Enchen Zhou
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Chih Kit Chung
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yuanqing Gao
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - José K van den Heuvel
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Jimmy F P Berbée
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke Heijink
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Albert K Groen
- Department of Vascular Medicine, Amsterdam Diabetes Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, University of Groningen, Groningen, The Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Yanan Wang
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Pediatrics, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
84
|
Brubaker PL. Glucagon‐like Peptide‐2 and the Regulation of Intestinal Growth and Function. Compr Physiol 2018; 8:1185-1210. [DOI: 10.1002/cphy.c170055] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
85
|
Wang X, Wang X, Jiang H, Cai C, Li G, Hao J, Yu G. Marine polysaccharides attenuate metabolic syndrome by fermentation products and altering gut microbiota: An overview. Carbohydr Polym 2018; 195:601-612. [PMID: 29805017 DOI: 10.1016/j.carbpol.2018.05.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/20/2018] [Accepted: 05/01/2018] [Indexed: 12/19/2022]
Abstract
Marine polysaccharides (MPs), including plant, animal, and microbial-derived polysaccharides, can alleviate metabolic syndrome (MetS) by different regulation mechanisms. MPs and their derivatives can attenuate MetS by vary cellular signal pathways, such as peroxisome proliferator-activated receptor, 5' adenosine monophosphate-activated protein kinase, and CCAAT/enhancer binding protein-α. Also, most of MPs cannot be degraded by human innate enzymes, but they can be degraded and fermented by human gut microbiota. The final metabolic products of these polysaccharides are usually short-chain fatty acids (SCFAs), which can change the gut microbiota ecology by altering the existing percentage of special microorganisms. In addition, the SCFAs and changed gut microbiota can regulate enteroendocrine hormone secretion, blood glucose, lipid metabolism levels, and other MetS symptoms. Here, we summarize the up-to-date findings on the effects of MPs, particularly marine microbial-derived polysaccharides, and their metabolites on attenuating MetS.
Collapse
Affiliation(s)
- Xueliang Wang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China
| | - Xin Wang
- Obstetrics Department (Work Number 002312), Qingdao Municipal Hospital (Group), Qingdao, 266000, China
| | - Hao Jiang
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China
| | - Chao Cai
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China
| | - Guoyun Li
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China
| | - Jiejie Hao
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China.
| | - Guangli Yu
- Key Laboratory of Marine Drugs of Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266003, China.
| |
Collapse
|
86
|
Rocha DMUP, Ribeiro PVDM, Caldas APS, da Silva BP, da Silva A, de Almeida AP, da Silva NBM, Machado AM, Alfenas RDCG. Acute consumption of yacon shake did not affect glycemic response in euglycemic, normal weight, healthy adults. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.02.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
87
|
Abstract
The human GI tract harbors a diverse and dynamic microbial community comprising bacteria, archaea, viruses and eukaryotic microbes, which varies in composition from individual to individual. A healthy microbiota metabolizes various indigestible dietary components of the host, maintains host immune homeostasis and nutrient intake, but, an imbalanced microbiota has been reported to be associated with many diseases, including obesity. Rodent studies have produced evidence in support of the causal role of the gut microbiota in the development of obesity, however, such causal relationship is lacking in humans. The objective of this review is to critically analyze the vast information available on the composition, function and alterations of the gut microbiota in obesity and explore the future prospects of this research area.
Collapse
Affiliation(s)
- Shabana
- Department of Microbiology & Molecular Genetics, University of the Punjab, Lahore 54590, Pakistan
| | - Saleem U Shahid
- Department of Microbiology & Molecular Genetics, University of the Punjab, Lahore 54590, Pakistan
| | - Uzma Irfan
- Women University Multan, Multan 66000, Pakistan
| |
Collapse
|
88
|
Padilla-Camberos E, Barragán-Álvarez CP, Diaz-Martinez NE, Rathod V, Flores-Fernández JM. Effects of Agave fructans (Agave tequilana Weber var. azul) on Body Fat and Serum Lipids in Obesity. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2018; 73:34-39. [PMID: 29417384 DOI: 10.1007/s11130-018-0654-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Obesity affects millions of people worldwide, constituting a public health problem associated with premature mortality. Agave fructans decrease fat mass, body and liver weight, and generate satiety in rodents. In the present study the effects of agave fructans on weight control, lipid profile, and physical tolerability were evaluated in obese people. Twenty-eight obese volunteers were randomly divided into two groups. In the first group, 96 mg/bw of agave fructans was administered for 12 weeks; in the second group, maltodextrin as a placebo was administered for 12 weeks. All participants consumed a low-calorie diet of 1500 kcal/day. Anthropometric and biochemical measurements were taken at baseline and at the end of the study. The body mass index (BMI) of the agave fructans treated group was reduced significantly from the baseline to the final measurements. Hip and waist circumferences decreased statistically in both groups. A decrease of 10% in total body fat was observed in the agave fructans treated group, resulting in a statistically significant difference in the final versus baseline measurements between the Agave fructans treated group and the placebo treated group. Triglycerides were reduced significantly in the agave fructans treated group. Glucose values did not change in either group. Agave fructans intake was safe and well tolerated throughout the study. The results showed that the ingestion of agave fructans enhanced the decrease in BMI, the decrease in total body fat, and the decrease in triglycerides in obese individuals who consume a low-calorie diet.
Collapse
Affiliation(s)
- Eduardo Padilla-Camberos
- Unit of Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Normalistas 800, 44270, Guadalajara, State of Jalisco, Mexico
| | - Carla P Barragán-Álvarez
- Unit of Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Normalistas 800, 44270, Guadalajara, State of Jalisco, Mexico
| | - Nestor E Diaz-Martinez
- Unit of Medical and Pharmaceutical Biotechnology, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, A.C. (CIATEJ), Normalistas 800, 44270, Guadalajara, State of Jalisco, Mexico
| | - Vineet Rathod
- Department of Biochemistry, University of Alberta, 474 Medical Sciences Building, Edmonton, AB, T6G 2R3, Canada
| | - José Miguel Flores-Fernández
- Department of Biochemistry, University of Alberta, 474 Medical Sciences Building, Edmonton, AB, T6G 2R3, Canada.
- División de Ingeniería en Industrias Alimentarias e Innovación Agrícola Sustentable, Tecnológico de Estudios Superiores de Villa Guerrero, Highway Toluca-Ixtapan de la Sal, Km 64.5, La Finca, 61763, Villa Guerrero, State of Mexico, Mexico.
| |
Collapse
|
89
|
The efficacy of daily snack replacement with oligofructose-enriched granola bars in overweight and obese adults: a 12-week randomised controlled trial. Br J Nutr 2018; 119:1076-1086. [DOI: 10.1017/s0007114518000211] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AbstractOligofructose is a prebiotic dietary fibre obtained from chicory root inulin. Oligofructose supplementation may affect satiety, food intake, body weight and/or body composition. The aim was to examine the efficacy of oligofructose-supplemented granola bars on the following weight management outcomes: satiety, energy intake, body weight and body composition in overweight or obese adults. In all, fifty-five adults with overweight or obesity (thirty-six females/nineteen males; age: 41 (sd 12) years; 90·6 (sd 11·8) kg; BMI: 29·4 (sd 2·6) kg/m2) participated in a parallel, triple-blind, placebo-controlled intervention. A total of twenty-nine subjects replaced their snacks twice a day with an equienergetic granola bar supplemented with 8 g of oligofructose (OF-Bar). Subjects in the control group (n 26) replaced their snack with a control granola bar without added oligofructose (Co-Bar). Satiety, 24-h energy intake, body weight and body composition (fat mass and waist circumference) were measured at baseline, weeks 6 and 12. In addition, weekly appetite and gastrointestinal side effects were measured. During the intervention, energy intake, body weight and fat mass remained similar in the Co-Bar and OF-Bar groups (all P>0·05). Both groups lost 0·3 (sd 1·2) kg lean mass (P<0·01) and reduced their waist circumference with −2·2 (sd 3·6) cm (P<0·0001) after 12 weeks. The OF-Bar group reported decreased hunger in later weeks of the intervention (P=0·04), less prospective food consumption (P=0·03) and less thirst (P=0·003). To conclude, replacing daily snacks for 12 weeks with oligofructose-supplemented granola bars does not differentially affect energy intake, body weight and body composition compared with a control bar. However, there was an indication that appetite was lower after oligofructose bar consumption.
Collapse
|
90
|
Zhang Q, Yu H, Xiao X, Hu L, Xin F, Yu X. Inulin-type fructan improves diabetic phenotype and gut microbiota profiles in rats. PeerJ 2018; 6:e4446. [PMID: 29507837 PMCID: PMC5835350 DOI: 10.7717/peerj.4446] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/10/2018] [Indexed: 12/18/2022] Open
Abstract
Background & Aims Accumulating research has addressed the linkage between the changes to gut microbiota structure and type 2 diabetes (T2D). Inulin is one type of soluble dietary fiber that can alleviate T2D. As a prebiotic, inulin cannot be digested by humans, but rather is digested by probiotics. However, whether inulin treatment can benefit the entire gut bacteria community remains unknown. In this study, we evaluated the differences in gut microbiota composition among diabetic, inulin-treated diabetic, normal control, and inulin-treated normal control rats. Methods A diabetic rat model was generated by a high-fat diet and streptozotocin injections (HF/STZ). Inulin was orally administered to normal and diabetic rats. To determine the composition of the gut microbiota, fecal DNA extraction and 16S rRNA gene 454 pyrosequencing were performed. Results We found that inulin treatment reduced fasting blood glucose levels and alleviated glucose intolerance and blood lipid panels in diabetic rats. Additionally, inulin treatment increased the serum glucagon-like peptide-1 (GLP-1) level, reduced serum IL-6 level, Il6 expression in epididymal adipose tissue, and Pepck, G6pc expression in liver of diabetic rats. Pyrophosphate sequencing of the 16s V3–V4 region demonstrated an elevated proportion of Firmicutes and a reduced abundance of Bacteroidetes at the phylogenetic level in diabetic rats compared to normal control rats. The characteristics of the gut microbiota in control and inulin-treated rats were similar. Inulin treatment can normalize the composition of the gut microbiota in diabetic rats. At the family and genus levels, probiotic bacteria Lactobacillus and short-chain fatty acid (SCFA)-producing bacteria Lachnospiraceae, Phascolarctobacterium, and Bacteroides were found to be significantly more abundant in the inulin-treated diabetic group than in the non-treated diabetic group. In addition, inulin-treated rats had a lower abundance of Desulfovibrio, which produce lipopolysaccharide (LPS). The abundance of Lachnospiraceae was negatively correlated with the blood glucose response after a glucose load. Conclusion In summary, diabetic rats have different gut microbiota from control rats. Inulin treatment can alleviate gut microbiota dysbiosis in T2D model rats. Moreover, inulin treatment enhanced serum GLP-1 level to suppress IL-6 secretion and production and hepatic gluconeogenesis, resulted in moderation of insulin tolerance.
Collapse
Affiliation(s)
- Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongyue Yu
- Department of Endocrinology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ling Hu
- Department of Endocrinology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Fengjiao Xin
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Xiaobing Yu
- Fengning Ping'an High-tech Industrial Co., Ltd., Hebei Province, China
| |
Collapse
|
91
|
Cawthon CR, de La Serre CB. Gut bacteria interaction with vagal afferents. Brain Res 2018; 1693:134-139. [PMID: 29360469 DOI: 10.1016/j.brainres.2018.01.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/25/2022]
Abstract
Contemporary techniques including the use of germ-free models and next generation sequencing have deepened our understanding of the gut microbiota dynamics and its influence on host physiology. There is accumulating evidence that the gut microbiota can communicate to the CNS and is involved in the development of metabolic and behavioral disorders. Vagal afferent terminals are positioned beneath the gut epithelium where they can receive, directly or indirectly, signals produced by the gut microbiota, to affect host behavior, including feeding behavior. Supplementation with L. Rhamnosus in mice notably causes a decrease in anxiety and these effects are abolished by vagotomy. Additionally, chronic treatment with bacterial byproduct lipopolysaccharide (LPS) blunts vagally-mediated post-ingestive feedback and is associated with increased food intake. Inflammation in the nodose ganglion (NG), the location of vagal afferent neurons' cell bodies, may be a key triggering factor of microbiota-driven vagal alteration. Interestingly, several models show that vagal damage leads to an increase in immune cell (microglia) activation in the NG and remodeling of the vagal pathway. Similarly, diet-driven microbiota dysbiosis is associated with NG microglia activation and decreased vagal outputs to the CNS. Crucially, preventing dysbiosis and microglia activation in high-fat diet fed rodents normalizes vagal innervation and energy intake, highlighting the importance of microbiota/vagal communication in controlling feeding behavior. As of today, new consideration of potential roles for glial influence on vagal communication and new methods of vagal afferent ablation open opportunities to increase our understanding of how the gut microbiota influence its host's health and behavior.
Collapse
Affiliation(s)
- Carolina R Cawthon
- Department of Foods and Nutrition, University of Georgia, 372 Dawson Hall, 305 Sanford Drive, Athens, GA 30602 USA.
| | - Claire B de La Serre
- Department of Foods and Nutrition, University of Georgia, 372 Dawson Hall, 305 Sanford Drive, Athens, GA 30602 USA.
| |
Collapse
|
92
|
Le Bourgot C, Apper E, Blat S, Respondek F. Fructo-oligosaccharides and glucose homeostasis: a systematic review and meta-analysis in animal models. Nutr Metab (Lond) 2018; 15:9. [PMID: 29416552 PMCID: PMC5785862 DOI: 10.1186/s12986-018-0245-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022] Open
Abstract
The aim of this systematic review was to assess the effect of fructo-oligosaccharide supplementation on glucose homeostasis. The search process was based on the selection of publications listed in the Pubmed-Medline database until April 2016 to identify studies evaluating the impact of short-chain fructo-oligosaccharides or oligofructose on glucose homeostasis. Twenty-nine trials were included in the systematic review and the meta-analysis was performed on twelve of these papers according to the inclusion criteria. Fasting blood concentrations of glucose and insulin were selected as pertinent criteria of glucose homeostasis for the meta-analysis. The consumption of fructo-oligosaccharides decreased fasting blood glycaemia levels, whatever the metabolic status (healthy, obese or diabetic) and diet (low-fat or high-fat) throughout the experiment. This reduction was linear with prebiotic dose (from 0 to 13% of the feed). Fasting insulinaemia also decreased linearly with fructo-oligosaccharide supplementation but the reduction was only significant in rodents fed a low-fat diet. Potential underlying mechanisms include gut bacterial fermentation of fructo-oligosaccharides to short-chain fatty acids (SCFA) and bacterial modulation of bile acids, both interacting with host metabolism. This systemic review, followed by the meta-analysis, provides evidence that fructo-oligosaccharide supplementation has a significant effect on glucose homeostasis whatever the health status and diet consumed by animals.
Collapse
Affiliation(s)
- Cindy Le Bourgot
- R&D Department, Tereos, ZI et portuaire, 67390 Marckolsheim, France
| | - Emmanuelle Apper
- R&D Department, Tereos, ZI et portuaire, 67390 Marckolsheim, France
| | - Sophie Blat
- 2INRA, INSERM, Univ Rennes 1, Nutrition Metabolisms and Cancer (NuMeCan), Rennes, France
| | | |
Collapse
|
93
|
Lach G, Schellekens H, Dinan TG, Cryan JF. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018; 15:36-59. [PMID: 29134359 PMCID: PMC5794698 DOI: 10.1007/s13311-017-0585-0] [Citation(s) in RCA: 318] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complex bidirectional communication between the gut and the brain is finely orchestrated by different systems, including the endocrine, immune, autonomic, and enteric nervous systems. Moreover, increasing evidence supports the role of the microbiome and microbiota-derived molecules in regulating such interactions; however, the mechanisms underpinning such effects are only beginning to be resolved. Microbiota-gut peptide interactions are poised to be of great significance in the regulation of gut-brain signaling. Given the emerging role of the gut-brain axis in a variety of brain disorders, such as anxiety and depression, it is important to understand the contribution of bidirectional interactions between peptide hormones released from the gut and intestinal bacteria in the context of this axis. Indeed, the gastrointestinal tract is the largest endocrine organ in mammals, secreting dozens of different signaling molecules, including peptides. Gut peptides in the systemic circulation can bind cognate receptors on immune cells and vagus nerve terminals thereby enabling indirect gut-brain communication. Gut peptide concentrations are not only modulated by enteric microbiota signals, but also vary according to the composition of the intestinal microbiota. In this review, we will discuss the gut microbiota as a regulator of anxiety and depression, and explore the role of gut-derived peptides as signaling molecules in microbiome-gut-brain communication. Here, we summarize the potential interactions of the microbiota with gut hormones and endocrine peptides, including neuropeptide Y, peptide YY, pancreatic polypeptide, cholecystokinin, glucagon-like peptide, corticotropin-releasing factor, oxytocin, and ghrelin in microbiome-to-brain signaling. Together, gut peptides are important regulators of microbiota-gut-brain signaling in health and stress-related psychiatric illnesses.
Collapse
Affiliation(s)
- Gilliard Lach
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Harriet Schellekens
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Food for Health Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Food for Health Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
94
|
Delbès AS, Castel J, Denis RGP, Morel C, Quiñones M, Everard A, Cani PD, Massiera F, Luquet SH. Prebiotics Supplementation Impact on the Reinforcing and Motivational Aspect of Feeding. Front Endocrinol (Lausanne) 2018; 9:273. [PMID: 29896158 PMCID: PMC5987188 DOI: 10.3389/fendo.2018.00273] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/09/2018] [Indexed: 11/25/2022] Open
Abstract
Energy homeostasis is tightly regulated by the central nervous system which responds to nervous and circulating inputs to adapt food intake and energy expenditure. However, the rewarding and motivational aspect of food is tightly dependent of dopamine (DA) release in mesocorticolimbic (MCL) system and could be operant in uncontrolled caloric intake and obesity. Accumulating evidence indicate that manipulating the microbiota-gut-brain axis through prebiotic supplementation can have beneficial impact of the host appetite and body weight. However, the consequences of manipulating the implication of the microbiota-gut-brain axis in the control motivational and hedonic/reinforcing aspects of food are still underexplored. In this study, we investigate whether and how dietary prebiotic fructo-oligosaccharides (FOS) could oppose, or revert, the change in hedonic and homeostatic control of feeding occurring after a 2-months exposure to high-fat high-sugar (HFHS) diet. The reinforcing and motivational components of food reward were assessed using a two-food choice paradigm and a food operant behavioral test in mice exposed to FOS either during or after HFHS exposure. We also performed mRNA expression analysis for key genes involved in limbic and hypothalamic control of feeding. We show in a preventive-like approach, FOS addition of HFHS diet had beneficial impact of hypothalamic neuropeptides, and decreased the operant performance for food but only after an overnight fast while it did not prevent the imbalance in mesolimbic markers for DA signaling induced by palatable diet exposure nor the spontaneous tropism for palatable food when given the choice. However, when FOS was added to control diet after chronic HFHS exposure, although it did not significantly alter body weight loss, it greatly decreased palatable food tropism and consumption and was associated with normalization of MCL markers for DA signaling. We conclude that the nature of the diet (regular chow or HFHS) as well as the timing at which prebiotic supplementation is introduced (preventive or curative) greatly influence the efficacy of the gut-microbiota-brain axis. This crosstalk selectively alters the hedonic or motivational drive to eat and triggers molecular changes in neural substrates involved in the homeostatic and non-homeostatic control of body weight.
Collapse
Affiliation(s)
- Anne-Sophie Delbès
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Julien Castel
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Raphaël G. P. Denis
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Chloé Morel
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Mar Quiñones
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Amandine Everard
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D. Cani
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Florence Massiera
- Laboratoire de Recherche Nutritionnelle KOT CEPRODI SA, Paris, France
| | - Serge H. Luquet
- Université Paris Diderot, Sorbonne Paris Cité, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
- *Correspondence: Serge H. Luquet,
| |
Collapse
|
95
|
Hadri Z, Rasoamanana R, Fromentin G, Azzout-Marniche D, Even PC, Gaudichon C, Darcel N, Bouras AD, Tomé D, Chaumontet C. Fructo-oligosaccharides reduce energy intake but do not affect adiposity in rats fed a low-fat diet but increase energy intake and reduce fat mass in rats fed a high-fat diet. Physiol Behav 2017; 182:114-120. [PMID: 29030250 DOI: 10.1016/j.physbeh.2017.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 11/17/2022]
Abstract
The ingestion of low or high lipid diets enriched with fructo-oligosaccharide (FOS) affects energy homeostasis. Ingesting protein diets also induces a depression of energy intake and decreases body weight. The goal of this study was to investigate the ability of FOS, combined or not with a high level of protein (P), to affect energy intake and body composition when included in diets containing different levels of lipids (L). We performed two studies of similar design over a period of 5weeks. During the first experiment (exp1), after a 3-week period of adaptation to a normal protein-low fat diet, the rats received one of the following four diets for 5weeks (6 rats per group): (i) normal protein (14% P/E (Energy) low fat (10% L/E) diet, (ii) normal protein, low fat diet supplemented with 10% FOS, (iii) high protein (55%P/E) low fat diet, and (iv) high protein, low fat diet supplemented with 10% FOS. In a second experiment (exp2) after the 3-week period of adaptation to a normal protein-high fat diet, the rats received one of the following 4 diets for 5weeks (6 rats per group): (i) normal protein, high fat diet (35% of fat), (ii) normal protein, high fat diet supplemented with 10% FOS, (iii) high protein high fat diet and (iv) high protein high fat diet supplemented with 10% FOS. In low-fat fed rats, FOS did not affect lean body mass (LBM) and fat mass but the protein level reduced fat mass and tended to reduce adiposity. In high-fat fed rats, FOS did not affect LBM but reduced fat mass and adiposity. No additive or antagonistic effects between FOS and the protein level were observed. FOS reduced energy intake in low-fat fed rats, did not affect energy intake in normal-protein high-fat fed rats but surprisingly, and significantly, increased energy intake in high-protein high-fat fed rats. The results thus showed that FOS added to a high-fat diet reduced body fat and body adiposity.
Collapse
Affiliation(s)
- Zouheyr Hadri
- Laboratoire de Bioressources Naturelles Locales, Département de biologie, Faculté des sciences de la nature et de la vie, Université Hassiba Ben Bouali - Chlef, Algeria
| | - Rojo Rasoamanana
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Gilles Fromentin
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Dalila Azzout-Marniche
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Patrick C Even
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Claire Gaudichon
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Nicolas Darcel
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Abdelkader Dilmi Bouras
- Laboratoire de Bioressources Naturelles Locales, Département de biologie, Faculté des sciences de la nature et de la vie, Université Hassiba Ben Bouali - Chlef, Algeria
| | - Daniel Tomé
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France
| | - Catherine Chaumontet
- UMR Physiologie de la Nutrition et du Comportement Alimentaire, AgroParisTech, INRA, Université Paris-Saclay, 16, rue Claude Bernard, F-75005 Paris, France.
| |
Collapse
|
96
|
Modulation of Immune Function in Rats Using Oligosaccharides Extracted from Palm Kernel Cake. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2576921. [PMID: 29349067 PMCID: PMC5733945 DOI: 10.1155/2017/2576921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/12/2017] [Accepted: 10/22/2017] [Indexed: 12/31/2022]
Abstract
To investigate the prebiotic and immunomodulatory effects of PKC extract (OligoPKC) a total of 24 male rats were randomly assigned to three treatment groups receiving basal diet (control), basal diet containing 0.5% OligoPKC, or basal diet containing 1% OligoPKC for four weeks. We found that OligoPKC had no significant effect on the tested growth parameters. However, it increased the size of the total and beneficial bacterial populations while reducing pathogen populations. OligoPKC increased the concentration of immunoglobulins in the serum and cecal contents of rats. It also enhanced the antioxidant capacity of the liver while reducing lipid peroxidation in liver tissue. OligoPKC affected the expression of genes involved in immune system function in the intestine. Therefore, OligoPKC could be considered a potential mannan-based prebiotic for humans and animals due to its beneficial effects on the health and well-being of the model rats.
Collapse
|
97
|
Li KK, Tian PJ, Wang SD, Lei P, Qu L, Huang JP, Shan YJ, Li BL. Targeting gut microbiota: Lactobacillus alleviated type 2 diabetes via inhibiting LPS secretion and activating GPR43 pathway. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.09.049] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
98
|
He M, Shi B. Gut microbiota as a potential target of metabolic syndrome: the role of probiotics and prebiotics. Cell Biosci 2017; 7:54. [PMID: 29090088 PMCID: PMC5655955 DOI: 10.1186/s13578-017-0183-1] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022] Open
Abstract
Metabolic syndrome (MS) comprises central obesity, increased plasma glucose levels, hyperlipidemia and hypertension, and its incidence is increasing due to changes in lifestyle and dietary structure in recent years. MS has been proven to be associated with an increased incidence of cardiovascular diseases and type 2 diabetes mellitus, leading to morbidity and mortality. In this manuscript, we review recent studies concerning the role of the gut microbiota in MS modulation. Manipulation of the gut microbiota through the administration of prebiotics or probiotics may assist in weight loss and reduce plasma glucose and serum lipid levels, decreasing the incidence of cardiovascular diseases and type 2 diabetes mellitus. To the best of our knowledge, short-chain fatty acids (SCFAs), bile salt hydrolase (BSH), metabolic endotoxemia and the endocannabinoid (eCB) system are essential in regulating the initiation and progression of MS through the normalization of adipogenesis and the regulation of insulin secretion, fat accumulation, energy homeostasis, and plasma cholesterol levels. Therefore, the gut microbiota may serve as a potential therapeutic target for MS. However, further studies are needed to enhance our understanding of manipulating the gut microbiota and the role of the gut microbiota in MS prevention and treatment.
Collapse
Affiliation(s)
- Mingqian He
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061 Shaanxi People's Republic of China
| | - Bingyin Shi
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, 710061 Shaanxi People's Republic of China
| |
Collapse
|
99
|
Esgalhado M, Kemp JA, Damasceno NR, Fouque D, Mafra D. Short-chain fatty acids: a link between prebiotics and microbiota in chronic kidney disease. Future Microbiol 2017; 12:1413-1425. [PMID: 29027814 DOI: 10.2217/fmb-2017-0059] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Under physiologic conditions, the human gut microbiota performs several activities essential to the body health. In contrast, their imbalances exacerbate some actions which can promote a cascade of metabolic abnormalities, and vice versa. Numerous diseases, including chronic kidney disease, are associated with gut microbiota imbalance, and among several strategies to re-establish gut symbiosis, prebiotics seem to represent an effective nonpharmacological approach. Prebiotics fermentation by gut microbiota produce short-chain fatty acids, which improve the gut barrier integrity and function, and modulate the glucose and lipid metabolism as well as the inflammatory response and immune system. Therefore, this literature review intends to discuss the beneficial effects of prebiotics in human health through short-chain fatty acids production, with a particular interest on chronic kidney disease.
Collapse
Affiliation(s)
- Marta Esgalhado
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Julie A Kemp
- Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| | - Nagila Rt Damasceno
- Department of Nutrition, Faculty of Public Health Nutrition, São Paulo University, São Paulo, Brazil
| | - Denis Fouque
- Department of Nephrology, Centre Hospitalier Lyon Sud, University Lyon, UCBL, Inserm Carmen, CENS, F-69622 Lyon, France
| | - Denise Mafra
- Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil.,Graduate Program in Medical Sciences, Fluminense Federal University (UFF), Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
100
|
Cavallari JF, Schertzer JD. Intestinal Microbiota Contributes to Energy Balance, Metabolic Inflammation, and Insulin Resistance in Obesity. J Obes Metab Syndr 2017; 26:161-171. [PMID: 31089513 PMCID: PMC6484920 DOI: 10.7570/jomes.2017.26.3.161] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/01/2017] [Accepted: 07/19/2017] [Indexed: 01/01/2023] Open
Abstract
Obesity is associated with increased risk of developing metabolic diseases such as type 2 diabetes. The origins of obesity are multi-factorial, but ultimately rooted in increased host energy accumulation or retention. The gut microbiota has been implicated in control of host energy balance and nutrient extraction from dietary sources. The microbiota also impacts host immune status and dysbiosis-related inflammation can augment insulin resistance, independently of obesity. Advances in microbial metagenomic analyses and directly manipulating bacterial-host models of obesity have contributed to our understanding of the relationship between gut bacteria and metabolic disease. Foodborne, or drug-mediated perturbations to the gut microbiota can increase metabolic inflammation, insulin resistance, and dysglycemia. There is now some evidence that specific bacterial species can influence obesity and related metabolic defects such as insulin sensitivity. Components of bacteria are sufficient to impact obesity-related changes in metabolism. In fact, different microbial components derived from the bacterial cell wall can increase or decrease insulin resistance. Improving our understanding of the how components of the microbiota alter host metabolism is positioned to aid in the development of dietary interventions, avoiding triggers of dysbiosis, and generating novel therapeutic strategies to combat increasing rates of obesity and diabetes.
Collapse
Affiliation(s)
- Joseph F. Cavallari
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario,
Canada
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences and Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario,
Canada
| |
Collapse
|