201
|
Atypical immunometabolism and metabolic reprogramming in liver cancer: Deciphering the role of gut microbiome. Adv Cancer Res 2020; 149:171-255. [PMID: 33579424 DOI: 10.1016/bs.acr.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Much recent research has delved into understanding the underlying molecular mechanisms of HCC pathogenesis, which has revealed to be heterogenous and complex. Two major hallmarks of HCC include: (i) a hijacked immunometabolism and (ii) a reprogramming in metabolic processes. We posit that the gut microbiota is a third component in an entanglement triangle contributing to HCC progression. Besides metagenomic studies highlighting the diagnostic potential in the gut microbiota profile, recent research is pinpointing the gut microbiota as an instigator, not just a mere bystander, in HCC. In this chapter, we discuss mechanistic insights on atypical immunometabolism and metabolic reprogramming in HCC, including the examination of tumor-associated macrophages and neutrophils, tumor-infiltrating lymphocytes (e.g., T-cell exhaustion, regulatory T-cells, natural killer T-cells), the Warburg effect, rewiring of the tricarboxylic acid cycle, and glutamine addiction. We further discuss the potential involvement of the gut microbiota in these characteristics of hepatocarcinogenesis. An immediate highlight is that microbiota metabolites (e.g., short chain fatty acids, secondary bile acids) can impair anti-tumor responses, which aggravates HCC. Lastly, we describe the rising 'new era' of immunotherapies (e.g., immune checkpoint inhibitors, adoptive T-cell transfer) and discuss for the potential incorporation of gut microbiota targeted therapeutics (e.g., probiotics, fecal microbiota transplantation) to alleviate HCC. Altogether, this chapter invigorates for continuous research to decipher the role of gut microbiome in HCC from its influence on immunometabolism and metabolic reprogramming.
Collapse
|
202
|
Seo KH, Kim DH, Yokoyama WH, Kim H. Synbiotic Effect of Whole Grape Seed Flour and Newly Isolated Kefir Lactic Acid Bacteria on Intestinal Microbiota of Diet-Induced Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:13131-13137. [PMID: 32124605 DOI: 10.1021/acs.jafc.0c01240] [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] [Indexed: 06/10/2023]
Abstract
Alterations of intestinal microbiota by synbiotic action of pre- and probiotics may confer health benefits to the host. In this study, high-throughput sequencing of 16S rRNA was used to analyze intestinal microbiota in feces, and the relative abundance of intestinal bacteria was correlated with physiological data from a prior study of a synbiotic combination of flavonoid-rich wine grape seed flour (WGF) and two newly isolated kefir lactic acid bacteria (LAB) in diet-induced obese mice. The combination of WGF and LAB enhanced observed operational taxonomic units and Chao1 index compared to WGF alone, indicating an increase in the microbial community richness. The combination significantly enhanced abundance of Akkermansia muciniphila and Nocardia coeliaca and their abundance had an inverse relationship with body weight gain and adipose weight. In conclusion, the synbiotic effects of WGF and LAB on improvement of high-fat-diet-induced obesity are strongly linked to remodeling intestinal microbiota.
Collapse
Affiliation(s)
- Kun-Ho Seo
- KU Center for One Health, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, South Korea
| | - Dong-Hyeon Kim
- KU Center for One Health, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 05029, South Korea
| | - Wallace H Yokoyama
- Agricultural Research Service (ARS), United States Department of Agriculture (USDA), Albany, California 94710, United States
| | - Hyunsook Kim
- Department of Food and Nutrition, Hanyang University, Seongdong-gu, Seoul 04763, South Korea
| |
Collapse
|
203
|
Molinaro A, Bel Lassen P, Henricsson M, Wu H, Adriouch S, Belda E, Chakaroun R, Nielsen T, Bergh PO, Rouault C, André S, Marquet F, Andreelli F, Salem JE, Assmann K, Bastard JP, Forslund S, Le Chatelier E, Falony G, Pons N, Prifti E, Quinquis B, Roume H, Vieira-Silva S, Hansen TH, Pedersen HK, Lewinter C, Sønderskov NB, Køber L, Vestergaard H, Hansen T, Zucker JD, Galan P, Dumas ME, Raes J, Oppert JM, Letunic I, Nielsen J, Bork P, Ehrlich SD, Stumvoll M, Pedersen O, Aron-Wisnewsky J, Clément K, Bäckhed F. Imidazole propionate is increased in diabetes and associated with dietary patterns and altered microbial ecology. Nat Commun 2020; 11:5881. [PMID: 33208748 PMCID: PMC7676231 DOI: 10.1038/s41467-020-19589-w] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Microbiota-host-diet interactions contribute to the development of metabolic diseases. Imidazole propionate is a novel microbially produced metabolite from histidine, which impairs glucose metabolism. Here, we show that subjects with prediabetes and diabetes in the MetaCardis cohort from three European countries have elevated serum imidazole propionate levels. Furthermore, imidazole propionate levels were increased in subjects with low bacterial gene richness and Bacteroides 2 enterotype, which have previously been associated with obesity. The Bacteroides 2 enterotype was also associated with increased abundance of the genes involved in imidazole propionate biosynthesis from dietary histidine. Since patients and controls did not differ in their histidine dietary intake, the elevated levels of imidazole propionate in type 2 diabetes likely reflects altered microbial metabolism of histidine, rather than histidine intake per se. Thus the microbiota may contribute to type 2 diabetes by generating imidazole propionate that can modulate host inflammation and metabolism. Gut microbial metabolism of nutrients contributes to metabolic diseases, and the histidine metabolite imidazole propionate (ImP) is produced by type 2 diabetes (T2D) associated microbiome. Here the authors report that circulating ImP levels are increased in subjects with prediabetes or T2D in three European populations, and this increase associates with altered gut microbiota rather than dietary histidine.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45, Gothenburg, Sweden.,Department of Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Pierre Bel Lassen
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France.,Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France
| | - Marcus Henricsson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Hao Wu
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Solia Adriouch
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Eugeni Belda
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France.,Integromics Unit, Institute of Cardiometabolism and Nutrition, 75013, Paris, France
| | - Rima Chakaroun
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Per-Olof Bergh
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Christine Rouault
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Sébastien André
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Florian Marquet
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Fabrizio Andreelli
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Joe-Elie Salem
- Assistance Publique Hôpitaux de Paris, Clinical Investigation Center Paris East, 75013, Paris, France
| | - Karen Assmann
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France
| | - Jean-Philippe Bastard
- Assistance Publique Hôpitaux de Paris, Biochemistry and Hormonology Department, Tenon Hospital, 75020, Paris, France
| | - Sofia Forslund
- Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin and the Max-Delbrück Center, Berlin, Germany
| | | | - Gwen Falony
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Leuven, Belgium
| | - Nicolas Pons
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Paris, France
| | - Edi Prifti
- Integromics Unit, Institute of Cardiometabolism and Nutrition, 75013, Paris, France.,Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, 93143, Bondy, France
| | - Benoit Quinquis
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Paris, France
| | - Hugo Roume
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Paris, France
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium.,Center for Microbiology, VIB, Leuven, Belgium
| | - Tue H Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Helle Krogh Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Christian Lewinter
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Nadja B Sønderskov
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | | | - Lars Køber
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Henrik Vestergaard
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Jean-Daniel Zucker
- Unité de Modélisation Mathématique et Informatique des Systèmes Complexes, UMMISCO, 93143, Bondy, France
| | - Pilar Galan
- Sorbonne Paris Cité Epidemiology and Statistics Research Centre (CRESS), U1153 Inserm, U1125, Inra, Cnam, University of Paris 13, Nutritional Epidemiology Research Team (EREN), 93017, Bobigny, France
| | - Marc-Emmanuel Dumas
- Computational and Systems Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, SW7 2AZ, UK.,Genomic and Environmental Medicine, National Heart & Lung Institute, Faculty of Medicine, Imperial College London, London, SW3 6KY, UK
| | - Jeroen Raes
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Paris, France.,Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven, Belgium
| | - Jean-Michel Oppert
- Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France
| | - Ivica Letunic
- Biobyte Solutions GmbH, Bothestr. 142, 69117, Heidelberg, Germany
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE41128, Gothenburg, Sweden
| | - Peer Bork
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg and European Molecular Biology Laboratory, Heidelberg, Germany
| | - S Dusko Ehrlich
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, Paris, France
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Judith Aron-Wisnewsky
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France.,Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France
| | - Karine Clément
- INSERM, Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, Paris, France. .,Assistance Publique Hôpitaux de Paris, Pitie-Salpêtrière Hospital, Nutrition department, CRNH Ile de France, Paris, France.
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine and Sahlgrenska Center for Cardiovascular and Metabolic Research, University of Gothenburg, 413 45, Gothenburg, Sweden. .,Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark. .,Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
204
|
Džunková M, Lipták R, Vlková B, Gardlík R, Čierny M, Moya A, Celec P. Salivary microbiome composition changes after bariatric surgery. Sci Rep 2020; 10:20086. [PMID: 33208788 PMCID: PMC7674438 DOI: 10.1038/s41598-020-76991-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies show that the salivary microbiome in subjects with obesity differ from those without obesity, but the mechanism of interaction between the salivary microbiome composition and body weight is unclear. Herein we investigate this relation by analyzing saliva samples from 35 adult patients with obesity undergoing bariatric surgery. Our aim was to describe salivary microbiome changes during body weight loss on an individual-specific level, and to elucidate the effect of bariatric surgery on the salivary microbiome which has not been studied before. Analysis of samples collected before and 1 day after surgery, as well as 3 and 12 months after surgery, showed that the salivary microbiome changed in all study participants, but these changes were heterogeneous. In the majority of participants proportions of Gemella species, Granulicatella elegans, Porphyromonas pasteri, Prevotella nanceiensis and Streptococcus oralis decreased, while Veillonella species, Megasphaera micronuciformis and Prevotella saliva increased. Nevertheless, we found participants deviating from this general trend which suggests that a variety of individual-specific factors influence the salivary microbiome composition more effectively than the body weight dynamics alone. The observed microbiome alternations could be related to dietary changes. Therefore, further studies should focus on association with altered taste preferences and potential oral health consequences.
Collapse
Affiliation(s)
- Mária Džunková
- Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Róbert Lipták
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Barbora Vlková
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Roman Gardlík
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Michal Čierny
- Department of Bariatric Surgery, Břeclav Hospital, Břeclav, Czech Republic
| | - Andrés Moya
- Department of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencian Community (FISABIO-Public Health), Valencia, Spain.
- CIBER in Epidemiology and Public Health (CIBEResp), Madrid, Spain.
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain.
| | - Peter Celec
- Institute of Molecular Biomedicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
| |
Collapse
|
205
|
Galyean S, Sawant D, Shin AC. Immunometabolism, Micronutrients, and Bariatric Surgery: The Use of Transcriptomics and Microbiota-Targeted Therapies. Mediators Inflamm 2020; 2020:8862034. [PMID: 33281501 PMCID: PMC7685844 DOI: 10.1155/2020/8862034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Obesity is associated with the gut microbiota and decreased micronutrient status. Bariatric surgery is a recommended therapy for obesity. It can positively affect the composition of the gut bacteria but also disrupt absorption of nutrients. Low levels of micronutrients can affect metabolic processes, like glycolysis, TCA cycle, and oxidative phosphorylation, that are associated with the immune system also known as immunometabolism. METHODS MEDLINE, PUBMED, and Google Scholar were searched. Articles involving gut microbiome, micronutrient deficiency, gut-targeted therapies, transcriptome analysis, micronutrient supplementation, and bariatric surgery were included. RESULTS Studies show that micronutrients play a pivotal role in the intestinal immune system and regulating immunometabolism. Research demonstrates that gut-targeting therapies may improve the microbiome health for bariatric surgery populations. There is limited research that examines the role of micronutrients in modulating the gut microbiota among the bariatric surgery population. CONCLUSIONS Investigations are needed to understand the influence that micronutrient deficiencies have on the gut, particularly immunometabolism. Nutritional transcriptomics shows great potential in providing this type of analysis to develop gut-modulating therapies as well as more personalized nutrition recommendations for bariatric surgery patients.
Collapse
Affiliation(s)
- Shannon Galyean
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Dhanashree Sawant
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Andrew C. Shin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
206
|
Van Hul M, Le Roy T, Prifti E, Dao MC, Paquot A, Zucker JD, Delzenne NM, Muccioli GG, Clément K, Cani PD. From correlation to causality: the case of Subdoligranulum. Gut Microbes 2020; 12:1-13. [PMID: 33323004 PMCID: PMC7744154 DOI: 10.1080/19490976.2020.1849998] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gut microbes are considered as major factors contributing to human health. Nowadays, the vast majority of the data available in the literature are mostly exhibiting negative or positive correlations between specific bacteria and metabolic parameters. From these observations, putative detrimental or beneficial effects are then inferred. Akkermansia muciniphila is one of the unique examples for which the correlations with health benefits have been causally validated in vivo in rodents and humans. In this study, based on available metagenomic data in overweight/obese population and clinical variables that we obtained from two cohorts of individuals (n = 108) we identified several metagenomic species (MGS) strongly associated with A. muciniphila with one standing out: Subdoligranulum. By analyzing both qPCR and shotgun metagenomic data, we discovered that the abundance of Subdoligranulum was correlated positively with microbial richness and HDL-cholesterol levels and negatively correlated with fat mass, adipocyte diameter, insulin resistance, levels of leptin, insulin, CRP, and IL6 in humans. Therefore, to further explore whether these strong correlations could be translated into causation, we investigated the effects of the unique cultivated strain of Subdoligranulum (Subdoligranulum variabile DSM 15176 T) in obese and diabetic mice as a proof-of-concept. Strikingly, there were no significant difference in any of the hallmarks of obesity and diabetes measured (e.g., body weight gain, fat mass gain, glucose tolerance, liver weight, plasma lipids) at the end of the 8 weeks of treatment. Therefore, the absence of effect following the supplementation with S. variabile indicates that increasing the intestinal abundance of this bacterium is not translated into beneficial effects in mice. In conclusion, we demonstrated that despite the fact that numerous strong correlations exist between a given bacteria and health, proof-of-concept experiments are required to be further validated or not in vivo. Hence, an urgent need for causality studies is warranted to move from human observations to preclinical validations.
Collapse
Affiliation(s)
- Matthias Van Hul
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Tiphaine Le Roy
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Edi Prifti
- Institut de Recherche et Developpement, IRD, Sorbonne Unive.rsity, UMMISCO, Bondy, France,Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Maria Carlota Dao
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Jean-Daniel Zucker
- Institut de Recherche et Developpement, IRD, Sorbonne Unive.rsity, UMMISCO, Bondy, France,Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesities: Systemic Approaches (Nutriomics) Research Unit, Paris, France,Assistance Publique Hôpitaux de Paris, Nutrition Department, Pitié-Salpêtrière Hospital, CRNH Ile de France, Paris, France
| | - Patrice D. Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), UCLouvain, Université Catholique de Louvain, Brussels, Belgium,CONTACT Patrice D. Cani UCLouvain, Université Catholique de Louvain, LDRI, Metabolism and Nutrition Research Group, BrusselsB-1200, Belgium
| |
Collapse
|
207
|
Obesity and Related Type 2 Diabetes: A Failure of the Autonomic Nervous System Controlling Gastrointestinal Function? GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2040039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The pandemic spread of obesity and type 2 diabetes is a serious health problem that cannot be contained with common therapies. At present, the most effective therapeutic tool is metabolic surgery, which substantially modifies the gastrointestinal anatomical structure. This review reflects the state of the art research in obesity and type 2 diabetes, describing the probable reason for their spread, how the various brain sectors are involved (with particular emphasis on the role of the vagal system controlling different digestive functions), and the possible mechanisms for the effectiveness of bariatric surgery. According to the writer’s interpretation, the identification of drugs that can modulate the activity of some receptor subunits of the vagal neurons and energy-controlling structures of the central nervous system (CNS), and/or specific physical treatment of cortical areas, could reproduce, non-surgically, the positive effects of metabolic surgery.
Collapse
|
208
|
Gut Microbiota in Patients with Morbid Obesity Before and After Bariatric Surgery: a Ten-Year Review Study (2009-2019). Obes Surg 2020; 31:317-326. [PMID: 33130944 DOI: 10.1007/s11695-020-05074-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
The changes in the composition and function of gut microbiota affect the metabolic functions (which are mediated by microbial effects) in patients with obesity, resulting in significant physiological regulation in these patients. Most of the studies emphasize that the Western-style diet (high fat and low vegetable consumption) leads to significant changes in the intestinal microbiome in individuals with metabolic syndrome. A deeper understanding of the profiles of gut microbes will contribute to the development of new therapeutic strategies for the management of metabolic syndrome and other metabolic diseases and related disorders. The aim of this review is to evaluate recent experimental evidence outlining the alterations of gut microbiota composition and function in recovery from bariatric surgical operations with an emphasis on sleeve gastrectomy and gastric bypass.
Collapse
|
209
|
Menni C, Zhu J, Le Roy CI, Mompeo O, Young K, Rebholz CM, Selvin E, North KE, Mohney RP, Bell JT, Boerwinkle E, Spector TD, Mangino M, Yu B, Valdes AM. Serum metabolites reflecting gut microbiome alpha diversity predict type 2 diabetes. Gut Microbes 2020; 11:1632-1642. [PMID: 32576065 PMCID: PMC7524143 DOI: 10.1080/19490976.2020.1778261] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/10/2020] [Accepted: 05/21/2020] [Indexed: 02/03/2023] Open
Abstract
Type 2 diabetes (T2D) is associated with reduced gut microbiome diversity, although the cause is unclear. Metabolites generated by gut microbes also appear to be causative factors in T2D. We therefore searched for serum metabolites predictive of gut microbiome diversity in 1018 females from TwinsUK with concurrent metabolomic profiling and microbiome composition. We generated a Microbial Metabolites Diversity (MMD) score of six circulating metabolites that explained over 18% of the variance in microbiome alpha diversity. Moreover, the MMD score was associated with a significantly lower odds of prevalent (OR[95%CI] = 0.22[0.07;0.70], P = .01) and incident T2D (HR[95%CI] = 0.31[0.11,0.90], P = .03). We replicated our results in 1522 individuals from the ARIC study (prevalent T2D: OR[95%CI] = 0.79[0.64,0.96], P = .02, incident T2D: HR[95%CI] = 0.87[0.79,0.95], P = .003). The MMD score mediated 28%[15%,94%] of the total effect of gut microbiome on T2D after adjusting for confounders. Metabolites predicting higher microbiome diversity included 3-phenylpropionate(hydrocinnamate), indolepropionate, cinnamoylglycine and 5-alpha-pregnan-3beta,20 alpha-diol monosulfate(2) of which indolepropionate and phenylpropionate have already been linked to lower incidence of T2D. Metabolites correlating with lower microbial diversity included glutarate and imidazole propionate, of which the latter has been implicated in insulin resistance. Our results suggest that the effect of gut microbiome diversity on T2D is largely mediated by microbial metabolites, which might be modifiable by diet.
Collapse
Affiliation(s)
- Cristina Menni
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Jialing Zhu
- School of Public Health, University of Texas Health Science Center, Houston, TX, USA
| | - Caroline I Le Roy
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Olatz Mompeo
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Kristin Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Casey M. Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kari E. North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Eric Boerwinkle
- School of Public Health, University of Texas Health Science Center, Houston, TX, USA
- Baylor College of Medicine, Houston, TX, USA
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
| | - Bing Yu
- School of Public Health, University of Texas Health Science Center, Houston, TX, USA
| | - Ana M Valdes
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, UK
- School of Medicine, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham, UK
| |
Collapse
|
210
|
Härma MA, Adeshara K, Istomin N, Lehto M, Blaut M, Savolainen MJ, Hörkkö S, Groop PH, Koivukangas V, Hukkanen J. Gastrointestinal manifestations after Roux-en-Y gastric bypass surgery in individuals with and without type 2 diabetes. Surg Obes Relat Dis 2020; 17:585-594. [PMID: 33246847 DOI: 10.1016/j.soard.2020.10.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) surgery is an effective treatment for obesity, which improves cardiovascular health and reduces the risk of premature mortality. However, some reports have suggested that RYGB may predispose patients to adverse health outcomes, such as inflammatory bowel disease (IBD) and colorectal cancer. OBJECTIVES The present prospective study aimed to evaluate the impact of RYGB surgery on cardiovascular risk factors and gastrointestinal inflammation in individuals with and without type 2 diabetes (T2D). SETTING University hospital setting in Finland. METHODS Blood and fecal samples were collected at baseline and 6 months after surgery from 30 individuals, of which 16 had T2D and 14 were nondiabetics. There were also single study visits for 6 healthy reference patients. Changes in cardiovascular risk factors, serum cholesterol, and triglycerides were investigated before and after surgery. Fecal samples were analyzed for calprotectin, anti-Saccharomyces cerevisiae immunoglobulin A antibodies (ASCA), active lipopolysaccharide (LPS) concentration, short-chain fatty acids (SCFAs), intestinal alkaline phosphatase activity, and methylglyoxal-hydro-imidazolone (MG-H1) protein adducts formation. RESULTS After RYGB, weight decreased on average -21.6% (-27.2 ± 7.8 kg), excess weight loss averaged 51%, and there were improvements in cardiovascular risk factors. Fecal calprotectin levels (P < .001), active LPS concentration (P < .002), ASCA (P < .02), and MG-H1 (P < .02) values increased significantly, whereas fecal SCFAs, especially acetate (P < .002) and butyrate (P < .03) levels, were significantly lowered. CONCLUSION The intestinal homeostasis is altered after RYGB, with several fecal markers suggesting increased inflammation; however, clinical significance of the detected changes is currently uncertain. As chronic inflammation may predispose patients to adverse health effects, our findings may have relevance for the suggested association between RYGB and increased risks of incident IBD and colorectal cancer.
Collapse
Affiliation(s)
- Mari-Anne Härma
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Krishna Adeshara
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Natalie Istomin
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Nordlab, Oulu University Hospital, Oulu, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Michael Blaut
- Department of Gastrointestinal Microbiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Markku J Savolainen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Research Unit of Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sohvi Hörkkö
- Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Nordlab, Oulu University Hospital, Oulu, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland; Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Vesa Koivukangas
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Surgery, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Janne Hukkanen
- Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Research Unit of Internal Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
211
|
Osinski C, Le Gléau L, Poitou C, de Toro-Martin J, Genser L, Fradet M, Soula HA, Leturque A, Blugeon C, Jourdren L, Hubert EL, Clément K, Serradas P, Ribeiro A. Type 2 diabetes is associated with impaired jejunal enteroendocrine GLP-1 cell lineage in human obesity. Int J Obes (Lond) 2020; 45:170-183. [PMID: 33037328 PMCID: PMC7752761 DOI: 10.1038/s41366-020-00694-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 09/07/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022]
Abstract
Objectives Altered enteroendocrine cell (EEC) function in obesity and type 2 diabetes is not fully understood. Understanding the transcriptional program that controls EEC differentiation is important because some EEC types harbor significant therapeutic potential for type 2 diabetes. Methods EEC isolation from jejunum of obese individuals with (ObD) or without (Ob) type 2 diabetes was obtained with a new method of cell sorting. EEC transcriptional profiles were established by RNA-sequencing in a first group of 14 Ob and 13 ObD individuals. EEC lineage and densities were studied in the jejunum of a second independent group of 37 Ob, 21 ObD and 22 non obese (NOb) individuals. Results The RNA seq analysis revealed a distinctive transcriptomic signature and a decreased differentiation program in isolated EEC from ObD compared to Ob individuals. In the second independent group of ObD, Ob and NOb individuals a decreased GLP-1 cell lineage and GLP-1 maturation from proglucagon, were observed in ObD compared to Ob individuals. Furthermore, jejunal density of GLP-1-positive cells was significantly reduced in ObD compared to Ob individuals. Conclusions These results highlight that the transcriptomic signature of EEC discriminate obese subjects according to their diabetic status. Furthermore, type 2 diabetes is associated with reduced GLP-1 cell differentiation and proglucagon maturation leading to low GLP-1-cell density in human obesity. These mechanisms could account for the decrease plasma GLP-1 observed in metabolic diseases.
Collapse
Affiliation(s)
- Céline Osinski
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France
| | - Léa Le Gléau
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France
| | - Christine Poitou
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France.,Nutrition Department, Pitié-Salpêtrière hospital, Assistance Publique/Hôpitaux de Paris, F-75013, Paris, France
| | - Juan de Toro-Martin
- Sorbonne Université, Université de Paris, INSERM, Cordeliers Research Center, F-75006, Paris, France.,Institute of Nutrition and Functional Foods (INAF), School of Nutrition, Université Laval, Quebec, QC, Canada
| | - Laurent Genser
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France.,Hepato-Biliary-Pancreatic Gastrointestinal Surgery and Liver Transplantation, Pitié-Salpêtrière Hospital, Assistance Publique/Hôpitaux de Paris, F-75013, Paris, France
| | - Magali Fradet
- Cytometry platform, Institut Cardiometabolism and Nutrition, F-75013, Paris, France.,Institut de Biologie, CIRB, Collège de France, F-75005, Paris, France
| | - Hédi Antoine Soula
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France
| | - Armelle Leturque
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France
| | - Corinne Blugeon
- Genomics core facility, Département de biologie, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Laurent Jourdren
- Genomics core facility, Département de biologie, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Edwige Ludiwyne Hubert
- Sorbonne Université, Université de Paris, INSERM, Cordeliers Research Center, F-75006, Paris, France.,SERVIER, ADIR, F-92284, Suresnes, cedex, France
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France.,Nutrition Department, Pitié-Salpêtrière hospital, Assistance Publique/Hôpitaux de Paris, F-75013, Paris, France
| | - Patricia Serradas
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France.
| | - Agnès Ribeiro
- Sorbonne Université, INSERM, Nutrition and obesities: systemic approaches, F-75013, Paris, France.
| |
Collapse
|
212
|
Nogacka AM, de Los Reyes-Gavilán CG, Martínez-Faedo C, Ruas-Madiedo P, Suarez A, Mancabelli L, Ventura M, Cifuentes A, León C, Gueimonde M, Salazar N. Impact of Extreme Obesity and Diet-Induced Weight Loss on the Fecal Metabolome and Gut Microbiota. Mol Nutr Food Res 2020; 65:e2000030. [PMID: 32966685 DOI: 10.1002/mnfr.202000030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
SCOPE A limited number of human studies have characterized fecal microbiota and metabolome in extreme obesity and after diet-induced weight loss. METHODS AND RESULTS Fecal samples from normal-weight and extremely obese adults and from obese participants before and after moderate diet-induced weight loss are evaluated for their interaction with the intestinal adenocarcinoma cell line HT29 using an impedance-based in vitro model, which reveals variations in the interaction between the gut microbiota and host linked to obesity status. Microbiota composition, short chain fatty acids, and other intestinal metabolites are further analyzed to assess the interplay among diet, gut microbiota, and host in extreme obesity. Microbiota profiles are distinct between normal-weight and obese participants and are accompanied by fecal signatures in the metabolism of biliary compounds and catecholamines. Moderate diet-induced weight loss promotes shifts in the gut microbiota, and the primary fecal metabolomics features are associated with diet and the gut-liver and gut-brain axes. CONCLUSIONS Analyses of the fecal microbiota and metabolome enable assessment of the impact of diet on gut microbiota composition and activity, supporting the potential use of certain fecal metabolites or members of the gut microbiota as biomarkers for the efficacy of weight loss in extreme obesity.
Collapse
Affiliation(s)
- Alicja M Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Asturias, 33300, Spain.,Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| | - Clara G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Asturias, 33300, Spain.,Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| | - Ceferino Martínez-Faedo
- Endocrinology and Nutrition Service, Central University Hospital of Asturias (HUCA), Oviedo, Asturias, 33011, Spain.,Endocrinology, Nutrition, Diabetes and Obesity Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Asturias, 33300, Spain.,Functionality and Ecology of Beneficial Microorganisms, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| | - Adolfo Suarez
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain.,Digestive Service, Central University Hospital of Asturias (HUCA), Oviedo, Asturias, 33011, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, 43121, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, 43121, Italy
| | - Alejandro Cifuentes
- Laboratory of Foodomics, Institute of Food Science Research, CIAL, CSIC, Nicolás Cabrera 9, Madrid, 28049, Spain
| | - Carlos León
- Department of Bioengineering, Universidad Carlos III de Madrid, Leganés, Madrid, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Asturias, 33300, Spain.,Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Asturias, 33300, Spain.,Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, 33011, Spain
| |
Collapse
|
213
|
Huybrechts I, Zouiouich S, Loobuyck A, Vandenbulcke Z, Vogtmann E, Pisanu S, Iguacel I, Scalbert A, Indave I, Smelov V, Gunter MJ, Michels N. The Human Microbiome in Relation to Cancer Risk: A Systematic Review of Epidemiologic Studies. Cancer Epidemiol Biomarkers Prev 2020; 29:1856-1868. [PMID: 32727720 PMCID: PMC7541789 DOI: 10.1158/1055-9965.epi-20-0288] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/06/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The microbiome has been hypothesized to play a role in cancer development. Because of the diversity of published data, an overview of available epidemiologic evidence linking the microbiome with cancer is now needed. We conducted a systematic review using a tailored search strategy in Medline and EMBASE databases to identify and summarize the current epidemiologic literature on the relationship between the microbiome and different cancer outcomes published until December 2019. We identified 124 eligible articles. The large diversity of parameters used to describe microbial composition made it impossible to harmonize the different studies in a way that would allow meta-analysis, therefore only a qualitative description of results could be performed. Fifty studies reported differences in the gut microbiome between patients with colorectal cancer and various control groups. The most consistent findings were for Fusobacterium, Porphyromonas, and Peptostreptococcus being significantly enriched in fecal and mucosal samples from patients with colorectal cancer. For the oral microbiome, significantly increased and decreased abundance was reported for Fusobacterium and Streptococcus, respectively, in patients with oral cancer compared with controls. Overall, although there was a large amount of evidence for some of these alterations, most require validation in high-quality, preferably prospective, epidemiologic studies.
Collapse
Affiliation(s)
| | - Semi Zouiouich
- International Agency for Research on Cancer, Lyon, France
| | - Astrid Loobuyck
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Zeger Vandenbulcke
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Emily Vogtmann
- Division of Cancer Epidemiology & Genetics, NCI, Bethesda, Maryland
| | - Silvia Pisanu
- International Agency for Research on Cancer, Lyon, France
- Department of Biomedical Sciences, Section of Microbiology and Virology, University of Cagliari, Cagliari, Italy
| | - Isabel Iguacel
- International Agency for Research on Cancer, Lyon, France
- GENUD (Growth, Exercise, NUtrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza, Zaragoza, Spain
| | | | - Iciar Indave
- International Agency for Research on Cancer, Lyon, France
| | - Vitaly Smelov
- International Agency for Research on Cancer, Lyon, France
- Division of Noncommunicable Diseases and Promoting Health through the Life-course, WHO Regional Office for Europe, Copenhagen, Denmark
| | - Marc J Gunter
- International Agency for Research on Cancer, Lyon, France
| | - Nathalie Michels
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| |
Collapse
|
214
|
Ntemiri A, Ghosh TS, Gheller ME, Tran TTT, Blum JE, Pellanda P, Vlckova K, Neto MC, Howell A, Thalacker-Mercer A, O’Toole PW. Whole Blueberry and Isolated Polyphenol-Rich Fractions Modulate Specific Gut Microbes in an In Vitro Colon Model and in a Pilot Study in Human Consumers. Nutrients 2020; 12:E2800. [PMID: 32932733 PMCID: PMC7551244 DOI: 10.3390/nu12092800] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Blueberry (BB) consumption is linked to improved health. The bioconversion of the polyphenolic content of BB by fermentative bacteria in the large intestine may be a necessary step for the health benefits attributed to BB consumption. The identification of specific gut microbiota taxa that respond to BB consumption and that mediate the bioconversion of consumed polyphenolic compounds into bioactive forms is required to improve our understanding of how polyphenols impact human health. We tested the ability of polyphenol-rich fractions purified from whole BB-namely, anthocyanins/flavonol glycosides (ANTH/FLAV), proanthocyanidins (PACs), the sugar/acid fraction (S/A), and total polyphenols (TPP)-to modulate the fecal microbiota composition of healthy adults in an in vitro colon system. In a parallel pilot study, we tested the effect of consuming 38 g of freeze-dried BB powder per day for 6 weeks on the fecal microbiota of 17 women in two age groups (i.e., young and older). The BB ingredients had a distinct effect on the fecal microbiota composition in the artificial colon model. The ANTH/FLAV and PAC fractions were more effective in promoting microbiome alpha diversity compared to S/A and TPP, and these effects were attributed to differentially responsive taxa. Dietary enrichment with BB resulted in a moderate increase in the diversity of the microbiota of the older subjects but not in younger subjects, and certain health-relevant taxa were significantly associated with BB consumption. Alterations in the abundance of some gut bacteria correlated not only with BB consumption but also with increased antioxidant activity in blood. Collectively, these pilot data support the notion that BB consumption is associated with gut microbiota changes and health benefits.
Collapse
Affiliation(s)
- Alexandra Ntemiri
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Tarini S. Ghosh
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Molly E. Gheller
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
| | - Tam T. T. Tran
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Jamie E. Blum
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
| | - Paola Pellanda
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Klara Vlckova
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Marta C. Neto
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| | - Amy Howell
- Marucci Center for Blueberry Cranberry Research, Rutgers University, Chatsworth, NJ 08019, USA;
| | - Anna Thalacker-Mercer
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; (M.E.G.); (J.E.B.); (A.T.-M.)
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, AL 35294, USA
| | - Paul W. O’Toole
- School of Microbiology, University College Cork, T12 K8AF Cork, Ireland; (A.N.); (T.S.G.); (T.T.T.T.); (P.P.); (K.V.); (M.C.N.)
- APC Microbiome Ireland, University College Cork, T12 K8AF Cork, Ireland
| |
Collapse
|
215
|
Gut Microbiota Modifications and Weight Regain in Morbidly Obese Women After Roux-en-Y Gastric Bypass. Obes Surg 2020; 30:4958-4966. [PMID: 32915360 DOI: 10.1007/s11695-020-04956-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/01/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
|
216
|
Proffitt C, Bidkhori G, Moyes D, Shoaie S. Disease, Drugs and Dysbiosis: Understanding Microbial Signatures in Metabolic Disease and Medical Interventions. Microorganisms 2020; 8:microorganisms8091381. [PMID: 32916966 PMCID: PMC7565856 DOI: 10.3390/microorganisms8091381] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Since the discovery of the potential role for the gut microbiota in health and disease, many studies have gone on to report its impact in various pathologies. These studies have fuelled interest in the microbiome as a potential new target for treating disease Here, we reviewed the key metabolic diseases, obesity, type 2 diabetes and atherosclerosis and the role of the microbiome in their pathogenesis. In particular, we will discuss disease associated microbial dysbiosis; the shift in the microbiome caused by medical interventions and the altered metabolite levels between diseases and interventions. The microbial dysbiosis seen was compared between diseases including Crohn’s disease and ulcerative colitis, non-alcoholic fatty liver disease, liver cirrhosis and neurodegenerative diseases, Alzheimer’s and Parkinson’s. This review highlights the commonalities and differences in dysbiosis of the gut between diseases, along with metabolite levels in metabolic disease vs. the levels reported after an intervention. We identify the need for further analysis using systems biology approaches and discuss the potential need for treatments to consider their impact on the microbiome.
Collapse
Affiliation(s)
- Ceri Proffitt
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Correspondence: (C.P.); (S.S.)
| | - Gholamreza Bidkhori
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - David Moyes
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
| | - Saeed Shoaie
- Centre for Host–Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK; (G.B.); (D.M.)
- Science for Life Laboratory, Department of Protein Science, KTH Royal Institute of Technology, 114 17 Stockholm, Sweden
- Correspondence: (C.P.); (S.S.)
| |
Collapse
|
217
|
Swierz MJ, Storman D, Staskiewicz W, Gorecka M, Jasinska KW, Swierz AM, Tobola P, Skuza A, Bala MM. Efficacy of probiotics in patients with morbid obesity undergoing bariatric surgery: a systematic review and meta-analysis. Surg Obes Relat Dis 2020; 16:2105-2116. [PMID: 33069600 DOI: 10.1016/j.soard.2020.08.038] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 12/15/2022]
Abstract
Bariatric surgery is considered effective for morbid obesity, and probiotic supplementation might provide some benefits. We aimed to revise the evidence regarding probiotic supplementation in patients with morbid obesity undergoing bariatric surgery. MEDLINE, Embase, Web of Science, CENTRAL, and trial registers were searched up to April 1, 2020. We included randomized controlled trials and controlled clinical trials, and outcomes of interest were weight change, quality of life, gastrointestinal symptoms, and adverse events. All stages of the review were done by 2 authors independently and we followed Cochrane Handbook guidance. We screened 2541 references and included 5 studies. Probiotics may have minor to no effect regarding percentage excess weight loss (%EWL) at 6 weeks (mean difference [MD], .28; 95% CI, -9.53 to 10.09; 44 participants, 2 studies), 3 months (MD, 5.47; 95% CI, -3.22 to 14.17; 165 participants, 3 studies), 6 months (MD, .46; 95% CI, -8.14 to 9.07; 115 participants, 2 studies), and 12 months post surgery (MD, .35; 95% CI, -8.66 to 9.37; 123 participants, 2 studies). We observed short-term improvement in gastrointestinal symptoms. There was no important effect on quality of life and no meaningful adverse events. Because probiotic supplementation might provide some benefit with respect to weight loss, might alleviate some gastrointestinal symptoms, and is associated with minor or no adverse events, continuous supplementation might be worth considering in certain individuals. Our findings are based on the body of evidence of very low certainty, and further well-designed randomized controlled trials are required to elucidate the effect and strengthen the certainty in the estimates.
Collapse
Affiliation(s)
- Mateusz J Swierz
- Chair of Epidemiology and Preventive Medicine, Department of Hygiene and Dietetics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Dawid Storman
- Chair of Epidemiology and Preventive Medicine, Department of Hygiene and Dietetics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Wojciech Staskiewicz
- Students' Scientific Research Group of Systematic Reviews, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Gorecka
- Students' Scientific Research Group of Systematic Reviews, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Katarzyna W Jasinska
- Students' Scientific Research Group of Systematic Reviews, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | | | - Paulina Tobola
- Chair of Epidemiology and Preventive Medicine, Department of Hygiene and Dietetics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Skuza
- Students' Scientific Research Group of Systematic Reviews, Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland
| | - Malgorzata M Bala
- Chair of Epidemiology and Preventive Medicine, Department of Hygiene and Dietetics, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland; Systematic Reviews Unit, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
218
|
Jacobson R, Terranella S, Booker C, Khalid S, Torquati A, Omotosho P. The Impact of Perioperative Antibiotic Prophylaxis on Weight Loss Following Laparoscopic Sleeve Gastrectomy. J Laparoendosc Adv Surg Tech A 2020; 30:998-1000. [DOI: 10.1089/lap.2020.0449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Richard Jacobson
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Samantha Terranella
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Connor Booker
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Syed Khalid
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Alfonso Torquati
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| | - Philip Omotosho
- Department of Surgery, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
219
|
Recent advances in the mechanisms underlying the beneficial effects of bariatric and metabolic surgery. Surg Obes Relat Dis 2020; 17:231-238. [PMID: 33036939 DOI: 10.1016/j.soard.2020.08.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/11/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
Bariatric and metabolic surgery (BMS) is the most effective treatment for obesity, type 2 diabetes and co-morbidities, including nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. The beneficial effects of BMS are beyond the primary goal of gastric restriction and nutrients malabsorption. Roux-en-Y gastric bypass and vertical sleeve gastrectomy are the 2 most commonly performed procedures of BMS. Both surgeries lead to physiologic changes in gastrointestinal tract; subsequently alter bile acids pool and composition, gut microbial activities, gut hormones, and circulating exosomes; and ultimately contribute to the improved glycemic control, insulin sensitivity, lipid metabolism, energy expenditure, and weight loss. The mechanisms underlying the benefits of BMS likely involve the bile acid-signaling pathway mediated mainly by nuclear farnesoid X receptor and the membrane Takeda G protein-coupled receptor, bile acids-gut microbiota interaction, and exosomes. In this review, we focus on recent advances in potential mechanisms and aim to learn novel insights into the molecular mechanisms underlying metabolic disorders.
Collapse
|
220
|
Yoshino M, Kayser BD, Yoshino J, Stein RI, Reeds D, Eagon JC, Eckhouse SR, Watrous JD, Jain M, Knight R, Schechtman K, Patterson BW, Klein S. Effects of Diet versus Gastric Bypass on Metabolic Function in Diabetes. N Engl J Med 2020; 383:721-732. [PMID: 32813948 PMCID: PMC7456610 DOI: 10.1056/nejmoa2003697] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Some studies have suggested that in people with type 2 diabetes, Roux-en-Y gastric bypass has therapeutic effects on metabolic function that are independent of weight loss. METHODS We evaluated metabolic regulators of glucose homeostasis before and after matched (approximately 18%) weight loss induced by gastric bypass (surgery group) or diet alone (diet group) in 22 patients with obesity and diabetes. The primary outcome was the change in hepatic insulin sensitivity, assessed by infusion of insulin at low rates (stages 1 and 2 of a 3-stage hyperinsulinemic euglycemic pancreatic clamp). Secondary outcomes were changes in muscle insulin sensitivity, beta-cell function, and 24-hour plasma glucose and insulin profiles. RESULTS Weight loss was associated with increases in mean suppression of glucose production from baseline, by 7.04 μmol per kilogram of fat-free mass per minute (95% confidence interval [CI], 4.74 to 9.33) in the diet group and by 7.02 μmol per kilogram of fat-free mass per minute (95% CI, 3.21 to 10.84) in the surgery group during clamp stage 1, and by 5.39 (95% CI, 2.44 to 8.34) and 5.37 (95% CI, 2.41 to 8.33) μmol per kilogram of fat-free mass per minute in the two groups, respectively, during clamp stage 2; there were no significant differences between the groups. Weight loss was associated with increased insulin-stimulated glucose disposal, from 30.5±15.9 to 61.6±13.0 μmol per kilogram of fat-free mass per minute in the diet group and from 29.4±12.6 to 54.5±10.4 μmol per kilogram of fat-free mass per minute in the surgery group; there was no significant difference between the groups. Weight loss increased beta-cell function (insulin secretion relative to insulin sensitivity) by 1.83 units (95% CI, 1.22 to 2.44) in the diet group and by 1.11 units (95% CI, 0.08 to 2.15) in the surgery group, with no significant difference between the groups, and it decreased the areas under the curve for 24-hour plasma glucose and insulin levels in both groups, with no significant difference between the groups. No major complications occurred in either group. CONCLUSIONS In this study involving patients with obesity and type 2 diabetes, the metabolic benefits of gastric bypass surgery and diet were similar and were apparently related to weight loss itself, with no evident clinically important effects independent of weight loss. (Funded by the National Institutes of Health and others; ClinicalTrials.gov number, NCT02207777.).
Collapse
Affiliation(s)
- Mihoko Yoshino
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Brandon D Kayser
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Jun Yoshino
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Richard I Stein
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Dominic Reeds
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - J Christopher Eagon
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Shaina R Eckhouse
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Jeramie D Watrous
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Mohit Jain
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Rob Knight
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Kenneth Schechtman
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Bruce W Patterson
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| | - Samuel Klein
- From the Center for Human Nutrition (M.Y., B.D.K., J.Y., R.I.S., D.R., K.S., B.W.P., S.K.) and the Department of Surgery (J.C.E., S.R.E.), Washington University School of Medicine, St. Louis; and the Departments of Medicine (J.D.W., M.J.), Pharmacology (J.D.W., M.J.), Pediatrics (R.K.), and Computer Science and Engineering (R.K.), University of California San Diego, San Diego
| |
Collapse
|
221
|
Cook J, Lehne C, Weiland A, Archid R, Ritze Y, Bauer K, Zipfel S, Penders J, Enck P, Mack I. Gut Microbiota, Probiotics and Psychological States and Behaviors after Bariatric Surgery-A Systematic Review of Their Interrelation. Nutrients 2020; 12:nu12082396. [PMID: 32785153 PMCID: PMC7468806 DOI: 10.3390/nu12082396] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
The gastrointestinal (GI) microbiota plays an important role in health and disease, including brain function and behavior. Bariatric surgery (BS) has been reported to result in various changes in the GI microbiota, therefore demanding the investigation of the impact of GI microbiota on treatment success. The goal of this systematic review was to assess the effects of BS on the microbiota composition in humans and other vertebrates, whether probiotics influence postoperative health, and whether microbiota and psychological and behavioral factors interact. A search was conducted using PubMed and Web of Science to find relevant studies with respect to the GI microbiota and probiotics after BS, and later screened for psychological and behavioral parameters. Studies were classified into groups and subgroups to provide a clear overview of the outcomes. Microbiota changes were further assessed for whether they were specific to BS in humans through the comparison to sham operated controls in other vertebrate studies. Changes in alpha diversity appear not to be specific, whereas dissimilarity in overall microbial community structure, and increases in the abundance of the phylum Proteobacteria and Akkermansia spp. within the phylum Verrucomicrobia after surgery were observed in both human and other vertebrates studies and may be specific to BS in humans. Human probiotic studies differed regarding probiotic strains and dosages, however it appeared that probiotic interventions were not superior to a placebo for quality of life scores or weight loss after BS. The relationship between GI microbiota and psychological diseases in this context is unclear due to insufficient available data.
Collapse
Affiliation(s)
- Jessica Cook
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - Christine Lehne
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - Alisa Weiland
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - Rami Archid
- Department of General, Visceral and Transplant Surgery, University Hospital, 72072 Tübingen, Germany;
| | - Yvonne Ritze
- Institute for Medical Psychology and Behavioral Neurobiology, University Hospital, 72072 Tübingen, Germany;
| | - Kerstin Bauer
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - Stephan Zipfel
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - John Penders
- Department of Medical Microbiology, School of Nutrition and Translational Research in Metabolism (NUTRIM) and Care and Public Health Research Institute(Caphri), Maastricht University Medical Centre, 6211 Maastricht, The Netherlands;
| | - Paul Enck
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
| | - Isabelle Mack
- Department of Psychosomatic Medicine and Psychotherapy, University Medical Hospital, 72072 Tübingen, Germany; (J.C.); (C.L.); (A.W.); (K.B.); (S.Z.); (P.E.)
- Correspondence: ; Tel.: +49-7071-2985614; Fax: +49-7071-294382
| |
Collapse
|
222
|
Morales-Marroquin E, Hanson B, Greathouse L, de la Cruz-Munoz N, Messiah SE. Comparison of methodological approaches to human gut microbiota changes in response to metabolic and bariatric surgery: A systematic review. Obes Rev 2020; 21:e13025. [PMID: 32249534 DOI: 10.1111/obr.13025] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 02/07/2023]
Abstract
Substantial differences in the response of gut microbial composition to metabolic and bariatric surgery have been reported. Therefore, the goal of the present review is to evaluate if methodological differences could be driving this lack of consistency. A search was conducted using PUBMED, Web of Science, Science Direct and COCHRANE using the following inclusion criteria: human studies written in English with a baseline sampling point, using gut microbiota as an outcome and either Roux-n-Y gastric bypass or sleeve gastrectomy. Sixteen articles were selected (total 221 participants). Roux-n-Y gastric bypass caused more alterations in gut microbial composition in comparison with sleeve gastrectomy. Substantial variability was found in study designs, data collection and analyses across studies. Increases in several families and genera from the phylum Proteobacteria and Bacteroidetes, the family Streptococcaceae, the species Akkermansia muciniphila and Streptococcus salivarius and a decrease in the phylum Firmicutes and the family Bifidobacteriaceae were reported. There is a need for standardization not only of microbial analysis but also of study designs when analysing the effect of bariatric surgery on the human gut microbiome. In addition, outcomes from different surgical procedures should not be combined as they produce distinctive effects on gut microbial composition.
Collapse
Affiliation(s)
- Elisa Morales-Marroquin
- School of Public Health, Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center, Texas, USA
| | - Blake Hanson
- School of Public Health, Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center, Texas, USA
| | - Leigh Greathouse
- Robbins College of Health and Human Services, Baylor University Waco, Texas, USA
| | | | - Sarah E Messiah
- School of Public Health, Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center, Texas, USA
| |
Collapse
|
223
|
Erawijantari PP, Mizutani S, Shiroma H, Shiba S, Nakajima T, Sakamoto T, Saito Y, Fukuda S, Yachida S, Yamada T. Influence of gastrectomy for gastric cancer treatment on faecal microbiome and metabolome profiles. Gut 2020; 69:1404-1415. [PMID: 31953253 PMCID: PMC7398469 DOI: 10.1136/gutjnl-2019-319188] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/26/2019] [Accepted: 12/02/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Recent evidence points to the gut microbiome's involvement in postoperative outcomes, including after gastrectomy. Here, we investigated the influence of gastrectomy for gastric cancer on the gut microbiome and metabolome, and how it related to postgastrectomy conditions. DESIGN We performed shotgun metagenomics sequencing and capillary electrophoresis time-of-flight mass spectrometry-based metabolomics analyses on faecal samples collected from participants with a history of gastrectomy for gastric cancer (n=50) and compared them with control participants (n=56). RESULTS The gut microbiota in the gastrectomy group showed higher species diversity and richness (p<0.05), together with greater abundance of aerobes, facultative anaerobes and oral microbes. Moreover, bile acids such as genotoxic deoxycholic acid and branched-chain amino acids were differentially abundant between the two groups (linear discriminant analysis (LDA) effect size (LEfSe): p<0.05, q<0.1, LDA>2.0), as were also Kyoto Encyclopedia of Genes and Genomes modules involved in nutrient transport and organic compounds biosynthesis (LEfSe: p<0.05, q<0.1, LDA>2.0). CONCLUSION Our results reveal alterations of gut microbiota after gastrectomy, suggesting its association with postoperative comorbidities. The multi-omic approach applied in this study could complement the follow-up of patients after gastrectomy.
Collapse
Affiliation(s)
- Pande Putu Erawijantari
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| | - Sayaka Mizutani
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Hirotsugu Shiroma
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| | - Satoshi Shiba
- Division of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Takeshi Nakajima
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Taku Sakamoto
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Yutaka Saito
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Tokyo, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan,Intestinal Microbiota Project, Kanagawa Institute of Industrial Science and Technology, Ebina, Kanagawa, Japan,Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| | - Shinichi Yachida
- Division of Cancer Genomics, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan .,Department of Cancer Genome Informatics, Graduate School of Medicine/Faculty of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takuji Yamada
- School of Life Science and Technology, Tokyo Institute of Technology, Meguro-ku, Tokyo, Japan
| |
Collapse
|
224
|
Qualls-Creekmore E, Marlatt KL, Aarts E, Bruce-Keller A, Church TS, Clément K, Fisher JO, Gordon-Larsen P, Morrison CD, Raybould HE, Ryan DH, Schauer PR, Spector AC, Spetter MS, Stuber GD, Berthoud HR, Ravussin E. What Should I Eat and Why? The Environmental, Genetic, and Behavioral Determinants of Food Choice: Summary from a Pennington Scientific Symposium. Obesity (Silver Spring) 2020; 28:1386-1396. [PMID: 32520444 PMCID: PMC7501251 DOI: 10.1002/oby.22806] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/29/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
This review details the proceedings of a Pennington Biomedical scientific symposium titled, "What Should I Eat and Why? The Environmental, Genetic, and Behavioral Determinants of Food Choice." The symposium was designed to review the literature about energy homeostasis, particularly related to food choice and feeding behaviors, from psychology to physiology. This review discusses the intrinsic determinants of food choice, including biological mechanisms (genetics), peripheral and central signals, brain correlates, and the potential role of the microbiome. This review also address the extrinsic determinants (environment) of food choice within our physical and social environments. Finally, this review reports the current treatment practices for the clinical management of eating-induced overweight and obesity. An improved understanding of these determinants will inform best practices for the clinical treatment and prevention of obesity. Strategies paired with systemic shifts in our public health policies and changes in our "obesogenic" environment will be most effective at attenuating the obesity epidemic.
Collapse
Affiliation(s)
- Emily Qualls-Creekmore
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Kara L. Marlatt
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Esther Aarts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Annadora Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Tim S. Church
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
- ACAP Health, Dallas, TX, USA
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesity: Systemic Approaches (NutriOmics) Research Unit, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition e, Pitié-Salpêtrière Hospital, 47-83 bd de l’Hôpital, Paris, France
| | - Jennifer O. Fisher
- Center for Obesity Research and Education, Temple University, Philadelphia, PA, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher D. Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Helen E. Raybould
- Department of Anatomy, Physiology, Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Donna H. Ryan
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Philip R. Schauer
- Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Maartje S. Spetter
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Germany; School of Psychology, University of Birmingham, Edgbaston, Birmingham, UK
| | - Garret D. Stuber
- Departments of Anesthesiology, Pain Medicine & Pharmacology, University of Washington, Seattle, WA, USA
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
225
|
Sookoian S, Salatino A, Castaño GO, Landa MS, Fijalkowky C, Garaycoechea M, Pirola CJ. Intrahepatic bacterial metataxonomic signature in non-alcoholic fatty liver disease. Gut 2020; 69:1483-1491. [PMID: 31900291 DOI: 10.1136/gutjnl-2019-318811] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 12/12/2019] [Accepted: 12/16/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE We aimed to characterise the liver tissue bacterial metataxonomic signature in two independent cohorts of patients with biopsy-proven non-alcoholic fatty liver disease (NAFLD) diagnosis, as differences in the host phenotypic features-from moderate to severe obesity-may be associated with significant changes in the microbial DNA profile. DESIGN AND METHODS Liver tissue samples from 116 individuals, comprising of 47 NAFLD overweight or moderately obese patients, 50 NAFLD morbidly obese patients elected for bariatric surgery and 19 controls, were analysed using high-throughput 16S rRNA gene sequencing. RESULTS Liver bacterial DNA profile significantly differs between morbidly obese and non-morbidly obese patients with NAFLD. Bacteroidetes (p=1.8e-18) and Firmicutes (p=0.0044) were over-represented in morbidly obese patients and Proteobacteria (p=5.2e-10)-specifically Gammaproteobacteria and Alphaproteobacteria, and Deinococcus-Thermus (p=0.00012)-were over-represented in the non-morbidly obese cohort. Cohort-specific analysis of liver microbial DNA signatures shows patterns linked to obesity. The imbalance in Proteobacteria (Alpha or Gamma) among non-morbidly obese patients, and Peptostreptococcaceae, Verrucomicrobia, Actinobacteria and Gamma Proteobacteria DNA among morbidly obese patients was associated with histological severity. Decreased amounts of bacterial DNA from the Lachnospiraceae family were associated with more severe histological features. Proteobacteria DNA was consistently associated with lobular and portal inflammation scores. Microbial DNA composition corresponded to predicted functional differences. CONCLUSION This is the first comprehensive study showing that the liver tissue of NAFLD patients contains a diverse repertoire of bacterial DNA (up to 2.5×104 read counts). The liver metataxonomic signature may explain differences in the NAFLD pathogenic mechanisms as well as physiological functions of the host.
Collapse
Affiliation(s)
- Silvia Sookoian
- Institute of Medical Research A Lanari, University of Buenos Aires Faculty of Medicine, Buenos Aires, Argentina .,Institute of Medical Research (IDIM), Department of Clinical and Molecular Hepatology, National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Adrian Salatino
- Institute of Medical Research A Lanari, University of Buenos Aires Faculty of Medicine, Buenos Aires, Argentina.,Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Gustavo Osvaldo Castaño
- Liver Unit, Medicine and Surgery Department, Hospital General de Agudos Dr Abel Zubizarreta, Buenos Aires, Argentina
| | - Maria Silvia Landa
- Institute of Medical Research A Lanari, University of Buenos Aires Faculty of Medicine, Buenos Aires, Argentina.,Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council, Buenos Aires, Argentina
| | - Cinthia Fijalkowky
- Institute of Medical Research A Lanari, University of Buenos Aires Faculty of Medicine, Buenos Aires, Argentina.,Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council, Buenos Aires, Argentina
| | | | - Carlos Jose Pirola
- Institute of Medical Research A Lanari, University of Buenos Aires Faculty of Medicine, Buenos Aires, Argentina .,Institute of Medical Research (IDIM), Department of Molecular Genetics and Biology of Complex Diseases, National Scientific and Technical Research Council, Buenos Aires, Argentina
| |
Collapse
|
226
|
Steinert RE, Rehman A, Souto Lima EJ, Agamennone V, Schuren FHJ, Gero D, Schreiner P, Vonlanthen R, Ismaeil A, Tzafos S, Hosa H, Vetter D, Misselwitz B, Bueter M. Roux-en-Y gastric bypass surgery changes fungal and bacterial microbiota in morbidly obese patients-A pilot study. PLoS One 2020; 15:e0236936. [PMID: 32735609 PMCID: PMC7394366 DOI: 10.1371/journal.pone.0236936] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023] Open
Abstract
The Roux-en-Y gastric bypass (RYGB) remains the most effective treatment for morbidly obese patients to lower body weight and improve glycemic control. There is recent evidence that the mycobiome (fungal microbiome) can aggravate disease severity in a number of diseases including inflammatory bowel disease (IBD), irritable bowel syndrome (IBS) and hepatitis; moreover, a dysbiotic fungal microbiota has been reported in the obese. We characterized fungal and bacterial microbial composition in fecal samples of 16 morbidly obese patients before and three months after RYGB surgery and compared with nine healthy controls. We found that RYGB surgery induced a clear alteration in structure and composition of the gut fungal and bacterial microbiota. Beta diversity analysis revealed significant differences in bacterial microbiota between obese patients before surgery and healthy controls (P < 0.005) and a significant, unidirectional shift in RYGB patients after surgery (P < 0.001 vs. before surgery). In contrast, there was no significant difference in fungal microbiota between groups but individually specific changes after RYGB surgery. Interestingly, RYGB surgery induced a significant reduction in fungal alpha diversity namely Chao1, Sobs, and Shannon diversity index (P<0.05, respectively) which contrasts the trend for uniform changes in bacteria towards increased richness and diversity post-surgery. We did not observe any inter-kingdom relations in RYGB patients but in the healthy control cohort and there were several correlations between fungi and bacteria and clinical parameters (P<0.05, respectively) that warrant further research. Our study identifies changes in intestinal fungal communities in RYGB patients that are distinct to changes in the bacterial microbiota.
Collapse
Affiliation(s)
- Robert E. Steinert
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
- * E-mail:
| | - Ateequr Rehman
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Everton Job Souto Lima
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Valeria Agamennone
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Frank H. J. Schuren
- Microbiology and Systems Biology, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Daniel Gero
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Phillip Schreiner
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - René Vonlanthen
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Aiman Ismaeil
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
- Department of Surgery, Aswan University, Tingar, Egypt
| | - Stefanos Tzafos
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Hanna Hosa
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Diana Vetter
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Benjamin Misselwitz
- Division of Visceral Surgery and Medicine, Inselspital Bern and Bern University, Bern, Switzerland
| | - Marco Bueter
- Department of Surgery, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
227
|
Obesity and Cardio-vascular Disorders: Review. Fam Med 2020. [DOI: 10.30841/2307-5112.3.2020.211860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
228
|
Marzullo P, Di Renzo L, Pugliese G, De Siena M, Barrea L, Muscogiuri G, Colao A, Savastano S. From obesity through gut microbiota to cardiovascular diseases: a dangerous journey. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:35-49. [PMID: 32714511 PMCID: PMC7371682 DOI: 10.1038/s41367-020-0017-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The co-existence of humans and gut microbiota started millions of years ago. Until now, a balance gradually developed between gut bacteria and their hosts. It is now recognized that gut microbiota are key to form adequate immune and metabolic functions and, more in general, for the maintenance of good health. Gut microbiota are established before birth under the influence of maternal nutrition and metabolic status, which can impact the future metabolic risk of the offspring in terms of obesity, diabetes, and cardiometabolic disorders during the lifespan. Obesity and diabetes are prone to disrupt the gut microbiota and alter the gut barrier permeability, leading to metabolic endotoxaemia with its detrimental consequences on health. Specific bacterial sequences are now viewed as peculiar signatures of the metabolic syndrome across life stages in each individual, and are linked to pathogenesis of cardiovascular diseases (CVDs) via metabolic products (metabolites) and immune modulation. These mechanisms have been linked, in association with abnormalities in microbial richness and diversity, to an increased risk of developing arterial hypertension, systemic inflammation, nonalcoholic fatty liver disease, coronary artery disease, chronic kidney disease, and heart failure. Emerging strategies for the manipulation of intestinal microbiota represent a promising therapeutic option for the prevention and treatment of CVD especially in individuals prone to CV events.
Collapse
Affiliation(s)
- Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
| | - Gabriella Pugliese
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Martina De Siena
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luigi Barrea
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Giovanna Muscogiuri
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Annamaria Colao
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Silvia Savastano
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - on behalf of Obesity Programs of nutrition, Education, Research and Assessment (OPERA) Group
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
229
|
Olsson LM, Poitou C, Tremaroli V, Coupaye M, Aron-Wisnewsky J, Bäckhed F, Clément K, Caesar R. Gut microbiota of obese subjects with Prader-Willi syndrome is linked to metabolic health. Gut 2020; 69:1229-1238. [PMID: 31611297 PMCID: PMC7306984 DOI: 10.1136/gutjnl-2019-319322] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The gut microbiota has been implicated in the aetiology of obesity and associated comorbidities. Patients with Prader-Willi syndrome (PWS) are obese but partly protected against insulin resistance. We hypothesised that the gut microbiota of PWS patients differs from that of non-genetically obese controls and correlate to metabolic health. Therefore, here we used PWS as a model to study the role of gut microbiota in the prevention of metabolic complications linked to obesity. DESIGN We conducted a case-control study with 17 adult PWS patients and 17 obese subjects matched for body fat mass index, gender and age. The subjects were metabolically characterised and faecal microbiota was profiled by 16S ribosomal RNA gene sequencing. The patients' parents were used as a non-obese control group. Stool samples from two PWS patients and two obese controls were used for faecal microbiota transplantations in germ-free mice to examine the impact of the microbiota on glucose metabolism. RESULTS The composition of the faecal microbiota in patients with PWS differed from that of obese controls, and was characterised by higher phylogenetic diversity and increased abundance of several taxa such as Akkermansia, Desulfovibrio and Archaea, and decreased abundance of Dorea. Microbial taxa prevalent in the PWS microbiota were associated with markers of insulin sensitivity. Improved insulin resistance of PWS was partly transmitted by faecal microbiota transplantations into germ-free mice. CONCLUSION The gut microbiota of PWS patients is similar to that of their non-obese parents and might play a role for the protection of PWS patients from metabolic complications.
Collapse
Affiliation(s)
- Lisa M Olsson
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christine Poitou
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Muriel Coupaye
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Judith Aron-Wisnewsky
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karine Clément
- INSERM, Nutrition and obesities; systemic approaches (NutriOmics) research Unit, Sorbonne Universite, Paris, Île-de-France, France
- Assistance Publique-Hôpitaux de Paris, Reference Center for Rare Diseases (Prader-Willi Syndrome), Nutrition Department, University Hospital Pitié Salpêtrière, Paris, Île-de-France, France
| | - Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
230
|
Aoun A, Darwish F, Hamod N. The Influence of the Gut Microbiome on Obesity in Adults and the Role of Probiotics, Prebiotics, and Synbiotics for Weight Loss. Prev Nutr Food Sci 2020; 25:113-123. [PMID: 32676461 PMCID: PMC7333005 DOI: 10.3746/pnf.2020.25.2.113] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/08/2020] [Indexed: 12/23/2022] Open
Abstract
The link between the gut microbiome and obesity is not well defined. Understanding of the role of the gut microbiome in weight and health management may lead to future revolutionary changes for treating obesity. This review examined the relationship between obesity and the gut microbiome, and the role of probiotics, prebiotics, and synbiotics for preventing and treating obesity. We used PubMed and Google Scholar to collect appropriate articles for the review. We showed that the gut microbiome has an impact on nutrient metabolism and energy expenditure. Moreover, different modalities of obesity treatment have been shown to change the diversity and composition of the gut microbiome; this raises questions about the role these changes may play in weight loss. In addition, studies have shown that supplementation with probiotics, prebiotics, and synbiotics may alter the secretion of hormones, neurotransmitters, and inflammatory factors, thus preventing food intake triggers that lead to weight gain. Further clinical studies are needed to better understand how different species of bacteria in the gut microbiome may affect weight gain, and to determine the most appropriate doses, compositions, and regimens of probiotics, prebiotics, and synbiotics supplementation for long-term weight control.
Collapse
Affiliation(s)
- Antoine Aoun
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| | - Fatima Darwish
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| | - Natacha Hamod
- Faculty of Nursing and Health Sciences, Notre Dame University-Louaize, Zouk Mosbeh 72, Lebanon
| |
Collapse
|
231
|
Yu D, Shu XO, Howard EF, Long J, English WJ, Flynn CR. Fecal metagenomics and metabolomics reveal gut microbial changes after bariatric surgery. Surg Obes Relat Dis 2020; 16:1772-1782. [PMID: 32747219 DOI: 10.1016/j.soard.2020.06.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/06/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Evidence from longitudinal patient studies regarding gut microbial changes after bariatric surgery is limited. OBJECTIVE To examine intraindividual changes in fecal microbiome and metabolites among patients undergoing Roux-en-Y gastric bypass or vertical sleeve gastrectomy. SETTING Observational study. METHODS Twenty patients were enrolled and provided stool samples before and 1 week, 1 month, and/or 3 months after surgery. Shallow shotgun metagenomics and untargeted fecal metabolomics were performed. Zero-inflated generalized additive models and linear mixed models were applied to identify fecal microbiome and metabolites changes, with adjustment for potential confounders and correction for multiple testing. RESULTS We enrolled 16 women and 4 men, including 16 white and 4 black participants (median age = 45 years; presurgery body mass index = 47.7 kg/m2). Ten patients had Roux-en-Y gastric bypass, 10 had vertical sleeve gastrectomy, and 14 patients provided postsurgery stool samples. Of 47 samples, median sequencing depth was 6.3 million reads and 1073 metabolites were identified. Microbiome alpha-diversity increased after surgery, especially at 3 months. Significant genus-level changes included increases in Odoribacter, Streptococcus, Anaerotruncus, Alistipes, Klebsiella, and Bifidobacterium, while decreases in Bacteroides, Coprocosccus, Dorea, and Faecalibacterium. Large increases in Streptococcus, Akkermansia, and Prevotella were observed at 3 months. Beta-diversity and fecal metabolites were also changed, including reduced caffeine metabolites, indoles, and butyrate. CONCLUSIONS Despite small sample size and missing repeated samples in some participants, our pilot study showed significant postsurgery changes in fecal microbiome and metabolites among bariatric surgery patients. Future large-scale, longitudinal studies are warranted to investigate gut microbial changes and their associations with metabolic outcomes after bariatric surgery.
Collapse
Affiliation(s)
- Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Eric F Howard
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Wayne J English
- Division of General Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charles R Flynn
- Division of General Surgery, Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
232
|
Revealing links between gut microbiome and its fungal community in Type 2 Diabetes Mellitus among Emirati subjects: A pilot study. Sci Rep 2020; 10:9624. [PMID: 32541680 PMCID: PMC7295773 DOI: 10.1038/s41598-020-66598-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/22/2020] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) drastically affects the population of Middle East countries with an ever-increasing number of overweight and obese individuals. The precise links between T2DM and gut microbiome composition remain elusive in these populations. Here, we performed 16 S rRNA and ITS2- gene based microbial profiling of 50 stool samples from Emirati adults with or without T2DM. The four major enterotypes initially described in westernized cohorts were retrieved in this Emirati population. T2DM and non-T2DM healthy controls had different microbiome compositions, with an enrichment in Prevotella enterotype in non-T2DM controls whereas T2DM individuals had a higher proportion of the dysbiotic Bacteroides 2 enterotype. No significant differences in microbial diversity were observed in T2DM individuals after controlling for cofounding factors, contrasting with reports from westernized cohorts. Interestingly, fungal diversity was significantly decreased in Bacteroides 2 enterotype. Functional profiling from 16 S rRNA gene data showed marked differences between T2DM and non-T2DM controls, with an enrichment in amino acid degradation and LPS-related modules in T2DM individuals, whereas non-T2DM controls had increased abundance of carbohydrate degradation modules in concordance with enterotype composition. These differences provide an insight into gut microbiome composition in Emirati population and its potential role in the development of diabetes mellitus.
Collapse
|
233
|
Redondo-Useros N, Nova E, González-Zancada N, Díaz LE, Gómez-Martínez S, Marcos A. Microbiota and Lifestyle: A Special Focus on Diet. Nutrients 2020; 12:E1776. [PMID: 32549225 PMCID: PMC7353459 DOI: 10.3390/nu12061776] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023] Open
Abstract
It is widely known that a good balance and healthy function for bacteria groups in the colon are necessary to maintain homeostasis and preserve health. However, the lack of consensus on what defines a healthy gut microbiota and the multitude of factors that influence human gut microbiota composition complicate the development of appropriate dietary recommendations for our gut microbiota. Furthermore, the varied response to the intake of probiotics and prebiotics observed in healthy adults suggests the existence of potential inter- and intra-individual factors, which might account for gut microbiota changes to a greater extent than diet. The changing dietary habits worldwide involving consumption of processed foods containing artificial ingredients, such as sweeteners; the coincident rise in emotional disorders; and the worsening of other lifestyle habits, such as smoking habits, drug consumption, and sleep, can together contribute to gut dysbiosis and health impairment, as well as the development of chronic diseases. This review summarizes the current literature on the effects of specific dietary ingredients (probiotics, prebiotics, alcohol, refined sugars and sweeteners, fats) in the gut microbiota of healthy adults and the potential inter- and intra-individual factors involved, as well as the influence of other potential lifestyle factors that are dramatically increasing nowadays.
Collapse
Affiliation(s)
| | | | | | | | | | - Ascensión Marcos
- Immunonutrition Group, Department of Metabolism and Nutrition, Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), Jose Antonio Novais, St.10, 28040 Madrid, Spain; (N.R.-U.); (E.N.); (N.G.-Z.); (L.E.D.); (S.G.-M.)
| |
Collapse
|
234
|
Abstract
Abstract
Collapse
Affiliation(s)
- Nicholas Finer
- National Centre for Cardiovascular Prevention and Outcomes, UCL Institute of Cardiovascular Science, University College London, UK
| |
Collapse
|
235
|
Affiliation(s)
- Patrice D Cani
- WELBIO - Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, UCLouvain, Université Catholique de Louvain , Brussels, Belgium
| | - Matthias Van Hul
- WELBIO - Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Metabolism and Nutrition Research Group, UCLouvain, Université Catholique de Louvain , Brussels, Belgium
| |
Collapse
|
236
|
Le transfert de microbiote fécal : quel potentiel thérapeutique dans le traitement des maladies métaboliques ? NUTR CLIN METAB 2020. [DOI: 10.1016/j.nupar.2019.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
237
|
Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol 2020; 17:279-297. [PMID: 32152478 DOI: 10.1038/s41575-020-0269-9] [Citation(s) in RCA: 648] [Impact Index Per Article: 129.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota dysbiosis has been repeatedly observed in obesity and type 2 diabetes mellitus, two metabolic diseases strongly intertwined with non-alcoholic fatty liver disease (NAFLD). Animal studies have demonstrated a potential causal role of gut microbiota in NAFLD. Human studies have started to describe microbiota alterations in NAFLD and have found a few consistent microbiome signatures discriminating healthy individuals from those with NAFLD, non-alcoholic steatohepatitis or cirrhosis. However, patients with NAFLD often present with obesity and/or insulin resistance and type 2 diabetes mellitus, and these metabolic confounding factors for dysbiosis have not always been considered. Patients with different NAFLD severity stages often present with heterogeneous lesions and variable demographic characteristics (including age, sex and ethnicity), which are known to affect the gut microbiome and have been overlooked in most studies. Finally, multiple gut microbiome sequencing tools and NAFLD diagnostic methods have been used across studies that could account for discrepant microbiome signatures. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling these signatures from underlying metabolic disorders. More advanced metagenomics and multi-omics studies using system biology approaches are needed to improve microbiome biomarkers.
Collapse
|
238
|
Aron-Wisnewsky J, Warmbrunn MV, Nieuwdorp M, Clément K. Nonalcoholic Fatty Liver Disease: Modulating Gut Microbiota to Improve Severity? Gastroenterology 2020; 158:1881-1898. [PMID: 32044317 DOI: 10.1053/j.gastro.2020.01.049] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Gut microbiota plays a role in the pathophysiology of metabolic diseases, which include nonalcoholic fatty liver diseases, through the gut-liver axis. To date, clinical guidelines recommend a weight loss goal of 7%-10% to improve features of nonalcoholic fatty liver diseases. Because this target is not easily achieved by all patients, alternative therapeutic options are currently being evaluated. This review focuses on therapeutics that aim to modulate the gut microbiota and the gut-liver axis. We discuss how probiotics, prebiotics, synbiotic, fecal microbiota transfer, polyphenols, specific diets, and exercise interventions have been found to modify gut microbiota signatures; improve nonalcoholic fatty liver disease outcomes; and detail, when available, the different mechanisms by which these beneficial outcomes might occur. Apart from probiotics that have already been tested in human randomized controlled trials, most of these potential therapeutics have been studied in animals. Their efficacy still warrants confirmation in humans using appropriate design.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Sorbonne Université, INSERM, UMRS U1269, Nutriomics Research Unit, Paris, France; Nutrition Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centre de Recherche en Nutrition Humaine d'Ile de France, Paris, France; Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands.
| | - Moritz V Warmbrunn
- Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands; Department of Internal Medicine, University of Amsterdam Medical Center, Free University, Amsterdam, The Netherlands
| | - Karine Clément
- Sorbonne Université, INSERM, UMRS U1269, Nutriomics Research Unit, Paris, France; Nutrition Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centre de Recherche en Nutrition Humaine d'Ile de France, Paris, France.
| |
Collapse
|
239
|
Postal BG, Ghezzal S, Aguanno D, André S, Garbin K, Genser L, Brot-Laroche E, Poitou C, Soula H, Leturque A, Clément K, Carrière V. AhR activation defends gut barrier integrity against damage occurring in obesity. Mol Metab 2020; 39:101007. [PMID: 32360426 PMCID: PMC7267728 DOI: 10.1016/j.molmet.2020.101007] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/03/2020] [Accepted: 04/22/2020] [Indexed: 12/24/2022] Open
Abstract
Objective Obesity is characterized by systemic and low-grade tissue inflammation. In the intestine, alteration of the intestinal barrier and accumulation of inflammatory cells in the epithelium are important contributors of gut inflammation. Recent studies demonstrated the role of the aryl hydrocarbon receptor (AhR) in the maintenance of immune cells at mucosal barrier sites. A wide range of ligands of external and local origin can activate this receptor. We studied the causal relationship between AhR activation and gut inflammation in obesity. Methods Jejunum samples from subjects with normal weight and severe obesity were phenotyped according to T lymphocyte infiltration in the epithelium from lamina propria and assayed for the mRNA level of AhR target genes. The effect of an AhR agonist was studied in mice and Caco-2/TC7 cells. AhR target gene expression, permeability to small molecules and ions, and location of cell-cell junction proteins were recorded under conditions of altered intestinal permeability. Results We showed that a low AhR tone correlated with a high inflammatory score in the intestinal epithelium in severe human obesity. Moreover, AhR activation protected junctional complexes in the intestinal epithelium in mice challenged by an oral lipid load. AhR ligands prevented chemically induced damage to barrier integrity and cytokine expression in Caco-2/TC7 cells. The PKC and p38MAPK signaling pathways were involved in this AhR action. Conclusions The results of these series of human, mouse, and cell culture experiments demonstrate the protective effect of AhR activation in the intestine targeting particularly tight junctions and cytokine expression. We propose that AhR constitutes a valuable target to protect intestinal functions in metabolic diseases, which can be achieved in the future via food or drug ligands. Intestinal inflammation in obese subjects correlates with low AhR activity. AhR activation protects intestinal epithelium from damages. AhR activation prevents cytokine induction in response to damages. Protective role of AhR activation targets junctional complexes. Protein kinases-dependent signaling pathways are involved in AhR action.
Collapse
Affiliation(s)
- Bárbara G Postal
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France
| | - Sara Ghezzal
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France
| | - Doriane Aguanno
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France; EPHE, PSL Research University, F-75006, Paris, France
| | - Sébastien André
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Kevin Garbin
- CHIC Platform of Cordeliers Research Center, Sorbonne University, UPD Univ Paris 05, INSERM, F-75006, Paris, France
| | - Laurent Genser
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Edith Brot-Laroche
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France
| | - Christine Poitou
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, CRNH Ile de France, F-75013, Paris, France
| | - Hédi Soula
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Armelle Leturque
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France
| | - Karine Clément
- Sorbonne University, INSERM, NutriOmics Research Unit Paris, F-75013, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Nutrition Department, CRNH Ile de France, F-75013, Paris, France
| | - Véronique Carrière
- Cordeliers Research Center, Sorbonne University, Paris Dauphine University 05, INSERM, CNRS, F-75006, Paris, France; Saint-Antoine Research Center, Sorbonne University, INSERM, F-75012, Paris, France.
| |
Collapse
|
240
|
Microbial Adaptation Due to Gastric Bypass Surgery: The Nutritional Impact. Nutrients 2020; 12:nu12041199. [PMID: 32344612 PMCID: PMC7230554 DOI: 10.3390/nu12041199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Bariatric surgery leads to sustained weight loss and the resolution of obesity-related comorbidities. Recent studies have suggested that changes in gut microbiota are associated with the weight loss induced by bariatric surgery. Several studies have observed major changes in the microbial composition following gastric bypass surgery. However, there are inconsistencies between the reported alterations in microbial compositions in different studies. Furthermore, it is well established that diet is an important factor shaping the composition and function of intestinal microbiota. However, most studies on gastric bypass have not assessed the impact of dietary intake on the microbiome composition in general, let alone the impact of restrictive diets prior to bariatric surgery, which are recommended for reducing liver fat content and size. Thus, the relative impact of bariatric surgery on weight loss and gut microbiota remains unclear. Therefore, this review aims to provide a deeper understanding of the current knowledge of the changes in intestinal microbiota induced by bariatric surgery considering pre-surgical nutritional changes.
Collapse
|
241
|
Affiliation(s)
- James N Luo
- Laboratory for Surgical and Metabolic Research, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eric G Sheu
- Laboratory for Surgical and Metabolic Research, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
242
|
Farup PG, Valeur J. Changes in Faecal Short-Chain Fatty Acids after Weight-Loss Interventions in Subjects with Morbid Obesity. Nutrients 2020; 12:nu12030802. [PMID: 32197409 PMCID: PMC7146446 DOI: 10.3390/nu12030802] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/09/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota and their metabolites, e.g., short-chain fatty acids (SCFA), are associated with obesity. The primary aims were to study faecal SCFA levels and the changes in SCFA levels after weight-loss interventions in subjects with obesity, and secondarily, to study factors associated with the faecal SCFA levels. In total, 90 subjects (men / women: 15/75) with a mean age of 44.4 (SD 8.4) years, BMI 41.7 (SD 3.7) kg/m2 and morbid obesity (BMI > 40 or > 35 kg/m2 with obesity-related complications) were included. Faecal SCFA and other variables were measured at inclusion and after a six-month conservative weight-loss intervention followed by bariatric surgery (Roux-en-Y gastric bypass or gastric sleeve). Six months after surgery, the total amount of SCFA was reduced, the total and relative amounts of the main straight SCFA (acetic-, propionic-, and butyric- acids) were reduced, and the total and relative amounts of branched SCFA (isobutyric-, isovaleric-, and isocaproic- acids) were increased. The changes indicate a shift toward a proteolytic fermentation pattern with unfavorable health effects. The amount of SCFA was associated with the diet but not with metabolic markers or makers of the faecal microbiota composition. Dietary interventions could counteract the unfavorable effects.
Collapse
Affiliation(s)
- Per G Farup
- Department of Research, Innlandet Hospital Trust, N-2381 Brumunddal, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, N-7491 Trondheim, Norway
- Correspondence: ; Tel.: +47-948-18-603
| | - Jørgen Valeur
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, N-0440 Oslo, Norway;
| |
Collapse
|
243
|
Ilhan ZE, DiBaise JK, Dautel SE, Isern NG, Kim YM, Hoyt DW, Schepmoes AA, Brewer HM, Weitz KK, Metz TO, Crowell MD, Kang DW, Rittmann BE, Krajmalnik-Brown R. Temporospatial shifts in the human gut microbiome and metabolome after gastric bypass surgery. NPJ Biofilms Microbiomes 2020; 6:12. [PMID: 32170068 PMCID: PMC7070067 DOI: 10.1038/s41522-020-0122-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
Although the etiology of obesity is not well-understood, genetic, environmental, and microbiome elements are recognized as contributors to this rising pandemic. It is well documented that Roux-en-Y gastric bypass (RYGB) surgery drastically alters the fecal microbiome, but data are sparse on temporal and spatial microbiome and metabolome changes, especially in human populations. We characterized the structure and function (through metabolites) of the microbial communities in the gut lumen and structure of microbial communities on mucosal surfaces in nine morbidly obese individuals before, 6 months, and 12 months after RYGB surgery. Moreover, using a comprehensive multi-omic approach, we compared this longitudinal cohort to a previously studied cross-sectional cohort (n = 24). In addition to the expected weight reduction and improvement in obesity-related comorbidities after RYGB surgery, we observed that the impact of surgery was much greater on fecal communities in comparison to mucosal ones. The changes in the fecal microbiome were linked to increased concentrations of branched-chain fatty acids and an overall decrease in secondary bile acid concentrations. The microbiome and metabolome data sets for this longitudinal cohort strengthen our understanding of the persistent impact of RYGB on the gut microbiome and its metabolism. Our findings highlight the importance of changes in mucosal and fecal microbiomes after RYGB surgery. The spatial modifications in the microbiome after RYGB surgery corresponded to persistent changes in fecal fermentation and bile acid metabolism, both of which are associated with improved metabolic outcomes.
Collapse
Affiliation(s)
- Zehra Esra Ilhan
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ, USA.
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA.
| | - John K DiBaise
- Mayo Clinic, Division of Gastroenterology, Scottsdale, AZ, USA
| | - Sydney E Dautel
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Nancy G Isern
- William R. Wiley Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Young-Mo Kim
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - David W Hoyt
- William R. Wiley Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Athena A Schepmoes
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Heather M Brewer
- William R. Wiley Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Karl K Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Thomas O Metz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | | | - Dae-Wook Kang
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA
- Department of Civil & Environmental Engineering, The University of Toledo, Toledo, OH, USA
| | - Bruce E Rittmann
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ, USA
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA
| | - Rosa Krajmalnik-Brown
- Biodesign Swette Center for Environmental Biotechnology, Arizona State University, Tempe, AZ, USA.
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, USA.
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
244
|
Cӑtoi AF, Vodnar DC, Corina A, Nikolic D, Citarrella R, Pérez-Martínez P, Rizzo M. Gut Microbiota, Obesity and Bariatric Surgery: Current Knowledge and Future Perspectives. Curr Pharm Des 2020; 25:2038-2050. [PMID: 31298152 DOI: 10.2174/1381612825666190708190437] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND There is an urgent need for a better understanding and management of obesity and obesity- associated diseases. It is known that obesity is associated with structural and functional changes in the microbiome. METHODS The purpose of this review is to present current evidence from animal and human studies, demonstrating the effects and the potential efficacy of microbiota modulation in improving obesity and associated metabolic dysfunctions. RESULTS This review discusses possible mechanisms linking gut microbiota dysbiosis and obesity, since there is a dual interaction between the two of them. Furthermore, comments on bariatric surgery, as a favourable model to understand the underlying metabolic and inflammatory effects, as well as its association with changes in the composition of the gut microbiota, are included. Also, a possible impact of anti-obesity drugs and the novel antidiabetic drugs on the gut microbiota has been briefly discussed. CONCLUSION More research is needed to better understand here discussed the association between microbiota modulation and obesity. It is expected that research in this field, in the following years, will lead to a personalized therapeutic approach considering the patient's microbiome, and also give rise to the discovery of new drugs and/or the combination therapies for the management of obesity and obesity-related co-morbidities.
Collapse
Affiliation(s)
- Adriana Florinela Cӑtoi
- Pathophysiology Department, "Iuliu Hațieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Dan Cristian Vodnar
- Department of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Andreea Corina
- Lipids and Atherosclerosis Research Unit, 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
| | - Dragana Nikolic
- PROMISE Department, University of Palermo, Italy.,Euro-Mediterranean Institute of Science and Technology (IEMEST), Palermo, Italy
| | | | - Pablo Pérez-Martínez
- Lipids and Atherosclerosis Research Unit, 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
|
245
|
Prifti E, Chevaleyre Y, Hanczar B, Belda E, Danchin A, Clément K, Zucker JD. Interpretable and accurate prediction models for metagenomics data. Gigascience 2020; 9:giaa010. [PMID: 32150601 PMCID: PMC7062144 DOI: 10.1093/gigascience/giaa010] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/12/2019] [Accepted: 01/27/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Microbiome biomarker discovery for patient diagnosis, prognosis, and risk evaluation is attracting broad interest. Selected groups of microbial features provide signatures that characterize host disease states such as cancer or cardio-metabolic diseases. Yet, the current predictive models stemming from machine learning still behave as black boxes and seldom generalize well. Their interpretation is challenging for physicians and biologists, which makes them difficult to trust and use routinely in the physician-patient decision-making process. Novel methods that provide interpretability and biological insight are needed. Here, we introduce "predomics", an original machine learning approach inspired by microbial ecosystem interactions that is tailored for metagenomics data. It discovers accurate predictive signatures and provides unprecedented interpretability. The decision provided by the predictive model is based on a simple, yet powerful score computed by adding, subtracting, or dividing cumulative abundance of microbiome measurements. RESULTS Tested on >100 datasets, we demonstrate that predomics models are simple and highly interpretable. Even with such simplicity, they are at least as accurate as state-of-the-art methods. The family of best models, discovered during the learning process, offers the ability to distil biological information and to decipher the predictability signatures of the studied condition. In a proof-of-concept experiment, we successfully predicted body corpulence and metabolic improvement after bariatric surgery using pre-surgery microbiome data. CONCLUSIONS Predomics is a new algorithm that helps in providing reliable and trustworthy diagnostic decisions in the microbiome field. Predomics is in accord with societal and legal requirements that plead for an explainable artificial intelligence approach in the medical field.
Collapse
Affiliation(s)
- Edi Prifti
- IRD, Sorbonne University, UMMISCO, 32 Avenue Henri Varagnat, F-93143 Bondy, France
- Institute of Cardiometabolism and Nutrition, ICAN, Integromics, 91 Boulevard de l'Hopital, F-75013, Paris, France
| | - Yann Chevaleyre
- Paris-Dauphine University, PSL Research University, CNRS, UMR 7243, LAMSADE, place du Mal. de Lattre de Tassigny, F-75016, Paris, France
| | - Blaise Hanczar
- IBISC, University Paris-Saclay, University Evry, Evry, 23 Boulevard de France, F-91034, France
| | - Eugeni Belda
- Institute of Cardiometabolism and Nutrition, ICAN, Integromics, 91 Boulevard de l'Hopital, F-75013, Paris, France
| | - Antoine Danchin
- Institut Cochin INSERM U1016−CNRS UMR8104−Université Paris Descartes, 24 Rue du Faubourg Saint-Jacques, F-75014, Paris, France
| | - Karine Clément
- Sorbonne University, INSERM, Nutrition and Obesities; Systemic Approach Research Unit (NutriOmics), 91 Boulevard de l'Hopital, F-75013, Paris, France
- Assistance Publique-Hôpitaux de Paris, Nutrition Department, CRNH Ile de France, Pitié-Salpêtrière Hospital, 91 Boulevard de l'Hopital, F-75013, Paris, France
| | - Jean-Daniel Zucker
- IRD, Sorbonne University, UMMISCO, 32 Avenue Henri Varagnat, F-93143 Bondy, France
- Institute of Cardiometabolism and Nutrition, ICAN, Integromics, 91 Boulevard de l'Hopital, F-75013, Paris, France
- Sorbonne University, INSERM, Nutrition and Obesities; Systemic Approach Research Unit (NutriOmics), 91 Boulevard de l'Hopital, F-75013, Paris, France
| |
Collapse
|
246
|
Haange SB, Jehmlich N, Krügel U, Hintschich C, Wehrmann D, Hankir M, Seyfried F, Froment J, Hübschmann T, Müller S, Wissenbach DK, Kang K, Buettner C, Panagiotou G, Noll M, Rolle-Kampczyk U, Fenske W, von Bergen M. Gastric bypass surgery in a rat model alters the community structure and functional composition of the intestinal microbiota independently of weight loss. MICROBIOME 2020; 8:13. [PMID: 32033593 PMCID: PMC7007695 DOI: 10.1186/s40168-020-0788-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/13/2020] [Indexed: 05/02/2023]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) surgery is a last-resort treatment to induce substantial and sustained weight loss in cases of severe obesity. This anatomical rearrangement affects the intestinal microbiota, but so far, little information is available on how it interferes with microbial functionality and microbial-host interactions independently of weight loss. METHODS A rat model was employed where the RYGB-surgery cohort is compared to sham-operated controls which were kept at a matched body weight by food restriction. We investigated the microbial taxonomy and functional activity using 16S rRNA amplicon gene sequencing, metaproteomics, and metabolomics on samples collected from theileum, the cecum, and the colon, and separately analysed the lumen and mucus-associated microbiota. RESULTS Altered gut architecture in RYGB increased the relative occurrence of Actinobacteria, especially Bifidobacteriaceae and Proteobacteria, while in general, Firmicutes were decreased although Streptococcaceae and Clostridium perfringens were observed at relative higher abundances independent of weight loss. A decrease of conjugated and secondary bile acids was observed in the RYGB-gut lumen. The arginine biosynthesis pathway in the microbiota was altered, as indicated by the changes in the abundance of upstream metabolites and enzymes, resulting in lower levels of arginine and higher levels of aspartate in the colon after RYGB. CONCLUSION The anatomical rearrangement in RYGB affects microbiota composition and functionality as well as changes in amino acid and bile acid metabolism independently of weight loss. The shift in the taxonomic structure of the microbiota after RYGB may be mediated by the resulting change in the composition of the bile acid pool in the gut and by changes in the composition of nutrients in the gut. Video abstract.
Collapse
Affiliation(s)
- Sven-Bastiaan Haange
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Nico Jehmlich
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Ute Krügel
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Constantin Hintschich
- Neuroendocrine Regulation of Energy Homeostasis Group, IFB Adiposity Diseases, Leipzig, Germany
| | - Dorothee Wehrmann
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Mohammed Hankir
- Neuroendocrine Regulation of Energy Homeostasis Group, IFB Adiposity Diseases, Leipzig, Germany
- Current address: Department of Experimental Surgery, Wuerzburg University Hospital, Wuerzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Vascular and Pediatric Surgery, Wuerzburg University Hospital, Wuerzburg, Germany
| | - Jean Froment
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Thomas Hübschmann
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Susann Müller
- Department of Environmental Microbiology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Dirk K. Wissenbach
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Current address: Institute of Forensic Medicine, Jena University Hospital, Jena, Germany
| | - Kang Kang
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
| | - Christian Buettner
- Institute for Bioanalysis, Faculty of Applied Sciences, Coburg University of Applied Sciences and Arts, Coburg, Germany
| | - Gianni Panagiotou
- Systems Biology and Bioinformatics Group, School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Matthias Noll
- Institute for Bioanalysis, Faculty of Applied Sciences, Coburg University of Applied Sciences and Arts, Coburg, Germany
| | - Ulrike Rolle-Kampczyk
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Wiebke Fenske
- Neuroendocrine Regulation of Energy Homeostasis Group, IFB Adiposity Diseases, Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| |
Collapse
|
247
|
Nogacka AM, Salazar N, Arboleya S, Ruas-Madiedo P, Mancabelli L, Suarez A, Martinez-Faedo C, Ventura M, Tochio T, Hirano K, Endo A, G. de los Reyes-Gavilán C, Gueimonde M. In Vitro Evaluation of Different Prebiotics on the Modulation of Gut Microbiota Composition and Function in Morbid Obese and Normal-Weight Subjects. Int J Mol Sci 2020; 21:E906. [PMID: 32019174 PMCID: PMC7038051 DOI: 10.3390/ijms21030906] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/13/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota remains relatively stable during adulthood; however, certain intrinsic and environmental factors can lead to microbiota dysbiosis. Its restoration towards a healthy condition using best-suited prebiotics requires previous development of in vitro models for evaluating their functionality. Herein, we carried out fecal cultures with microbiota from healthy normal-weight and morbid obese adults. Cultures were supplemented with different inulin-type fructans (1-kestose, Actilight, P95, Synergy1 and Inulin) and a galactooligosaccharide. Their impact on the gut microbiota was assessed by monitoring gas production and evaluating changes in the microbiota composition (qPCR and 16S rRNA gene profiling) and metabolic activity (gas chromatography). Additionally, the effect on the bifidobacterial species was assessed (ITS-sequencing). Moreover, the functionality of the microbiota before and after prebiotic-modulation was determined in an in vitro model of interaction with an intestinal cell line. In general, 1-kestose was the compound showing the largest effects. The modulation with prebiotics led to significant increases in the Bacteroides group and Faecalibacterium in obese subjects, whereas in normal-weight individuals, substantial rises in Bifidobacterium and Faecalibacterium were appreciated. Notably, the results obtained showed differences in the responses among the tested compounds but also among the studied human populations, indicating the need for developing population-specific products.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
| | - Nuria Salazar
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
| | - Patricia Ruas-Madiedo
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Functionality and Ecology of Beneficial Microorganisms, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Leonardo Mancabelli
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, 43121 Parma, Italy; (L.M.); (M.V.)
| | - Adolfo Suarez
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
- Digestive Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Asturias, Spain
| | - Ceferino Martinez-Faedo
- Endocrinology and Nutrition Service, Central University Hospital of Asturias (HUCA), 33011 Oviedo, Asturias, Spain;
- Endocrinology, Nutrition, Diabetes and Obesity Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Life Sciences, University of Parma, 43121 Parma, Italy; (L.M.); (M.V.)
| | - Takumi Tochio
- β-Food Sciences Co., Chita 478-0046, Japan; (T.T.); (K.H.)
| | | | - Akihito Endo
- Department of Food and Cosmetic Science, Tokyo University of Agriculture, Abashiri 099-2493, Japan;
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (N.S.); (S.A.); (P.R.-M.); (C.G.d.l.R.-G.); (M.G.)
- Diet, Human Microbiota and Health Group, Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Spain;
| |
Collapse
|
248
|
Gut Microbiota Profile of Obese Diabetic Women Submitted to Roux-en-Y Gastric Bypass and Its Association with Food Intake and Postoperative Diabetes Remission. Nutrients 2020; 12:nu12020278. [PMID: 31973130 PMCID: PMC7071117 DOI: 10.3390/nu12020278] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/07/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota composition is influenced by environmental factors and has been shown to impact body metabolism. Objective: To assess the gut microbiota profile before and after Roux-en-Y gastric bypass (RYGB) and the correlation with food intake and postoperative type 2 diabetes remission (T2Dr). Design: Gut microbiota profile from obese diabetic women was evaluated before (n = 25) and 3 (n = 20) and 12 months (n = 14) after RYGB, using MiSeq Illumina-based V4 bacterial 16S rRNA gene profiling. Data on food intake (7-day record) and T2Dr (American Diabetes Association (ADA) criteria) were recorded. Results: Preoperatively, the abundance of five bacteria genera differed between patients with (57%) and without T2Dr (p < 0.050). Preoperative gut bacteria genus signature was able to predict the T2Dr status with 0.94 accuracy ROC curve (receiver operating characteristic curve). Postoperatively (vs. preoperative), the relative abundance of some gut bacteria genera changed, the gut microbial richness increased, and the Firmicutes to Bacteroidetes ratio (rFB) decreased (p < 0.05) regardless of T2Dr. Richness levels was correlated with dietary profile pre and postoperatively, mainly displaying positive and inverse correlations with fiber and lipid intakes, respectively (p < 0.05). Conclusions: Gut microbiota profile was influenced by RYGB and correlated with diet and T2Dr preoperatively, suggesting the possibility to assess its composition to predict postoperative T2Dr.
Collapse
|
249
|
Ciobârcă D, Cătoi AF, Copăescu C, Miere D, Crișan G. Bariatric Surgery in Obesity: Effects on Gut Microbiota and Micronutrient Status. Nutrients 2020; 12:E235. [PMID: 31963247 PMCID: PMC7019602 DOI: 10.3390/nu12010235] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 12/30/2019] [Accepted: 01/13/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is associated with reduced gut microbial diversity and a high rate of micronutrient deficiency. Bariatric surgery, the therapy of choice for severe obesity, produces sustained weight loss and improvements in obesity-related comorbidities. Also, it significantly alters the gut microbiota (GM) composition and function, which might have an important impact on the micronutrient status as GM is able to synthesize certain vitamins, such as riboflavin, folate, B12, or vitamin K2. However, recent data have reported that GM is not fully restored after bariatric surgery; therefore, manipulation of GM through probiotics represents a promising therapeutic approach in bariatric patients. In this review, we discuss the latest evidence concerning the relationship between obesity, GM and micronutrients, the impact of bariatric surgery on GM in relation with micronutrients equilibrium, and the importance of the probiotics' supplementation in obese patients submitted to surgical treatment.
Collapse
Affiliation(s)
- Daniela Ciobârcă
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Adriana Florinela Cătoi
- Department of Physiopathology, Faculty of Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 3-4 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Cătălin Copăescu
- General Surgery Department, Ponderas Hospital, 85A Nicolae G. Caramfil Street, 014142 Bucharest, Romania;
| | - Doina Miere
- Department of Bromatology, Hygiene, Nutrition, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania
| | - Gianina Crișan
- Department of Pharmaceutical Botany, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 23 Gheorghe Marinescu Street, 400337 Cluj-Napoca, Romania;
| |
Collapse
|
250
|
Leocádio PCL, Oriá RB, Crespo-Lopez ME, Alvarez-Leite JI. Obesity: More Than an Inflammatory, an Infectious Disease? Front Immunol 2020; 10:3092. [PMID: 31993062 PMCID: PMC6971046 DOI: 10.3389/fimmu.2019.03092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/17/2019] [Indexed: 12/23/2022] Open
Affiliation(s)
- Paola C L Leocádio
- Laboratório de Aterosclerose e Bioquímica Nutricional, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Reinaldo B Oriá
- Laboratório de Biologia da Cicatrização, Ontogenia e Nutrição de Tecidos, Faculdade de Medicina, Universidade Federal Do Ceará, Fortaleza, Brazil
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Jacqueline I Alvarez-Leite
- Laboratório de Aterosclerose e Bioquímica Nutricional, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|