501
|
Régnier M, Polizzi A, Guillou H, Loiseau N. Sphingolipid metabolism in non-alcoholic fatty liver diseases. Biochimie 2018; 159:9-22. [PMID: 30071259 DOI: 10.1016/j.biochi.2018.07.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) involves a panel of pathologies starting with hepatic steatosis and continuing to irreversible and serious conditions like steatohepatitis (NASH) and hepatocarcinoma. NAFLD is multifactorial in origin and corresponds to abnormal fat deposition in liver. Even if triglycerides are mostly associated with these pathologies, other lipid moieties seem to be involved in the development and severity of NAFLD. That is the case with sphingolipids and more particularly ceramides. In this review, we explore the relationship between NAFLD and sphingolipid metabolism. After providing an analysis of complex sphingolipid metabolism, we focus on the potential involvement of sphingolipids in the different pathologies associated with NAFLD. An unbalanced ratio between ceramides and terminal metabolic products in the liver and plasma promotes weight gain, inflammation, and insulin resistance. In the etiology of NAFLD, some sphingolipid species such as ceramides may be potential biomarkers for NAFLD. We review the clinical relevance of sphingolipids in liver diseases.
Collapse
Affiliation(s)
- Marion Régnier
- INRA UMR1331, ToxAlim, Chemin de Tournefeuille, 31027 Toulouse, France
| | - Arnaud Polizzi
- INRA UMR1331, ToxAlim, Chemin de Tournefeuille, 31027 Toulouse, France
| | - Hervé Guillou
- INRA UMR1331, ToxAlim, Chemin de Tournefeuille, 31027 Toulouse, France
| | - Nicolas Loiseau
- INRA UMR1331, ToxAlim, Chemin de Tournefeuille, 31027 Toulouse, France.
| |
Collapse
|
502
|
Abstract
In recent years, the gut microbiota (the microorganisms that live in our digestive tract) has become an area of great interest. Indeed, this intestinal microbial community performs essential functions in maintaining our health, and has been proven to influence host physiology and metabolism. Thereby, dysregulation of this gut microbiota may be implicated in the development of various diseases, including obesity. However, studies rarely assess causality, which requires the use of germ-free animals and microbiota transplant. Using these strategies, some gut microbiota were shown to confer obesity and associated metabolic disorders to mice, suggesting a causative link between gut bacteria and metabolic diseases.
Collapse
|
503
|
Wang R, Li H, Yang X, Xue X, Deng L, Shen J, Zhang M, Zhao L, Zhang C. Genetically Obese Human Gut Microbiota Induces Liver Steatosis in Germ-Free Mice Fed on Normal Diet. Front Microbiol 2018; 9:1602. [PMID: 30079055 PMCID: PMC6062601 DOI: 10.3389/fmicb.2018.01602] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 06/27/2018] [Indexed: 12/29/2022] Open
Abstract
Dysbiotic gut microbiota contributes to genetically obese phenotype in human. However, the effect of genetic obesity-associated gut microbiota on host hepatic metabolic deteriorations remains largely unknown. Gut microbiota from a genetically obese human donor before and after a dietary weight loss program was transplanted into germ-free C57BL/6J male mice, grouped as PreM and PostM groups, respectively. The gut microbiome, liver pathology and transcriptome response in the gnotobiotic mice were evaluated. After being fed on normal chow diet for 4 weeks, PreM group developed liver macrovesicular steatosis accompanied with higher concentrations of hepatic triglyceride and cholesterol, while PostM group exhibited normal hepatic physiology. The gut microbiota in PreM and PostM groups was significantly different from each other and was more resembling with their respective donor. RNA-sequencing revealed that, in comparison with PostM group, PreM group showed a foregoing pro-steatotic transcriptional response in liver featuring by the repression of lipid beta-oxidation and the activation of lipid absorption and cholesterol uptake before the pathology of liver steatosis. Moreover, peroxisome proliferator-activated receptor alpha (PPARα), which was repressed in PreM group, may act as crucial regulator of the hepatic transcriptional profile of lipid metabolism between two groups. Our results show that gut microbiota from a genetically obese human promotes the onset of liver steatosis by impacting hepatic transcriptional profile of lipid metabolism in mice. This adds new evidence that gut microbiota may play a causative role in the development of non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Ruirui Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xinhe Xue
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liman Deng
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Shen
- Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Menghui Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liping Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Ministry of Education Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers New Jersey Institute for Food, Nutrition, and Health, Rutgers University-New Brunswick, New Brunswick, NJ, United States
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
504
|
Kriss M, Hazleton KZ, Nusbacher NM, Martin CG, Lozupone CA. Low diversity gut microbiota dysbiosis: drivers, functional implications and recovery. Curr Opin Microbiol 2018; 44:34-40. [PMID: 30036705 DOI: 10.1016/j.mib.2018.07.003] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/07/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
Dysbiosis, an imbalance in microbial communities, is linked with disease when this imbalance disturbs microbiota functions essential for maintaining health or introduces processes that promote disease. Dysbiosis in disease is predicted when microbiota differ compositionally from a healthy control population, but only truly defined when these differences are mechanistically related to adverse phenotypes. For the human gut microbiota, dysbiosis varies across diseases. One common manifestation is replacement of the complex community of anaerobes typical of the healthy adult gut microbiome with a community of lower overall microbial diversity and increased facultative anaerobes. Here we review diseases in which low-diversity dysbiosis has been observed and mechanistically linked with disease, with a particular focus on liver disease, inflammatory bowel disease, and Clostridium difficile infection.
Collapse
Affiliation(s)
- Michael Kriss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado, 12700 East 19th Avenue, Campus Box B146, Aurora, CO 80045, USA
| | - Keith Z Hazleton
- Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of Colorado, 13123 East 16th Avenue, Aurora, CO 80045, USA; Digestive Health Institute, Children's Hospital Colorado, 13123 East 16th Avenue, Aurora, CO 80045, USA
| | - Nichole M Nusbacher
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado, 12700 East 19th Avenue, Campus Box 8617, Aurora, CO 80045, USA
| | - Casey G Martin
- Department of Immunology and Microbiology, University of Colorado, 12700 East 19th Avenue,Campus Box 8617, Aurora, CO 80045, USA
| | - Catherine A Lozupone
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado, 12700 East 19th Avenue, Campus Box 8617, Aurora, CO 80045, USA.
| |
Collapse
|
505
|
Comment on "Gut Microbiota as a Driver of Inflammation in Nonalcoholic Fatty Liver Disease". Mediators Inflamm 2018; 2018:3746509. [PMID: 30116145 PMCID: PMC6079468 DOI: 10.1155/2018/3746509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/27/2018] [Indexed: 01/22/2023] Open
|
506
|
Wahlström A. Outside the liver box: The gut microbiota as pivotal modulator of liver diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1865:912-919. [PMID: 31007175 DOI: 10.1016/j.bbadis.2018.07.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023]
Abstract
The gut microbiota affects host physiology and has evolved as an important contributor to health and disease. Gut and liver are closely connected and communicate via the portal vein and the biliary system so the liver is constantly exposed to gut-derived bacterial products and metabolites. The intestinal barrier is important for maintaining physical and functional separation between microbes in the gut and the interior of the host and disruption of the barrier function can lead to bacterial translocation and increased leakage of bacterial metabolites. Liver diseases have been associated with dysbiotic changes in the gut microbiota and impaired gut barrier integrity, thus a future strategy to treat liver disease may be to target the gut microbiota and thereby restore the gut barrier function. This review will summarize and discuss studies that have shown a link between the gut microbiota and liver disease with the main focus on non-alcoholic fatty liver disease and alcoholic liver disease.
Collapse
Affiliation(s)
- Annika Wahlström
- Sahlgrenska Academy, Institute of Medicine, Department of Molecular and Clinical Medicine, Wallenberg Laboratory, University of Gothenburg, S-413 45 Gothenburg, Sweden.
| |
Collapse
|
507
|
Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, Knight R. The gut-liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018; 15:397-411. [PMID: 29748586 PMCID: PMC6319369 DOI: 10.1038/s41575-018-0011-z] [Citation(s) in RCA: 925] [Impact Index Per Article: 132.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, an exciting realization has been that diverse liver diseases - ranging from nonalcoholic steatohepatitis, alcoholic steatohepatitis and cirrhosis to hepatocellular carcinoma - fall along a spectrum. Work on the biology of the gut-liver axis has assisted in understanding the basic biology of both alcoholic fatty liver disease and nonalcoholic fatty liver disease (NAFLD). Of immense importance is the advancement in understanding the role of the microbiome, driven by high-throughput DNA sequencing and improved computational techniques that enable the complexity of the microbiome to be interrogated, together with improved experimental designs. Here, we review gut-liver communications in liver disease, exploring the molecular, genetic and microbiome relationships and discussing prospects for exploiting the microbiome to determine liver disease stage and to predict the effects of pharmaceutical, dietary and other interventions at a population and individual level. Although much work remains to be done in understanding the relationship between the microbiome and liver disease, rapid progress towards clinical applications is being made, especially in study designs that complement human intervention studies with mechanistic work in mice that have been humanized in multiple respects, including the genetic, immunological and microbiome characteristics of individual patients. These 'avatar mice' could be especially useful for guiding new microbiome-based or microbiome-informed therapies.
Collapse
Affiliation(s)
- Anupriya Tripathi
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, USA
| | - Justine Debelius
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - David A Brenner
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Michael Karin
- Department of Pediatrics, University of California, San Diego, CA, USA
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, CA, USA.
- Department of Computer Science and Engineering, University of California, San Diego, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| |
Collapse
|
508
|
Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat Med 2018; 24:1070-1080. [PMID: 29942096 PMCID: PMC6140997 DOI: 10.1038/s41591-018-0061-3] [Citation(s) in RCA: 469] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2018] [Indexed: 02/07/2023]
Abstract
Hepatic steatosis is a multifactorial condition that is often observed in obese patients and is a prelude to non-alcoholic fatty liver disease. Here, we combine shotgun sequencing of fecal metagenomes with molecular phenomics (hepatic transcriptome and plasma and urine metabolomes) in two well-characterized cohorts of morbidly obese women recruited to the FLORINASH study. We reveal molecular networks linking the gut microbiome and the host phenome to hepatic steatosis. Patients with steatosis have low microbial gene richness and increased genetic potential for the processing of dietary lipids and endotoxin biosynthesis (notably from Proteobacteria), hepatic inflammation and dysregulation of aromatic and branched-chain amino acid metabolism. We demonstrated that fecal microbiota transplants and chronic treatment with phenylacetic acid, a microbial product of aromatic amino acid metabolism, successfully trigger steatosis and branched-chain amino acid metabolism. Molecular phenomic signatures were predictive (area under the curve = 87%) and consistent with the gut microbiome having an effect on the steatosis phenome (>75% shared variation) and, therefore, actionable via microbiome-based therapies.
Collapse
|
509
|
Tran M, Liu Y, Huang W, Wang L. Nuclear receptors and liver disease: Summary of the 2017 basic research symposium. Hepatol Commun 2018; 2:765-777. [PMID: 30129636 PMCID: PMC6049066 DOI: 10.1002/hep4.1203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/03/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
The nuclear receptor superfamily contains important transcriptional regulators that play pleiotropic roles in cell differentiation, development, proliferation, and metabolic processes to govern liver physiology and pathology. Many nuclear receptors are ligand-activated transcription factors that regulate the expression of their target genes by modulating transcriptional activities and epigenetic changes. Additionally, the protein complex associated with nuclear receptors consists of a multitude of coregulators, corepressors, and noncoding RNAs. Therefore, acquiring new information on nuclear receptors may provide invaluable insight into novel therapies and shed light on new interventions to reduce the burden and incidence of liver diseases. (Hepatology Communications 2018;2:765-777).
Collapse
Affiliation(s)
- Melanie Tran
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Yanjun Liu
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Li Wang
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT.,Veterans Affairs Connecticut Healthcare System West Haven CT.,Department of Internal Medicine, Section of Digestive Diseases Yale University New Haven CT
| |
Collapse
|
510
|
Wang RR, Zhang L, Xu JJ, Gu Z, Zhang L, Ji G, Liu BC. Human microbiome brings new insights to traditional Chinese medicine. JOURNAL OF BIO-X RESEARCH 2018; 01:41-44. [DOI: 10.1097/jbr.0000000000000007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Rui-Rui Wang
- Shanghai Innovation Center of TCM Health Service
| | - Lei Zhang
- Shanghai Innovation Center of TCM Health Service
| | - Jing-Juan Xu
- Shanghai Innovation Center of TCM Health Service
| | - Zhan Gu
- Shanghai Innovation Center of TCM Health Service
| | - Li Zhang
- Shanghai Innovation Center of TCM Health Service
| | - Guang Ji
- Shanghai Innovation Center of TCM Health Service
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | | |
Collapse
|
511
|
Li BL, Cheng L, Zhou XD, Peng X. [Research progress on the relationship between oral microbes and digestive system diseases]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2018; 36:331-335. [PMID: 29984938 DOI: 10.7518/hxkq.2018.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The human microbiome project promoted further understanding on human oral microbes. Besides oral diseases such as dental caries, periodontal disease, and oral cancer, oral microbes are closely associated with systematic diseases. They have a close connection with digestive system diseases and even contribute to the origination and progression of colorectal cancer. By reviewing recent studies involving oral microbe-related digestive systemic diseases, we aim to propose the considerable role of oral microbes in relation to digestive systemic diseases and the way of oral microbes to multiple organs of digestive system.
Collapse
Affiliation(s)
- Bo-Lei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xue-Dong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xian Peng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
512
|
Anania C, Perla FM, Olivero F, Pacifico L, Chiesa C. Mediterranean diet and nonalcoholic fatty liver disease. World J Gastroenterol 2018; 24:2083-2094. [PMID: 29785077 PMCID: PMC5960814 DOI: 10.3748/wjg.v24.i19.2083] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/27/2018] [Accepted: 05/05/2018] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is emerging as the most common chronic liver disease, and is characterized by a wide spectrum of fat-liver disorders that can result in severe liver disease and cirrhosis. Inflammation and oxidative stress are the major risk factors involved in the pathogenesis of NAFLD. Currently, there is no consensus concerning the pharmacological treatment of NAFLD. However, lifestyle interventions based on exercise and a balanced diet for quality and quantity, are considered the cornerstone of NAFLD management. Mediterranean diet (MD), rich in polyunsaturated fats, polyphenols, vitamins and carotenoids, with their anti-inflammatory and anti-oxidant effects, has been suggested to be effective in preventing cardiovascular risk factors. In adults, MD has also been demonstrated to be efficacious in reducing the risk of metabolic syndrome. However, few studies are available on the effects of the MD in both adult and pediatric subjects with NAFLD. Thus, the aims of the present narrative review are to analyze the current clinical evidence on the impact of MD in patients with NAFLD, and to summarize the main mechanisms of action of MD components on this condition.
Collapse
Affiliation(s)
- Caterina Anania
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome 00161, Italy
| | | | - Francesca Olivero
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome 00161, Italy
| | - Lucia Pacifico
- Policlinico Umberto I Hospital, Sapienza University of Rome, Rome 00161, Italy
| | - Claudio Chiesa
- Institute of Translational Pharmacology, National Research Council, Rome 00133, Italy
| |
Collapse
|
513
|
Histological improvement of non-alcoholic steatohepatitis with a prebiotic: a pilot clinical trial. Eur J Nutr 2018; 58:1735-1745. [PMID: 29779170 DOI: 10.1007/s00394-018-1721-2] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/15/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE In obesity and diabetes the liver is highly susceptible to abnormal uptake and storage of fat. In certain individuals hepatic steatosis predisposes to the development of non-alcoholic steatohepatitis (NASH), a disease marked by hepatic inflammation and fibrosis. Although the precise pathophysiology of NASH is unknown, it is believed that the gut microbiota-liver axis influences the development of this disease. With few treatment strategies available for NASH, exploration of gut microbiota-targeted interventions is warranted. We investigated the therapeutic potential of a prebiotic supplement to improve histological parameters of NASH. METHODS In a placebo-controlled, randomized pilot trial, 14 individuals with liver-biopsy-confirmed NASH [non-alcoholic fatty liver activity score (NAS) ≥ 5] were randomized to receive oligofructose (8 g/day for 12 weeks followed by 16 g/day for 24 weeks) or isocaloric placebo for 9 months. The primary outcome measure was the change in liver biopsy NAS score and the secondary outcomes included changes in body weight, body composition, glucose tolerance, inflammatory markers, and gut microbiota. RESULTS Independent of weight loss, oligofructose improved liver steatosis relative to placebo and improved overall NAS score (P = 0.016). Bifidobacterium was enhanced by oligofructose, whereas bacteria within Clostridium cluster XI and I were reduced with oligofructose (P < 0.05). There were no adverse side effects that deterred individuals from consuming oligofructose for treatment of this disease. CONCLUSIONS Independent of other lifestyle changes, prebiotic supplementation reduced histologically-confirmed steatosis in patients with NASH. Larger follow-up studies are warranted. CLINICAL TRIAL This trial was registered at Clinicaltrials.com as NCT03184376.
Collapse
|
514
|
Liu H, Zhang H, Wang X, Yu X, Hu C, Zhang X. The family Coriobacteriaceae is a potential contributor to the beneficial effects of Roux-en-Y gastric bypass on type 2 diabetes. Surg Obes Relat Dis 2018; 14:584-593. [DOI: 10.1016/j.soard.2018.01.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/04/2017] [Accepted: 01/08/2018] [Indexed: 01/22/2023]
|
515
|
van de Guchte M, Blottière HM, Doré J. Humans as holobionts: implications for prevention and therapy. MICROBIOME 2018; 6:81. [PMID: 29716650 PMCID: PMC5928587 DOI: 10.1186/s40168-018-0466-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/23/2018] [Indexed: 05/27/2023]
Abstract
The human gut microbiota is increasingly recognized for its important or even decisive role in health. As it becomes clear that microbiota and host mutually affect and depend on each other in an intimate relationship, a holistic view of the gut microbiota-host association imposes itself. Ideally, a stable state of equilibrium, homeostasis, is maintained and serves health, but signs are that perturbation of this equilibrium beyond the limits of resilience can propel the system into an alternative stable state, a pre-disease state, more susceptible to the development of chronic diseases. The microbiota-host equilibrium of a large and growing proportion of individuals in Western society may represent such a pre-disease state and explain the explosive development of chronic diseases such as inflammatory bowel disease, obesity, and other inflammatory diseases. These diseases themselves represent other alternative stable states again and are therefore hard to cure. The holistic view of the microbiota-host association where feedback loops between microbiota and host are thought to maintain the system in a stable state-be it a healthy, pre-disease, or disease state-implies that integrated approaches, addressing host processes and microbiota, should be used to treat or prevent (pre-)disease.
Collapse
Affiliation(s)
- Maarten van de Guchte
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| | - Hervé M Blottière
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- MetaGenoPolis, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Joël Doré
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
- MetaGenoPolis, INRA, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| |
Collapse
|
516
|
Wrzosek L, Ciocan D, Borentain P, Spatz M, Puchois V, Hugot C, Ferrere G, Mayeur C, Perlemuter G, Cassard AM. Transplantation of human microbiota into conventional mice durably reshapes the gut microbiota. Sci Rep 2018; 8:6854. [PMID: 29717179 PMCID: PMC5931539 DOI: 10.1038/s41598-018-25300-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Human microbiota-associated (HMA) mice are an important model to study the relationship between liver diseases and intestinal microbiota. We describe a new method to humanize conventional mice based on bowel cleansing with polyethylene glycol followed by fecal microbiota transplantation (FMT) from a human donor. Four successive bowel cleansings were sufficient to empty the intestine and decrease the microbiota by 90%. We then compared four different strategies based on the frequency of FMT over four weeks: (1) twice a week; (2) once a week; (3) two FMTs; (4) one FMT. We were able to transfer human bacteria to mice, irrespective of the strategy used. We detected human bacteria after four weeks, even if only one FMT was performed, but there was a shift of the microbiota over time. FMT twice a week for four weeks was too frequent and perturbed the stability of the newly formed ecosystem. FMT once a week appears to be the best compromise as it allowed engraftment of Faecalibacterium, and a higher diversity of bacteria belonging to the Bacteroidales order. Our easy to establish HMA mouse model could be used as an alternative to classical HMA mice to study the relationship between the liver and the microbiota.
Collapse
Affiliation(s)
- Laura Wrzosek
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Dragos Ciocan
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Patrick Borentain
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
- Service d'Hépato-Gastroentérologie, Hôpital de la Timone, Marseille, France
| | - Madeleine Spatz
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Virginie Puchois
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Cindy Hugot
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Gladys Ferrere
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
| | - Camille Mayeur
- INRA, UMR 1319 MICALIS, AgroParisTech, Jouy-en-Josas, France
| | - Gabriel Perlemuter
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France
- AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - Anne-Marie Cassard
- INSERM U996, Inflammation Chemokines and Immunopathology, Faculté de Médecine-Univ Paris-Sud, Université Paris-Saclay, Clamart, France.
| |
Collapse
|
517
|
Helicobacter pylori and Risk of Nonalcoholic Fatty Liver Disease: A Systematic Review and Meta-analysis. J Clin Gastroenterol 2018; 52:386-391. [PMID: 28098578 DOI: 10.1097/mcg.0000000000000784] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES Helicobacter pylori (H. pylori) is the most common chronic bacterial infection. Patients with H. pylori infection may be at an increased risk of nonalcoholic fatty liver disease (NAFLD) because of chronic inflammation and insulin resistance. Several epidemiologic studies attempting to determine this risk have yielded inconsistent results. This meta-analysis was conducted with the aims to summarize all available evidence and estimate the risk of NAFLD in patients with H. pylori infection. METHODS A literature search was performed using MEDLINE and EMBASE database from inception to June 2016. Studies that reported relative risks, odd ratios, or hazard ratios comparing the risk of NAFLD among patients with H. pylori infection versus without H. pylori infection were included. Pooled odds ratios and 95% confidence intervals were calculated using a random-effect, generic inverse variance method. RESULTS Six studies met our eligibility criteria and were included in this analysis. We found a statistically significant increased risk of NAFLD among patients with H. pylori infection with the pooled odds ratios of 1.21 (95% confidence interval, 1.07-1.37). The statistical heterogeneity was low with an I of 49%. CONCLUSIONS A significantly increased risk of NAFLD among patients with H. pylori infection was demonstrated in this meta-analysis. Further studies are required to clarify how this risk should be addressed in clinical practice.
Collapse
|
518
|
Even M, Davail S, Rey M, Tavernier A, Houssier M, Bernadet MD, Gontier K, Pascal G, Ricaud K. Probiotics Strains Modulate Gut Microbiota and Lipid Metabolism in Mule Ducks. Open Microbiol J 2018; 12:71-93. [PMID: 29755604 PMCID: PMC5925865 DOI: 10.2174/1874285801812010071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 01/22/2023] Open
Abstract
Background: Livestock production should respond to societal, environmental and economic changes. Since 2006 and the ban on antibiotics as growth factors in European Union, the use of probiotics has become widespread and has demonstrated the effect of intestinal microbiota on the performance of farm animals. Objective: The aim of this study was to investigate the effect of supplementation with Lactobacillus salivarius (as a probiotics strain or combined with other strains) on zootechnical performance, metabolic and immune gene expression and intestinal microbiota diversity in mule ducks using high-throughput sequencing and real-time PCR. Method: The mule ducks were reared for 79 days and overfed for 12 days with or without probiotics. Samples were collected at 14 (starting period) and 91 days (end of overfeeding period), 3 hours post feeding. Results: Irrespective of digestive content, age, level of feed intake or supplementation with probiotics, Firmicutes, Proteobacteria and Bacteroidetes were the dominant phyla in the bacterial community in mule ducks. At 14 days, both the ileal and cecal samples were dominated by Firmicutes (in particular the Clostridiales order). Overfeeding induced a shift between Clostridiales and Lactobacillales in the ileal samples whereas in the cecal samples, the relative abundance of Firmicutes decreased. Overfeeding also induced hepatic over-expression of Fatty Acid Synthase (FAS) and of the lipid transporter gene Fatty Acid Binding Protein 4 (FABP4). This increase in lipid metabolism genes is associated with a decrease in inflammatory response. Conclusion: Finally, probiotic supplementation had only a slight impact on gene expression and microbiota diversity, both at 14 days and after overfeeding.
Collapse
Affiliation(s)
- Maxime Even
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| | - Stéphane Davail
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| | - Mikael Rey
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France
| | - Annabelle Tavernier
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| | - Marianne Houssier
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| | - Marie Dominique Bernadet
- UEPFG INRA Bordeaux-Aquitaine, (Unité Expérimentale Palmipèdes à Foie Gras), Domaine d'Artiguères 1076, route de Haut Mauco, F-40280 Benquet, France
| | - Karine Gontier
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| | - Géraldine Pascal
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet-Tolosan, France
| | - Karine Ricaud
- UMR 1419 INRA UPPA NuMéA, 371 rue du ruisseau, 40000 Mont de Marsan, France.,UMR 1419 INRA UPPA NuMéA, Quartier Ibarron, 64310 Saint Pée sur Nivelle, France
| |
Collapse
|
519
|
Proton pump inhibitors as risk factor for metabolic syndrome and hepatic steatosis in coeliac disease patients on gluten-free diet. J Gastroenterol 2018; 53:507-516. [PMID: 28823009 DOI: 10.1007/s00535-017-1381-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 08/08/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recent research has shown that patients with coeliac disease (CD) are at risk of developing metabolic syndrome (MS) and hepatic steatosis (HS) after commencing a gluten-free diet (GFD). This study aimed to evaluate the predictive factors for MS and HS in CD after 1 year of GFD. METHODS All consecutive newly diagnosed CD patients were enrolled. We prospectively collected data about BMI; waist circumference; blood pressure; cholesterol; triglycerides, glucose and insulin blood levels; insulin resistance (through the homeostatic model assessment HOMA-IR) and treatment with proton pump inhibitors (PPI). Diagnosis of MS was made in accordance with current guidelines and HS was diagnosed by ultrasonography. The prevalence of MS and HS was re-assessed after 1 year of GFD. A logistic regression analysis was performed to identify risk factors for MS and HS occurrence after 1 year of GFD. RESULTS Of 301 patients with newly diagnosed CD, 4.3% met criteria for diagnosis of MS and 25.9% presented with HS at the time of CD diagnosis; 99 subjects (32.8%) had long-term exposure to PPI during the study period. After 1 year, 72 (23.9%) patients had developed MS (4.3 vs 23.9%; p < 0.001, OR 6.9) and 112 (37.2%) had developed HS (25.9 vs 37.2%; p < 0.01, OR 1.69). At multivariate analysis, high BMI at diagnosis (OR 10.8; p < 0.001) and PPI exposure (OR 22.9; p < 0.001) were the only factors associated with the occurrence of MS; HOMA-IR (OR 9.7; p < 0.001) and PPI exposure (OR 9.2; p < 0.001) were the only factors associated with the occurrence of HS. CONCLUSIONS PPI exposure adds further risk of occurrence of MS and HS for patients with CD on GFD. The use of PPI in patients with CD on GFD should be limited to strict indications.
Collapse
|
520
|
de Faria Ghetti F, Oliveira DG, de Oliveira JM, de Castro Ferreira LEVV, Cesar DE, Moreira APB. Influence of gut microbiota on the development and progression of nonalcoholic steatohepatitis. Eur J Nutr 2018; 57:861-876. [PMID: 28875318 DOI: 10.1007/s00394-017-1524-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 08/06/2017] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nonalcoholic steatohepatitis (NASH) is characterized by the presence of steatosis, inflammation, and ballooning degeneration of hepatocytes, with or without fibrosis. The prevalence of NASH has increased with the obesity epidemic, but its etiology is multifactorial. The current studies suggest the role of gut microbiota in the development and progression of NASH. The aim is to review the studies that investigate the relationship between gut microbiota and NASH. These review also discusses the pathophysiological mechanisms and the influence of diet on the gut-liver axis. RESULT The available literature has proposed mechanisms for an association between gut microbiota and NASH, such as: modification energy homeostasis, lipopolysaccharides (LPS)-endotoxemia, increased endogenous production of ethanol, and alteration in the metabolism of bile acid and choline. There is evidence to suggest that NASH patients have a higher prevalence of bacterial overgrowth in the small intestine and changes in the composition of the gut microbiota. However, there is still a controversy regarding the microbiome profile in this population. The abundance of Bacteroidetes phylum may be increased, decreased, or unaltered in NASH patients. There is an increase in the Escherichia and Bacteroides genus. There is depletion of certain taxa, such as Prevotella and Faecalibacterium. CONCLUSION Although few studies have evaluated the composition of the gut microbiota in patients with NASH, it is observed that these individuals have a distinct gut microbiota, compared to the control groups, which explains, at least in part, the genesis and progression of the disease through multiple mechanisms. Modulation of the gut microbiota through diet control offers new challenges for future studies.
Collapse
Affiliation(s)
- Fabiana de Faria Ghetti
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil.
- Unidade de Nutrição Clínica, Hospital Universitário, Rua Catulo Breviglieri, s/n, Bairro Santa Catarina, Juiz de Fora, Minas Gerais, CEP 36036-330, Brazil.
| | - Daiane Gonçalves Oliveira
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Juliano Machado de Oliveira
- Universitary Hospital and School of Medicine, Federal University of Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | | | | | | |
Collapse
|
521
|
Van Hul M, Geurts L, Plovier H, Druart C, Everard A, Ståhlman M, Rhimi M, Chira K, Teissedre PL, Delzenne NM, Maguin E, Guilbot A, Brochot A, Gérard P, Bäckhed F, Cani PD. Reduced obesity, diabetes, and steatosis upon cinnamon and grape pomace are associated with changes in gut microbiota and markers of gut barrier. Am J Physiol Endocrinol Metab 2018; 314:E334-E352. [PMID: 28874357 DOI: 10.1152/ajpendo.00107.2017] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Increasing evidence suggests that polyphenols have a significant potential in the prevention and treatment of risk factors associated with metabolic syndrome. The objective of this study was to assess the metabolic outcomes of two polyphenol-containing extracts from cinnamon bark (CBE) and grape pomace (GPE) on C57BL/6J mice fed a high-fat diet (HFD) for 8 wk. Both CBE and GPE were able to decrease fat mass gain and adipose tissue inflammation in mice fed a HFD without reducing food intake. This was associated with reduced liver steatosis and lower plasma nonesterified fatty acid levels. We also observed a beneficial effect on glucose homeostasis, as evidenced by an improved glucose tolerance and a lower insulin resistance index. These ameliorations of the overall metabolic profile were associated with a significant impact on the microbial composition, which was more profound for the GPE than for the CBE. At the genus level, Peptococcus were decreased in the CBE group. In the GPE-treated group, several key genera that have been previously found to be linked with HFD, metabolic effects, and gut barrier integrity were affected: we observed a decrease of Desulfovibrio, Lactococcus, whereas Allobaculum and Roseburia were increased. In addition, the expression of several antimicrobial peptides and tight junction proteins was increased in response to both CBE and GPE supplementation, indicating an improvement of the gut barrier function. Collectively, these data suggest that CBE and GPE can ameliorate the overall metabolic profile of mice on a high-fat diet, partly by acting on the gut microbiota.
Collapse
Affiliation(s)
- Matthias Van Hul
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Lucie Geurts
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Hubert Plovier
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Céline Druart
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Amandine Everard
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Marcus Ståhlman
- Wallenberg Laboratory, University of Gothenburg , Gothenburg , Sweden
| | - Moez Rhimi
- Micalis, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay , Jouy-en-Josas , France
| | - Kleopatra Chira
- Université Bordeaux, Institut des Sciences de la Vigne et du Vin (ISVV), EA 4577 Œnologie, Villenave d'Ornon, France
- INRA, ISVV, USC 1366 Œnologie, Villenave d'Ornon, France
| | - Pierre-Louis Teissedre
- Université Bordeaux, Institut des Sciences de la Vigne et du Vin (ISVV), EA 4577 Œnologie, Villenave d'Ornon, France
- INRA, ISVV, USC 1366 Œnologie, Villenave d'Ornon, France
| | - Nathalie M Delzenne
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| | - Emmanuelle Maguin
- Micalis, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay , Jouy-en-Josas , France
| | | | | | - Philippe Gérard
- Micalis, Institut National de la Recherche Agronomique (INRA), AgroParisTech, Université Paris-Saclay , Jouy-en-Josas , France
| | - Fredrik Bäckhed
- Wallenberg Laboratory, 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
| | - Patrice D Cani
- Université Catholique de Louvain, Louvain Drug Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group , Brussels , Belgium
| |
Collapse
|
522
|
García-Lezana T, Raurell I, Bravo M, Torres-Arauz M, Salcedo MT, Santiago A, Schoenenberger A, Manichanh C, Genescà J, Martell M, Augustin S. Restoration of a healthy intestinal microbiota normalizes portal hypertension in a rat model of nonalcoholic steatohepatitis. Hepatology 2018; 67:1485-1498. [PMID: 29113028 DOI: 10.1002/hep.29646] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/16/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022]
Abstract
UNLABELLED Portal hypertension (PH) drives most of the clinical complications in chronic liver diseases. However, its progression in nonalcoholic steatohepatitis (NASH) and its association with the intestinal microbiota (IM) have been scarcely studied. Our aim was to investigate the role of the IM in the mechanisms leading to PH in early NASH. The experimental design was divided in two stages. In stage 1, Sprague-Dawley rats were fed for 8 weeks a high-fat, high-glucose/fructose diet (HFGFD) or a control diet/water (CD). Representative rats were selected as IM donors for stage 2. In stage 2, additional HFGFD and CD rats underwent intestinal decontamination, followed by IM transplantation with feces from opposite-diet donors (heterologous transplant) or autologous fecal transplant (as controls), generating four groups: CD-autotransplanted, CD-transplanted, HFGFD-autotransplanted, HFGFD-transplanted. After IM transplantation, the original diet was maintained for 12-14 days until death. HFGFD rats developed obesity, insulin resistance, NASH without fibrosis but with PH, intrahepatic endothelial dysfunction, and IM dysbiosis. In HFGFD rats, transplantation with feces from CD donors caused a significant reduction of PH to levels comparable to CD without significant changes in NASH histology. The reduction in PH was due to a 31% decrease of intrahepatic vascular resistance compared to the HFGFD-autotransplanted group (P < 0.05). This effect occurs through restoration of the sensitivity to insulin of the hepatic protein kinase B-dependent endothelial nitric oxide synthase signaling pathway. CONCLUSION The IM exerts a direct influence in the development of PH in rats with diet-induced NASH and dysbiosis; PH, insulin resistance, and endothelial dysfunction revert when a healthy IM is restored. (Hepatology 2018;67:1485-1498).
Collapse
Affiliation(s)
- Teresa García-Lezana
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Imma Raurell
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Miren Bravo
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Manuel Torres-Arauz
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Alba Santiago
- Digestive System Research Unit, Institut de Recerca Vall d'Hebron, Barcelona, Spain
| | | | - Chaysavanh Manichanh
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain.,Digestive System Research Unit, Institut de Recerca Vall d'Hebron, Barcelona, Spain
| | - Joan Genescà
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - María Martell
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Salvador Augustin
- Liver Unit, Department of Internal Medicine, Hospital Universitari Vall d'Hebron, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
523
|
Bluemel S, Wang L, Martino C, Lee S, Wang Y, Williams B, Horvath A, Stadlbauer V, Zengler K, Schnabl B. The Role of Intestinal C-type Regenerating Islet Derived-3 Lectins for Nonalcoholic Steatohepatitis. Hepatol Commun 2018; 2:393-406. [PMID: 29619418 PMCID: PMC5880191 DOI: 10.1002/hep4.1165] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/15/2018] [Accepted: 02/08/2018] [Indexed: 12/14/2022] Open
Abstract
C-type regenerating islet derived-3 (Reg3) lectins defend against pathogens and keep commensal bacteria at a distance. Deficiency of Reg3g and Reg3b facilitates alcohol-induced bacterial translocation and alcoholic liver disease. Intestinal Reg3g is down-regulated in animal models of diet-induced obesity, but the functional consequences for nonalcoholic steatohepatitis (NASH) are unknown. The aim of this study was to investigate the role of Reg3 lectins in NASH. NASH was induced by a Western-style fast-food diet in mice deficient for Reg3g or Reg3b and in transgenic mice overexpressing Reg3g in intestinal epithelial cells (Reg3gTg). Glucose tolerance was assessed after 18 weeks and insulin resistance after 19 weeks of feeding. After 20 weeks, mice were assessed for features of the metabolic syndrome. Obesity was not different in genetically modified mice compared with their respective wild-type littermates. Glucose intolerance, liver injury, hepatic inflammation, steatosis, fibrosis, and bacterial translocation to mesenteric lymph nodes and to the liver were not different in Reg3g-deficient mice compared with wild-type littermates. Plasma endotoxin levels were higher in Reg3g-deficient mice. Reg3b deficiency protected against glucose intolerance, but liver disease, bacterial translocation, and plasma endotoxin levels were similar to wild-type littermates. Absence of either REG3G or REG3B protein in the ileum was not compensated for by up-regulation of the respective other REG3 protein. Transgenic Reg3g mice also developed liver injury, steatosis, and fibrosis similar to their wild-type littermates. Conclusion: In contrast to alcoholic liver disease, loss of intestinal Reg3 lectins is not sufficient to aggravate diet-induced obesity and NASH. This supports a multi-hit pathogenesis in NASH. Only glucose metabolism is affected by Reg3b deficiency. (Hepatology Communications 2018;2:393-406).
Collapse
Affiliation(s)
- Sena Bluemel
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - Lirui Wang
- Department of MedicineUniversity of California San DiegoLa JollaCA
- Department of MedicineVA San Diego Healthcare SystemSan DiegoCA
| | - Cameron Martino
- Department of PediatricsDivision of Host‐Microbe Systems and TherapeuticsSan DiegoCA
| | - Suhan Lee
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - Yanhan Wang
- Department of MedicineUniversity of California San DiegoLa JollaCA
- Department of MedicineVA San Diego Healthcare SystemSan DiegoCA
| | - Brandon Williams
- Department of MedicineUniversity of California San DiegoLa JollaCA
| | - Angela Horvath
- Department of Internal Medicine, Division of Gastroenterology and HepatologyMedical University of GrazGrazAustria
- Center of Biomarker Research in MedicineGrazAustria
| | - Vanessa Stadlbauer
- Department of Internal Medicine, Division of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - Karsten Zengler
- Department of PediatricsDivision of Host‐Microbe Systems and TherapeuticsSan DiegoCA
- Center for Microbiome InnovationUniversity of California San DiegoLa JollaCA
| | - Bernd Schnabl
- Department of MedicineUniversity of California San DiegoLa JollaCA
- Department of MedicineVA San Diego Healthcare SystemSan DiegoCA
| |
Collapse
|
524
|
Lyu Y, Wu L, Wang F, Shen X, Lin D. Carotenoid supplementation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis. Exp Biol Med (Maywood) 2018. [PMID: 29534601 DOI: 10.1177/1535370218763760] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis, a broad spectrum of imbalance of the gut microbiota, may progress to microbiota dysfunction. Dysbiosis is linked to some human diseases, such as inflammation-related disorders and metabolic syndromes. However, the underlying mechanisms of the pathogenesis of dysbiosis remain elusive. Recent findings suggest that the microbiome and gut immune responses, like immunoglobulin A production, play critical roles in the gut homeostasis and function, and the progression of dysbiosis. In the past two decades, much progress has been made in better understanding of production of immunoglobulin A and its association with commensal microbiota. The present minireview summarizes the recent findings in the gut microbiota dysbiosis and dysfunction of immunoglobulin A induced by the imbalance of pathogenic bacteria and commensal microbiota. We also propose the potentials of dietary carotenoids, such as β-carotene and astaxanthin, in the improvement of the gut immune system maturation and immunoglobulin A production, and the consequent promotion of the gut health. Impact statement The concept of carotenoid metabolism in the gut health has not been well established in the literature. Here, we review and discuss the roles of retinoic acid and carotenoids, including pro-vitamin A carotenoids and xanthophylls in the maturation of the gut immune system and IgA production. This is the first review article about the carotenoid supplements and the metabolites in the regulation of the gut microbiome. We hope this review would provide a new direction for the management of the gut microbiota dysbiosis by application of bioactive carotenoids and the metabolites.
Collapse
Affiliation(s)
- Yi Lyu
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Lei Wu
- 2 Department of Nutritional Sciences, 7618 Oklahoma State University, Stillwater , OK 74078, USA
| | - Fang Wang
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Xinchun Shen
- 1 College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, 12391 Nanjing University of Finance and Economics , Nanjing 210023, China
| | - Dingbo Lin
- 2 Department of Nutritional Sciences, 7618 Oklahoma State University, Stillwater , OK 74078, USA
| |
Collapse
|
525
|
Mardinoglu A, Wu H, Bjornson E, Zhang C, Hakkarainen A, Räsänen SM, Lee S, Mancina RM, Bergentall M, Pietiläinen KH, Söderlund S, Matikainen N, Ståhlman M, Bergh PO, Adiels M, Piening BD, Granér M, Lundbom N, Williams KJ, Romeo S, Nielsen J, Snyder M, Uhlén M, Bergström G, Perkins R, Marschall HU, Bäckhed F, Taskinen MR, Borén J. An Integrated Understanding of the Rapid Metabolic Benefits of a Carbohydrate-Restricted Diet on Hepatic Steatosis in Humans. Cell Metab 2018; 27:559-571.e5. [PMID: 29456073 PMCID: PMC6706084 DOI: 10.1016/j.cmet.2018.01.005] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/06/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
A carbohydrate-restricted diet is a widely recommended intervention for non-alcoholic fatty liver disease (NAFLD), but a systematic perspective on the multiple benefits of this diet is lacking. Here, we performed a short-term intervention with an isocaloric low-carbohydrate diet with increased protein content in obese subjects with NAFLD and characterized the resulting alterations in metabolism and the gut microbiota using a multi-omics approach. We observed rapid and dramatic reductions of liver fat and other cardiometabolic risk factors paralleled by (1) marked decreases in hepatic de novo lipogenesis; (2) large increases in serum β-hydroxybutyrate concentrations, reflecting increased mitochondrial β-oxidation; and (3) rapid increases in folate-producing Streptococcus and serum folate concentrations. Liver transcriptomic analysis on biopsy samples from a second cohort revealed downregulation of the fatty acid synthesis pathway and upregulation of folate-mediated one-carbon metabolism and fatty acid oxidation pathways. Our results highlight the potential of exploring diet-microbiota interactions for treating NAFLD.
Collapse
Affiliation(s)
- Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Hao Wu
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Elias Bjornson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Antti Hakkarainen
- HUS Medical Imaging Center, Radiology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Sari M Räsänen
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Sunjae Lee
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mattias Bergentall
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kirsi H Pietiläinen
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Sanni Söderlund
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Niina Matikainen
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland; Endocrinology, Abdominal Center, Helsinki University Hospital, Helsinki, Finland
| | - Marcus Ståhlman
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per-Olof Bergh
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Martin Adiels
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Brian D Piening
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Marit Granér
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Nina Lundbom
- HUS Medical Imaging Center, Radiology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Kevin J Williams
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Michael Snyder
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Mathias Uhlén
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rosie Perkins
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Fredrik Bäckhed
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Marja-Riitta Taskinen
- Research Programs Unit, Diabetes and Obesity, University of Helsinki and Department of Internal Medicine, Helsinki University Hospital, Helsinki, Finland
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg, and Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
526
|
Zhou XL, Yan BB, Xiao Y, Zhou YM, Liu TY. Tartary buckwheat protein prevented dyslipidemia in high-fat diet-fed mice associated with gut microbiota changes. Food Chem Toxicol 2018; 119:296-301. [PMID: 29481895 DOI: 10.1016/j.fct.2018.02.052] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 01/09/2023]
Abstract
As one of low-digestible proteins, tartary buckwheat protein (BWP) revealed a cholesterol-lowering activity. The relationship between the prevention of BWP on dyslipidemia and changes in the numbers of gut microbiota was investigated. The male C57BL/6 mice were separately fed on normal diet, high-fat diet (HFD) with casein, and HFD with BWP extract for 6 weeks. Quantitative PCR assay was applied to quantify the microbiota composition in feces. The levels of plasma total cholesterol (TC) and triglyceride (TG) in the mice fed on HFD with BWP were significantly lower than those on HFD with casein. BWP promoted the growth of Lactobacillus, Bifidobacterium and Enterococcus and inhibited the growth of Escherichia coli in HFD-fed mice. Moreover, Bifidobacterium population was closely related to contents of plasma lipids. Further, BWP significantly decreased the levels of plasma inflammation factors as induced by HFD, including lipopolysaccharide, tumor necrosis factor α and interleukin 6. BWP significantly increased the excretion of total bile acids and short-chain fatty acids in feces. In conlusion, BWP benefited cholesterol metabolism, which could be attributed to regulating composition of gut microbiota.
Collapse
Affiliation(s)
- Xiao-Li Zhou
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai 200436, China; School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Bei-Bei Yan
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Ying Xiao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Yi-Ming Zhou
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai 200436, China; School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Tai-Yi Liu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
527
|
Schuppan D, Surabattula R, Wang XY. Determinants of fibrosis progression and regression in NASH. J Hepatol 2018; 68:238-250. [PMID: 29154966 DOI: 10.1016/j.jhep.2017.11.012] [Citation(s) in RCA: 360] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/02/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023]
Abstract
Cirrhosis has become the major liver-related clinical endpoint in non-alcoholic steatohepatitis (NASH). However, progression to cirrhosis is less predictable in NASH than in other chronic liver diseases. This is due to the complex and multifactorial aetiology of NASH, which is determined by lifestyle and nutrition, multiple genetic and epigenetic factors, and a prominent role of hepatic and extrahepatic comorbidities. Thus, modest changes in these cofactors can also induce fibrosis regression, at least in patients with precirrhotic liver disease. Fibrogenesis in NASH correlates with, but is indirectly coupled to, classical inflammation, since fibrosis progression is driven by repetitive periods of repair. While hepatocyte lipoapoptosis is a key driving force of fibrosis progression, activated hepatic stellate cells, myofibroblasts, cholangiocytes, macrophages and components of the pathological extracellular matrix are major fibrogenic effectors and thus pharmacological targets for therapies aimed at inhibition of fibrosis progression or induction of fibrosis reversal. The advent of novel, highly sensitive and specific serum biomarkers and imaging methods to assess the dynamics of liver fibrosis in NASH will improve detection, stratification and follow-up of patients with progressive NASH . These non-invasive tools will also promote the clinical development of antifibrotic drugs, by permitting the design of lean proof-of-concept studies, and enabling development of a personalised antifibrotic therapy for patients with rapid fibrosis progression or advanced disease.
Collapse
Affiliation(s)
- Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| | - Rambabu Surabattula
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| | - Xiao Yu Wang
- Institute of Translational Immunology and Research Center for Immunotherapy, University of Mainz Medical Center, Mainz, Germany
| |
Collapse
|
528
|
Gao R, Zhu C, Li H, Yin M, Pan C, Huang L, Kong C, Wang X, Zhang Y, Qu S, Qin H. Dysbiosis Signatures of Gut Microbiota Along the Sequence from Healthy, Young Patients to Those with Overweight and Obesity. Obesity (Silver Spring) 2018; 26:351-361. [PMID: 29280312 DOI: 10.1002/oby.22088] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the gut microbiota in healthy volunteers (HVs), patients with overweight (OW), and patients with obesity (OB), including those with acanthosis nigricans (AN) or without AN (N-AN). METHODS Microbial 16S rRNA genes were examined by using pyrosequencing technology and analyzed by using bioinformatics methods. RESULTS Subjects in the OW and OB groups showed severe disturbances in glycemic control, lipid profile, and inflammatory markers (all P < 0.05); patients with AN had worse metabolic status (P < 0.001) and a lower diversity of microbiota (P < 0.05). The OB and HV groups showed totally different gut microbiota composition. In the OB group, beneficial microbiotas including Bifidobacterium (0.01% vs. 0.05%, false discovery rate [FDR] = 4.27*10-5 ), anti-inflammatory Faecalibacterium (6.70% vs. 13.82%, FDR = 0.010), and butyrate-producing Ruminococcaceae were significantly decreased, whereas Bacillus (0.58% vs. 0.04%, FDR = 0.013) and potential opportunistic pathogens such as Fusobacterium (1.44% vs. 0.11%, FDR < 0.01) and Escherichia-Shigella (6.01% vs. 0.76%, FDR = 0.041) had outgrown dramatically. Function prediction revealed a significant increase in lipopolysaccharide biosynthesis proteins and bacterial invasion of epithelial cell-associated genes and a significant decrease in glucose and essential amino acid-related genes. CONCLUSIONS Gut microbiotas and their functions were significantly changed in obesity. More prospective studies on association and causality between microbiota and obesity are imperative and might contribute to the prevention, diagnosis, and treatment of obesity.
Collapse
Affiliation(s)
- Renyuan Gao
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Cuiling Zhu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Li
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Mingming Yin
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Cheng Pan
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Linsheng Huang
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Cheng Kong
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Xingchun Wang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Zhang
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
529
|
Suriano F, Neyrinck AM, Verspreet J, Olivares M, Leclercq S, Van de Wiele T, Courtin CM, Cani PD, Bindels LB, Delzenne NM. Particle size determines the anti-inflammatory effect of wheat bran in a model of fructose over-consumption: Implication of the gut microbiota. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
530
|
Affiliation(s)
- Peng Chen
- Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou 510515, China.,Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
531
|
Saltzman ET, Palacios T, Thomsen M, Vitetta L. Intestinal Microbiome Shifts, Dysbiosis, Inflammation, and Non-alcoholic Fatty Liver Disease. Front Microbiol 2018; 9:61. [PMID: 29441049 PMCID: PMC5797576 DOI: 10.3389/fmicb.2018.00061] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/10/2018] [Indexed: 12/16/2022] Open
Abstract
Adverse fluctuations in the distribution of the intestinal microbiome cohort has been associated with the onset of intra- and extra-intestinal inflammatory conditions, like the metabolic syndrome (MetS) and it's hepatic manifestation, non-alcoholic fatty liver disease (NAFLD). The intestinal microbial community of obese compared to lean subjects has been shown to undergo configurational shifts in various genera, including but not limited to increased abundances of Prevotella, Escherichia, Peptoniphilus, and Parabacteroides and decreased levels of Bifidobacteria, Roseburia, and Eubacteria genera. At the phylum level, decreased Bacteroidetes and increased Firmicutes have been reported. The intestinal microbiota therefore presents an important target for designing novel therapeutic modalities that target extra-intestinal inflammatory disorders, such as NAFLD. This review hypothesizes that disruption of the intestinal-mucosal macrophage interface is a key factor in intestinal-liver axis disturbances. Intestinal immune responses implicated in the manifestation, maintenance and progression of NAFLD provide insights into the dialogue between the intestinal microbiome, the epithelia and mucosal immunity. The pro-inflammatory activity and immune imbalances implicated in NAFLD pathophysiology are reported to stem from dysbiosis of the intestinal epithelia which can serve as a source of hepatoxic effects. We posit that the hepatotoxic consequences of intestinal dysbiosis are compounded through intestinal microbiota-mediated inflammation of the local mucosa that encourages mucosal immune dysfunction, thus contributing important plausible insight in NAFLD pathogenesis. The administration of probiotics and prebiotics as a cure-all remedy for all chronic diseases is not advocated, instead, the incorporation of evidence based probiotic/prebiotic formulations as adjunctive modalities may enhance lifestyle modification management strategies for the amelioration of NAFLD.
Collapse
Affiliation(s)
- Emma T. Saltzman
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Talia Palacios
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Michael Thomsen
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Luis Vitetta
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| |
Collapse
|
532
|
Sun YL, Li WQ, Ding PX, Wang ZW, Wei CH, Ma XX, Zhang RF, Wu Y, Zhou L, Liang RP, Zhang YP, Zhao YP, Zhu RT, Li J. Specific alterations in gut microbiota are associated with prognosis of Budd-Chiari syndrome. Oncotarget 2018; 9:3303-3320. [PMID: 29423048 PMCID: PMC5790465 DOI: 10.18632/oncotarget.23234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Gut microbiota is associated with liver diseases. However, gut microbial characteristics of Budd-Chiari syndrome (B-CS) have not been reported. Here, by MiSeq sequencing, gut microbial alterations were characterized among 37 health controls, 20 liver cirrhosis (LC) patients, 31 initial B-CS patients (B-CS group), 33 stability patients after BCS treatment (stability group) and 23 recurrent patients after BCS treatment (recurrence group). Gut microbial diversity was increased in B-CS versus LC. Bacterial community of B-CS clustered with controls but separated from LC. Operational taxonomic units (OTUs) 421, 502 (Clostridium IV) and 141 (Megasphaera) were unique to B-CS. Genera Escherichia/Shigella and Clostridium XI were decreased in B-CS versus controls. Moreover, nine genera, mainly including Bacteroides and Megamonas, were enriched in B-CS versus LC. Notably, Megamonas could distinguish B-CS from LC with areas under the curve (AUCs) of 0.7904. Microbial function prediction revealed that L-amino acid transport system activity was decreased in B-CS versus both LC and controls. Furthermore, OTUs 27 (Clostridium XI), 137 (Clostridium XIVb) and 40 (Bacteroides) were associated with B-CS stability. Importantly, genus Clostridium XI was enriched in stability group versus both recurrence group and B-CS group. Also, PRPP glutamine biosynthesis was reduced in stability group versus recurrence group, but was enriched in stability group versus B-CS group. In conclusion, specific microbial alterations associated with diagnosis and prognosis were detected in B-CS patients. Correction of gut microbial alterations may be a potential strategy for B-CS prevention and treatment.
Collapse
Affiliation(s)
- Yu-Ling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Wen-Qi Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Peng-Xu Ding
- Department of Radioactive Intervention, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Zhi-Wei Wang
- Department of Radioactive Intervention, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Chang-Hua Wei
- Department of Ultrasound Diagnosis, The People’s Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiu-Xian Ma
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Rui-Fang Zhang
- Department of Ultrasound Diagnosis, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yan Wu
- Department of Radiology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Lin Zhou
- Department of Digestive Diseases, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Ruo-Peng Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Yan-Peng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Yi-Pu Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Rong-Tao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| | - Jian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
533
|
Milligan S, Martin GG, Landrock D, McIntosh AL, Mackie JT, Schroeder F, Kier AB. Ablating both Fabp1 and Scp2/Scpx (TKO) induces hepatic phospholipid and cholesterol accumulation in high fat-fed mice. Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:323-338. [PMID: 29307784 DOI: 10.1016/j.bbalip.2017.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/13/2017] [Accepted: 12/31/2017] [Indexed: 01/16/2023]
Abstract
Although singly ablating Fabp1 or Scp2/Scpx genes may exacerbate the impact of high fat diet (HFD) on whole body phenotype and non-alcoholic fatty liver disease (NAFLD), concomitant upregulation of the non-ablated gene, preference for ad libitum fed HFD, and sex differences complicate interpretation. Therefore, these issues were addressed in male and female mice ablated in both genes (Fabp1/Scp2/Scpx null or TKO) and pair-fed HFD. Wild-type (WT) males gained more body weight as fat tissue mass (FTM) and exhibited higher hepatic lipid accumulation than WT females. The greater hepatic lipid accumulation in WT males was associated with higher hepatic expression of enzymes in glyceride synthesis, higher hepatic bile acids, and upregulation of transporters involved in hepatic reuptake of serum bile acids. While TKO had little effect on whole body phenotype and hepatic bile acid accumulation in either sex, TKO increased hepatic accumulation of lipids in both, specifically phospholipid and cholesteryl esters in males and females and free cholesterol in females. TKO-induced increases in glycerides were attributed not only to complete loss of FABP1, SCP2 and SCPx, but also in part to sex-dependent upregulation of hepatic lipogenic enzymes. These data with WT and TKO mice pair-fed HFD indicate that: i) Sex significantly impacted the ability of HFD to increase body weight, induce hepatic lipid accumulation and increase hepatic bile acids; and ii) TKO exacerbated the HFD ability to induce hepatic lipid accumulation, regardless of sex, but did not significantly alter whole body phenotype in either sex.
Collapse
Affiliation(s)
- Sherrelle Milligan
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Gregory G Martin
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - Danilo Landrock
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Avery L McIntosh
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - John T Mackie
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA
| | - Friedhelm Schroeder
- Department of Physiology/Pharmacology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4466, USA
| | - Ann B Kier
- Department of Pathobiology, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA.
| |
Collapse
|
534
|
Wang W, Zhao J, Gui W, Sun D, Dai H, Xiao L, Chu H, Du F, Zhu Q, Schnabl B, Huang K, Yang L, Hou X. Tauroursodeoxycholic acid inhibits intestinal inflammation and barrier disruption in mice with non-alcoholic fatty liver disease. Br J Pharmacol 2018; 175:469-484. [PMID: 29139555 DOI: 10.1111/bph.14095] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 10/08/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The gut-liver axis is associated with the progression of non-alcoholic fatty liver disease (NAFLD). Targeting the gut-liver axis and bile acid-based pharmaceuticals are potential therapies for NAFLD. The effect of tauroursodeoxycholic acid (TUDCA), a candidate drug for NAFLD, on intestinal barrier function, intestinal inflammation, gut lipid transport and microbiota composition was analysed in a murine model of NAFLD. EXPERIMENTAL APPROACH The NAFLD mouse model was established by feeding mice a high-fat diet (HFD) for 16 weeks. TUDCA was administered p.o. during the last 4 weeks. The expression levels of intestinal tight junction genes, lipid metabolic and inflammatory genes were determined by quantitative PCR. Tissue inflammation was evaluated by haematoxylin and eosin staining. The gut microbiota was analysed by 16S rRNA gene sequencing. KEY RESULTS TUDCA administration attenuated HFD-induced hepatic steatosis, inflammatory responses, obesity and insulin resistance in mice. Moreover, TUDCA attenuated gut inflammatory responses as manifested by decreased intestinal histopathology scores and inflammatory cytokine levels. In addition, TUDCA improved intestinal barrier function by increasing levels of tight junction molecules and the solid chemical barrier. The components involved in ileum lipid transport were also reduced by TUDCA administration in HFD-fed mice. Finally, the TUDCA-treated mice showed a different gut microbiota composition compared with that in HFD-fed mice but similar to that in normal chow diet-fed mice. CONCLUSIONS AND IMPLICATIONS TUDCA attenuates the progression of HFD-induced NAFLD in mice by ameliorating gut inflammation, improving intestinal barrier function, decreasing intestinal fat transport and modulating intestinal microbiota composition.
Collapse
Affiliation(s)
- Weijun Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinfang Zhao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenfang Gui
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Sun
- Division of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haijiang Dai
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Du
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | - Bernd Schnabl
- Department of Medicine, Biomedical Research Facility 2 (BRF2), University of California, San Diego, La Jolla, CA, USA
| | - Kai Huang
- Division of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
535
|
Prieto I, Hidalgo M, Segarra AB, Martínez-Rodríguez AM, Cobo A, Ramírez M, Abriouel H, Gálvez A, Martínez-Cañamero M. Influence of a diet enriched with virgin olive oil or butter on mouse gut microbiota and its correlation to physiological and biochemical parameters related to metabolic syndrome. PLoS One 2018; 13:e0190368. [PMID: 29293629 PMCID: PMC5749780 DOI: 10.1371/journal.pone.0190368] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
The type of fat in the diet determinates the characteristics of gut microbiota, exerting a major role in the development of metabolic syndrome. We hypothesize that a diet enriched with extra virgin olive oil (EVOO) has a distinctive effect on the intestinal microbiome in comparison with an enriched butter diet (BT) and this effect is related to the physiological benefits exerted by EVOO. Swiss Webster mice were fed standard (SD) or two high fat diets enriched with EVOO or butter. Hormonal, physiological and metabolic parameters were evaluated. At the end of the feeding period, DNA was extracted from faeces and the 16S rRNA genes were pyrosequenced. Among the main significant differences found, BT triggered the highest values of systolic blood pressure, correlating positively with the percentage of Desulfovibrio sequences in faeces, which in turn showed significantly higher values in BT than in EVOO. EVOO had the lowest values of plasmatic insulin, correlating inversely with Desulfovibrio, and had the lowest plasmatic values of leptin which correlated inversely with Sutterellaceae, Marispirillum and Mucilaginibacter dageonensis, the three showing significantly higher percentages in EVOO. The lowest total cholesterol levels in plasma were detected in SD, correlating positively with Prevotella and Fusicatenibacter, both taxa with significantly greater presence in SD. These results may be indicative of a link between specific diets, certain physiological parameters and the prevalence of some taxa, supporting the possibility that in some of the proposed effects of virgin olive oil the modulation of intestinal microbiota could be involved.
Collapse
Affiliation(s)
- Isabel Prieto
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Marina Hidalgo
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Ana Belén Segarra
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | | | - Antonio Cobo
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Manuel Ramírez
- Área de Fisiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Hikmate Abriouel
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Antonio Gálvez
- Área de Microbiología, Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | | |
Collapse
|
536
|
Cassard AM, Gérard P, Perlemuter G. Microbiota, Liver Diseases, and Alcohol. BUGS AS DRUGS 2018:187-212. [DOI: 10.1128/9781555819705.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Affiliation(s)
- Anne-Marie Cassard
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay; 78350 Jouyen-Josas France
| | - Gabriel Perlemuter
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud; Université Paris-Saclay; 92140 Clamart France
- AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère; Clamart France
| |
Collapse
|
537
|
Hugenholtz F, de Vos WM. Mouse models for human intestinal microbiota research: a critical evaluation. Cell Mol Life Sci 2018; 75:149-160. [PMID: 29124307 PMCID: PMC5752736 DOI: 10.1007/s00018-017-2693-8] [Citation(s) in RCA: 365] [Impact Index Per Article: 52.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 09/29/2017] [Indexed: 02/06/2023]
Abstract
Since the early days of the intestinal microbiota research, mouse models have been used frequently to study the interaction of microbes with their host. However, to translate the knowledge gained from mouse studies to a human situation, the major spatio-temporal similarities and differences between intestinal microbiota in mice and humans need to be considered. This is done here with specific attention for the comparative physiology of the intestinal tract, the effect of dietary patterns and differences in genetics. Detailed phylogenetic and metagenomic analysis showed that while many common genera are found in the human and murine intestine, these differ strongly in abundance and in total only 4% of the bacterial genes are found to share considerable identity. Moreover, a large variety of murine strains is available yet most of the microbiota research is performed in wild-type, inbred strains and their transgenic derivatives. It has become increasingly clear that the providers, rearing facilities and the genetic background of these mice have a significant impact on the microbial composition and this is illustrated with recent experimental data. This may affect the reproducibility of mouse microbiota studies and their conclusions. Hence, future studies should take these into account to truly show the effect of diet, genotype or environmental factors on the microbial composition.
Collapse
Affiliation(s)
- Floor Hugenholtz
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, Building 124, 6708 WE, Wageningen, The Netherlands
- Division of Infectious Diseases, Department of Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, Building 124, 6708 WE, Wageningen, The Netherlands.
- Research Programme Unit Immunobiology, Department of Bacteriology and Immunology, Helsinki University, P.O. Box 21, 00014, Helsinki, Finland.
| |
Collapse
|
538
|
Wieser V, Adolph TE, Grander C, Grabherr F, Enrich B, Moser P, Moschen AR, Kaser S, Tilg H. Adipose type I interferon signalling protects against metabolic dysfunction. Gut 2018; 67:157-165. [PMID: 28011892 DOI: 10.1136/gutjnl-2016-313155] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/28/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Low-grade chronic inflammation emerges as a potent driver of insulin resistance and glucose dysregulation in obesity and associated non-alcoholic fatty liver disease (NAFLD). The liver, subcutaneous fat and the immune system participate in disturbances of metabolism. Type I interferon (IFN) signalling initiated by innate and adaptive immunity modulates inflammatory responses consequent to infection. However, little is known about the role of type I IFN signalling in metabolic diseases and the development of NAFLD. DESIGN We determined the impact of type I IFN signalling by tissue-specific deletion of interferon (α and β) receptor 1 (Ifnar1) in hepatocytes (Ifnar1Δhep ), adipocytes (Ifnar1Δat ), intestinal epithelial cells (Ifnar1ΔIEC ) or myelocytes (Ifnar1Δmyel ) on glucose metabolism, obesity and hepatic disease in mice exposed to a high-fat or methionine-choline-deficient (MCD) diet. Furthermore, we investigated the expression of type I IFN-regulated genes in patients with obesity undergoing laparoscopic adjustable gastric banding (LAGB). RESULTS Long chain fatty acids induce type I IFN responses in murine hepatocytes and macrophages and exposure to a high-fat diet elicited type I IFN-regulated gene expression in the liver of wild-type mice. Hepatocyte-specific, but not adipose tissue-specific deletion of Ifnar1 worsened steatosis and inflammation induced by the MCD diet. In contrast, adipose-specific, but not hepatocyte-specific deletion of Ifnar1 deteriorated metabolic dysregulation induced by a high-fat diet, indicated by increased weight gain, insulin resistance and an impaired glucose tolerance. Abrogated type I IFN signalling in myeloid or intestinal epithelial cells did not modulate susceptibility to metabolic or hepatic disease. Improved metabolic control in patients with obesity after LAGB was associated with increased expression of type I IFN-regulated genes in subcutaneous adipose tissue and liver. CONCLUSIONS Our study implicates a role for adipose and hepatocyte type I IFN signalling in diet-induced metabolic dysregulation and hepatic disease. Further studies on type I IFN signalling in metabolic diseases are warranted.
Collapse
Affiliation(s)
- Verena Wieser
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Timon Erik Adolph
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Barbara Enrich
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Patrizia Moser
- Institute of Pathology, Medical University Innsbruck, Innsbruck, Austria
| | - Alexander Rupert Moschen
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Susanne Kaser
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
539
|
Woodhouse CA, Patel VC, Singanayagam A, Shawcross DL. Review article: the gut microbiome as a therapeutic target in the pathogenesis and treatment of chronic liver disease. Aliment Pharmacol Ther 2018; 47:192-202. [PMID: 29083037 DOI: 10.1111/apt.14397] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 06/06/2017] [Accepted: 10/03/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Mortality from chronic liver disease is rising exponentially. The liver is intimately linked to the gut via the portal vein, and exposure to gut microbiota and their metabolites translocating across the gut lumen may impact upon both the healthy and diseased liver. Modulation of gut microbiota could prove to be a potential therapeutic target. AIM To characterise the changes in the gut microbiome that occur in chronic liver disease and to assess the impact of manipulation of the microbiome on the liver. METHODS We conducted a PubMed search using search terms including 'microbiome', 'liver' and 'cirrhosis' as well as 'non-alcoholic fatty liver disease', 'steatohepatitis', 'alcohol' and 'primary sclerosing cholangitis'. Relevant articles were also selected from references of articles and review of the ClinicalTrials.gov website. RESULTS Reduced bacterial diversity, alcohol sensitivity and the development of gut dysbiosis are seen in several chronic liver diseases, including non-alcoholic fatty liver disease, alcohol-related liver disease and primary sclerosing cholangitis. Perturbations in gut commensals could lead to deficient priming of the immune system predisposing the development of immune-mediated diseases. Furthermore, transfer of stool from an animal with the metabolic syndrome may induce steatosis in a healthy counterpart. Patients with cirrhosis develop dysbiosis, small bowel bacterial overgrowth and increased gut wall permeability, allowing bacterial translocation and uptake of endotoxin inducing hepatic and systemic inflammation. CONCLUSIONS Manipulation of the gut microbiota with diet, probiotics or faecal microbiota transplantation to promote the growth of "healthy" bacteria may ameliorate the dysbiosis and alter prognosis.
Collapse
Affiliation(s)
- C A Woodhouse
- Institute of Liver Studies and Transplantation, King's College London School of Medicine at King's College Hospital, London, UK
| | - V C Patel
- Institute of Liver Studies and Transplantation, King's College London School of Medicine at King's College Hospital, London, UK
| | - A Singanayagam
- Institute of Liver Studies and Transplantation, King's College London School of Medicine at King's College Hospital, London, UK
| | - D L Shawcross
- Institute of Liver Studies and Transplantation, King's College London School of Medicine at King's College Hospital, London, UK
| |
Collapse
|
540
|
Song M, Li X, Zhang X, Shi H, Vos MB, Wei X, Wang Y, Gao H, Rouchka EC, Yin X, Zhou Z, Prough RA, Cave MC, McClain CJ. Dietary copper-fructose interactions alter gut microbial activity in male rats. Am J Physiol Gastrointest Liver Physiol 2018; 314:G119-G130. [PMID: 29025734 PMCID: PMC5866377 DOI: 10.1152/ajpgi.00378.2016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 02/08/2023]
Abstract
Dietary copper-fructose interactions contribute to the development of nonalcoholic fatty liver disease (NAFLD). Gut microbiota play critical roles in the pathogenesis of NAFLD. The aim of this study was to determine the effect of different dietary doses of copper and their interactions with high fructose on gut microbiome. Male weanling Sprague-Dawley rats were fed diets with adequate copper (6 ppm CuA), marginal copper (1.5 ppm CuM) (low copper), or supplemented copper (20 ppm CuS) (high copper) for 4 wk. Deionized water or deionized water containing 30% fructose (wt/vol) was given ad libitum. Copper status, liver enzymes, gut barrier function, and gut microbiome were evaluated. Both low- and high-copper diets led to liver injury in high-fructose-fed rats, and this was associated with gut barrier dysfunction, as shown by the markedly decreased tight junction proteins and increased gut permeability. 16S rDNA sequencing analysis revealed distinct alterations of the gut microbiome associated with dietary low- and high-copper/high-fructose feeding. The common features of the alterations of the gut microbiome were the increased abundance of Firmicutes and the depletion of Akkermansia. However, they differed mainly within the phylum Firmicutes. Our data demonstrated that a complex interplay among host, microbes, and dietary copper-fructose interaction regulates gut microbial metabolic activity, which may contribute to the development of liver injury and hepatic steatosis. The distinct alterations of gut microbial activity, which were associated with the different dietary doses of copper and fructose, imply that separate mechanism(s) may be involved. NEW & NOTEWORTHY First, dietary low- and high-copper/high-fructose-induced liver injury are associated with distinct alterations of gut microbiome. Second, dietary copper level plays a critical role in maintaining the gut barrier integrity, likely by acting on the intestinal tight junction proteins and the protective commensal bacteria Akkermansia. Third, the alterations of gut microbiome induced by dietary low and high copper with or without fructose differ mainly within the phylum Firmicutes.
Collapse
Affiliation(s)
- Ming Song
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
| | - Xiaohong Li
- Bioinformatics Core. University of Louisville School of Medicine , Louisville, Kentucky
| | - Xiang Zhang
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Hongxue Shi
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
| | - Miriam B Vos
- Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, Georgia
| | - Xiaoli Wei
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Yuhua Wang
- College of Food Science and Engineering, Jilin Agricultural University , Changchun , China
| | - Hong Gao
- Genomics Facility, University of Louisville School of Medicine , Louisville, Kentucky
| | - Eric C Rouchka
- Bioinformatics Core. University of Louisville School of Medicine , Louisville, Kentucky
| | - Xinmin Yin
- Department of Chemistry, University of Louisville School of Medicine , Louisville, Kentucky
- Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, Kannapolis, North Carolina
- Department of Nutrition, University of North Carolina at Greensboro, Kannapolis, North Carolina
| | - Russell A Prough
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine , Louisville, Kentucky
| | - Matthew C Cave
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine , Louisville, Kentucky
- Robley Rex Louisville Veterans Afairs Medical Center , Louisville, Kentucky
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine , Louisville, Kentucky
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine , Louisville, Kentucky
- Hepatobiology and Toxicology Center, University of Louisville School of Medicine , Louisville, Kentucky
- University of Louisville Alcohol Research Center, University of Louisville School of Medicine , Louisville, Kentucky
- Robley Rex Louisville Veterans Afairs Medical Center , Louisville, Kentucky
| |
Collapse
|
541
|
Ikegami T, Honda A. Reciprocal interactions between bile acids and gut microbiota in human liver diseases. Hepatol Res 2018; 48:15-27. [PMID: 29150974 DOI: 10.1111/hepr.13001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/08/2023]
Abstract
The gut microbiota (GM) play a central role in their host's metabolism of bile acids (BAs) by regulating deconjugation, dehydroxylation, dehydrogenation, and epimerization reactions to generate unconjugated free BAs and secondary BAs. These BAs generated by the GM are potent signaling molecules that interact with BA receptors, such as the farnesoid X receptor and Takeda G-protein-coupled receptor 5. Each BA has a differential affinity to these receptors; therefore, alterations in BA composition by GM could modify the intensity of receptor signaling. Bile acids also act as antimicrobial agents by damaging bacterial membranes and as detergents by altering intracellular macromolecular structures. Therefore, BAs and the GM reciprocally control each other's compositions. In this review, we discuss the latest findings on the mutual effects of BAs and GM on each other; we also describe their roles in the pathophysiology of liver disease progression and potential therapeutic applications of targeting this cross-talk.
Collapse
Affiliation(s)
- Tadashi Ikegami
- Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Akira Honda
- Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan.,Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
542
|
Jia W, Rajani C. The Influence of Gut Microbial Metabolism on the Development and Progression of Non-alcoholic Fatty Liver Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1061:95-110. [PMID: 29956209 DOI: 10.1007/978-981-10-8684-7_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is defined as the presence of excess fat in the liver parenchyma in the absence of excess alcohol consumption and overt inflammation. It has also been described as the hepatic manifestation of metabolic syndrome (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015). The incidence of NAFLD has been reported to be 43-60% in diabetics, ~90% in patients with hyperlipidemia and 91% in morbidly obese patients (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015, Machado M, Marques-Vidal P, Cortez-Pinto H, J Hepatol, 45:600-606, 2006, Vernon G, Baranova A, Younossi ZM, Aliment Pharmacol Ther, 34:274-285, 2011). The risk factors that have been associated with the development of NAFLD include male gender, increasing age, obesity, insulin resistance, diabetes and hyperlipidemia (Attar BM, Van Thiel DH, Sci World J, 2013:481893, 2013, Gaggini M, Morelli M, Buzzigoli E, DeFronzo RA, Bugianesi E, Gastaldelli A, Forum Nutr, 5:1544-1460, 2013). All of these risk factors have been linked to alterations of the gut microbiota, ie., gut dysbiosis (He X, Ji G, Jia W, Li H, Int J Mol Sci, 17:300, 2016). However, it must be pointed out that the prevalence of NAFLD in normal weight individuals without metabolic risk factors is ~16% (Than NN, Newsome PN, Atherosclerosis. 239:192-202, 2015). This fact has led some investigators to hypothesize that the gut microbiota can impact lipid metabolism in the liver independently of obesity-related metabolic factors (Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold g, et al., Gut, 65:330 339, 2016) (Le Roy T, Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, et al., Gut, 62:1787-1794, 2013). In this chapter, we will explore the effect of the gut microbiota on hepatic lipid metabolism and how this affects the development of NAFLD.
Collapse
Affiliation(s)
- Wei Jia
- University of Hawaii Cancer Center, Honolulu, HI, USA.
- Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | | |
Collapse
|
543
|
Panasevich MR, Meers GM, Linden MA, Booth FW, Perfield JW, Fritsche KL, Wankhade UD, Chintapalli SV, Shankar K, Ibdah JA, Rector RS. High-fat, high-fructose, high-cholesterol feeding causes severe NASH and cecal microbiota dysbiosis in juvenile Ossabaw swine. Am J Physiol Endocrinol Metab 2018; 314:E78-E92. [PMID: 28899857 PMCID: PMC5866386 DOI: 10.1152/ajpendo.00015.2017] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 09/07/2017] [Accepted: 09/07/2017] [Indexed: 02/07/2023]
Abstract
Pediatric obesity and nonalcoholic steatohepatitis (NASH) are on the rise in industrialized countries, yet our ability to mechanistically examine this relationship is limited by the lack of a suitable higher animal models. Here, we examined the effects of high-fat, high-fructose corn syrup, high-cholesterol Western-style diet (WD)-induced obesity on NASH and cecal microbiota dysbiosis in juvenile Ossabaw swine. Juvenile female Ossabaw swine (5 wk old) were fed WD (43.0% fat; 17.8% high-fructose corn syrup; 2% cholesterol) or low-fat diet (CON/lean; 10.5% fat) for 16 wk ( n = 6 each) or 36 wk ( n = 4 each). WD-fed pigs developed obesity, dyslipidemia, and systemic insulin resistance compared with CON pigs. In addition, obese WD-fed pigs developed severe NASH, with hepatic steatosis, hepatocyte ballooning, inflammatory cell infiltration, and fibrosis after 16 wk, with further exacerbation of histological inflammation and fibrosis after 36 wk of WD feeding. WD feeding also resulted in robust cecal microbiota changes including increased relative abundances of families and genera in Proteobacteria ( P < 0.05) (i.e., Enterobacteriaceae, Succinivibrionaceae, and Succinivibrio) and LPS-containing Desulfovibrionaceae and Desulfovibrio and a greater ( P < 0.05) predicted microbial metabolic function for LPS biosynthesis, LPS biosynthesis proteins, and peptidoglycan synthesis compared with CON-fed pigs. Overall, juvenile Ossabaw swine fed a high-fat, high-fructose, high-cholesterol diet develop obesity and severe microbiota dysbiosis with a proinflammatory signature and a NASH phenotype directly relevant to the pediatric/adolescent and young adult population.
Collapse
Affiliation(s)
- M. R. Panasevich
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - G. M. Meers
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - M. A. Linden
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - F. W. Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - J. W. Perfield
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Department of Food Science, University of Missouri, Columbia, Missouri
| | - K. L. Fritsche
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
| | - Umesh D. Wankhade
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Sree V. Chintapalli
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - K. Shankar
- Department of Pediatrics, Arkansas Children’s Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - J. A. Ibdah
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| | - R. S. Rector
- Research Service, Harry S. Truman Memorial Veterans Affairs Hospital, Columbia, Missouri
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
544
|
Machado MV, Diehl AM. Pathogenesis of Nonalcoholic Fatty Liver Disease. ZAKIM AND BOYER'S HEPATOLOGY 2018:369-390.e14. [DOI: 10.1016/b978-0-323-37591-7.00025-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
545
|
Tang C, Sun J, Zhou B, Jin C, Liu J, Kan J, Qian C, Zhang N. Effects of polysaccharides from purple sweet potatoes on immune response and gut microbiota composition in normal and cyclophosphamide treated mice. Food Funct 2018; 9:937-950. [DOI: 10.1039/c7fo01302g] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Three polysaccharides were extracted from purple sweet potatoes and then administered to normal and cyclophosphamide treated mice by gavage.
Collapse
Affiliation(s)
- Chao Tang
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Jian Sun
- College of Chemistry and Chemical Engineering
- Yangzhou University
- Yangzhou 225002
- China
- Xuzhou Institute of Agricultural Sciences in Jiangsu Xuhuai Area
| | - Bo Zhou
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Changhai Jin
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Jun Liu
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Juan Kan
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Chunlu Qian
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| | - Nianfeng Zhang
- College of Food Science and Engineering
- Yangzhou University
- Yangzhou 225127
- China
| |
Collapse
|
546
|
Microbial Regulation of Glucose Metabolism and Insulin Resistance. Genes (Basel) 2017; 9:genes9010010. [PMID: 29286343 PMCID: PMC5793163 DOI: 10.3390/genes9010010] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes is a combined disease, resulting from a hyperglycemia and peripheral and hepatic insulin resistance. Recent data suggest that the gut microbiota is involved in diabetes development, altering metabolic processes including glucose and fatty acid metabolism. Thus, type 2 diabetes patients show a microbial dysbiosis, with reduced butyrate-producing bacteria and elevated potential pathogens compared to metabolically healthy individuals. Furthermore, probiotics are a known tool to modulate the microbiota, having a therapeutic potential. Current literature will be discussed to elucidate the complex interaction of gut microbiota, intestinal permeability and inflammation leading to peripheral and hepatic insulin resistance. Therefore, this review aims to generate a deeper understanding of the underlying mechanism of potential microbial strains, which can be used as probiotics.
Collapse
|
547
|
Prebiotic effect of predigested mango peel on gut microbiota assessed in a dynamic in vitro model of the human colon (TIM-2). Food Res Int 2017; 118:89-95. [PMID: 30898357 DOI: 10.1016/j.foodres.2017.12.024] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/05/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
Abstract
Mango (Mangifera indica L.) peel (MP), is a by-product from the industrial processing to obtain juices and concentrates, and is rich in polyphenols and dietary fiber (DF). DF content of dried MP is about 40%. The aim of this study was to determine the prebiotic potential of this by-product submitting predigested mango ('Ataulfo') peel to a dynamic in vitro model of the human colon. Dried MPs were predigested following an enzymatic treatment and separating digestion products and undigested material by diafiltration. The predigested samples were fermented in a validated in vitro model of the colon (TIM-2) using human fecal microbiota and sampled after 0, 24, 48 and 72h. A carbohydrate mixture of standard ileal effluent medium (SIEM) was used as control. Production of short chain fatty acids (SCFA), branched chain fatty acids (BCFA) and ammonia profiles were determined in both lumen and dialysates. Microbiota composition was determined by sequencing 16S rRNA gene V3-V4 region. Principal component (PC) analysis of fermentation metabolites and relative abundance of genera was carried out. Fermentation of MP resulted in SCFA concentrations resembling those found in the SIEM experiments, with a 56:19:24 molar ratio for acetic, propionic and butyric acids, respectively. BCFA and ammonia were produced in similar concentrations in both samples. About 80 bacterial genera were identified after fermentation of MP, with an 83% relative abundance of Bifidobacterium at 24h. Three PC were identified; PC1 was influenced by a high Bifidobacterium abundance and low metabolites production. PC2 resulted in a decrease of other genera and an increase of metabolites studied. The relative abundance at 72h in MP was distributed over 4 genera Bifidobacterium, Lactobacillus, Dorea, and Lactococcus. Our results suggest MP as a potential prebiotic ingredient.
Collapse
|
548
|
Mörkl S, Lackner S, Müller W, Gorkiewicz G, Kashofer K, Oberascher A, Painold A, Holl A, Holzer P, Meinitzer A, Mangge H, Holasek S. Gut microbiota and body composition in anorexia nervosa inpatients in comparison to athletes, overweight, obese, and normal weight controls. Int J Eat Disord 2017; 50:1421-1431. [PMID: 29131365 DOI: 10.1002/eat.22801] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/03/2017] [Accepted: 10/24/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Anorexia nervosa (AN) is a heterogeneous eating disorder associated with alterations of body structure and the gut microbiome. We aimed to investigate the gut microbiota composition of a large female cohort including different BMI groups and activity levels along with body composition parameters. METHOD 106 female participants were included in this cross-sectional study: AN patients (n = 18), athletes (n = 20), normal weight (n = 26), overweight (n = 22), and obese women (n = 20). DNA was extracted from stool samples and subjected to 16S rRNA gene analysis. The software Quantitative Insights Into Microbial Ecology (QIIME) was used to analyze data. Additionally, we performed anthropometric assessments, ultrasound measurements of subcutaneous adipose tissue thickness, bioimpedance analysis, administered depression inventories, and ascertained laboratory parameters and dietary intakes. RESULTS Alpha diversity was particularly lower in AN patients and obese participants compared to other groups, while athletes showed highest alpha diversity. Several categories significantly associated with community structure were identified: body fat parameters, serum lipids, CRP, depression scales and smoking. Comparative analysis revealed Coriobacteriaceae as the only enriched phylotype in AN compared to other entities (LDA score >3.5). DISCUSSION This study provides further evidence of intestinal dysbiosis in AN and sheds light on characteristics of the gut microbiome in different BMI and physical activity groups. These insights point to new modulation possibilities of the gut microbiota which could improve the standard therapy of AN.
Collapse
Affiliation(s)
- Sabrina Mörkl
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Sonja Lackner
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Wolfram Müller
- Institute of Biophysics, Medical University of Graz, Graz, Austria
| | | | - Karl Kashofer
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Andreas Oberascher
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Annamaria Painold
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Anna Holl
- Department of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Peter Holzer
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Andreas Meinitzer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sandra Holasek
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| |
Collapse
|
549
|
Castaño-Rodríguez N, Mitchell HM, Kaakoush NO. NAFLD, Helicobacter species and the intestinal microbiome. Best Pract Res Clin Gastroenterol 2017; 31:657-668. [PMID: 29566909 DOI: 10.1016/j.bpg.2017.09.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/03/2017] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide. It is well-accepted that gut dysbiosis is associated with NAFLD, however, there is some conflicting evidence regarding the nature of these alterations. Infection with Helicobacter species, mainly H. pylori, has also been associated with increased NAFLD risk, however, some studies have failed to reproduce this finding. Further studies including large study samples and standardised procedures for microbiota analyses, H. pylori detection and NAFLD diagnostic criteria, are required. The mechanisms involving Helicobacter species and the intestinal microbiome in NAFLD pathogenesis appear to be part of the multiple-hit theory, in which increased intestinal permeability, inflammatory responses, altered choline, bile acids and carbohydrate metabolism, production of short-chain fatty acids, urea cycle and urea transport systems, altered maintenance of hepatic γδT-17 cells, insulin resistance, hormones secreted by the adipose tissue, metabolic hormones, bacterial metabolites and Helicobacter toxins, are all implicated.
Collapse
Affiliation(s)
| | - Hazel M Mitchell
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Nadeem O Kaakoush
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
550
|
Impact of Time-Restricted Feeding and Dawn-to-Sunset Fasting on Circadian Rhythm, Obesity, Metabolic Syndrome, and Nonalcoholic Fatty Liver Disease. Gastroenterol Res Pract 2017; 2017:3932491. [PMID: 29348746 PMCID: PMC5733887 DOI: 10.1155/2017/3932491] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 10/01/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity now affects millions of people and places them at risk of developing metabolic syndrome, nonalcoholic fatty liver disease (NAFLD), and even hepatocellular carcinoma. This rapidly emerging epidemic has led to a search for cost-effective methods to prevent the metabolic syndrome and NAFLD as well as the progression of NAFLD to cirrhosis and hepatocellular carcinoma. In murine models, time-restricted feeding resets the hepatic circadian clock and enhances transcription of key metabolic regulators of glucose and lipid homeostasis. Studies of the effect of dawn-to-sunset Ramadan fasting, which is akin to time-restricted feeding model, have also identified significant improvement in body mass index, serum lipid profiles, and oxidative stress parameters. Based on the findings of studies conducted on human subjects, dawn-to-sunset fasting has the potential to be a cost-effective intervention for obesity, metabolic syndrome, and NAFLD.
Collapse
|