51
|
Characterization and description of Faecalibacterium butyricigenerans sp. nov. and F. longum sp. nov., isolated from human faeces. Sci Rep 2021; 11:11340. [PMID: 34059761 PMCID: PMC8166934 DOI: 10.1038/s41598-021-90786-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Exploiting a pure culture strategy to investigate the composition of the human gut microbiota, two novel anaerobes, designated strains AF52-21T and CM04-06T, were isolated from faeces of two healthy Chinese donors and characterized using a polyphasic approach. The two strains were observed to be gram-negative, non-motile, and rod-shaped. Both strains grew optimally at 37 °C and pH 7.0. Phylogenetic analysis based on 16S rRNA gene sequences revealed that the two strains clustered with species of the genus Faecalibacterium and were most closely related to Faecalibacterium prausnitzii ATCC 27768T with sequence similarity of 97.18% and 96.87%, respectively. The two isolates shared a 16S rRNA gene sequence identity of 98.69%. Draft genome sequencing was performed for strains AF52-21T and CM04-06T, generating genome sizes of 2.85 Mbp and 3.01 Mbp. The calculated average nucleotide identity values between the genomes of the strains AF52-21T and CM04-06T compared to Faecalibacterium prausnitzii ATCC 27768T were 83.20% and 82.54%, respectively, and 90.09% when comparing AF52-21T and CM04-06T. Both values were below the previously proposed species threshold (95–96%), supporting their recognition as novel species in the genus Faecalibacterium. The genomic DNA G + C contents of strains AF52-21T and CM04-06T calculated from genome sequences were 57.77 mol% and 57.51 mol%, respectively. Based on the phenotypic, chemotaxonomic and phylogenetic characteristics, we conclude that both strains represent two new Faecalibacterium species, for which the names Faecalibacterium butyricigenerans sp. nov. (type strain AF52-21T = CGMCC 1.5206T = DSM 103434T) and Faecalibacterium longum sp. nov. (type strain CM04-06T = CGMCC 1.5208T = DSM 103432T) are proposed.
Collapse
|
52
|
Vancells Lujan P, Viñas Esmel E, Sacanella Meseguer E. Overview of Non-Alcoholic Fatty Liver Disease (NAFLD) and the Role of Sugary Food Consumption and Other Dietary Components in Its Development. Nutrients 2021; 13:nu13051442. [PMID: 33923255 PMCID: PMC8145877 DOI: 10.3390/nu13051442] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
NAFLD is the world's most common chronic liver disease, and its increasing prevalence parallels the global rise in diabetes and obesity. It is characterised by fat accumulation in the liver evolving to non-alcoholic steatohepatitis (NASH), an inflammatory subtype that can lead to liver fibrosis and cirrhosis. Currently, there is no effective pharmacotherapeutic treatment for NAFLD. Treatment is therefore based on lifestyle modifications including changes to diet and exercise, although it is unclear what the most effective form of intervention is. The aim of this review, then, is to discuss the role of specific nutrients and the effects of different dietary interventions on NAFLD. It is well established that an unhealthy diet rich in calories, sugars, and saturated fats and low in polyunsaturated fatty acids, fibre, and micronutrients plays a critical role in the development and progression of this disease. However, few clinical trials have evaluated the effects of nutrition interventions on NAFLD. We, therefore, summarise what is currently known about the effects of macronutrients, foods, and dietary patterns on NAFLD prevention and treatment. Most current guidelines recommend low-calorie, plant-based diets, such as the Mediterranean diet, as the most effective dietary pattern to treat NAFLD. More clinical trials are required, however, to identify the best evidence-based dietary treatment approach.
Collapse
Affiliation(s)
- Pau Vancells Lujan
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (P.V.L.); (E.V.E.)
| | - Esther Viñas Esmel
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (P.V.L.); (E.V.E.)
- Department of Internal Medicine, Hospital Clínic de Barcelona, Villarroel 170, 08036 Barcelona, Spain
| | - Emilio Sacanella Meseguer
- Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain; (P.V.L.); (E.V.E.)
- Department of Internal Medicine, Hospital Clínic de Barcelona, Villarroel 170, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-932-275539
| |
Collapse
|
53
|
Abstract
The increasing prevalence of non-alcoholic fatty liver disease (NAFLD) poses a growing challenge in terms of its prevention and treatment. The 'multiple hits' hypothesis of multiple insults, such as dietary fat intake, de novo lipogenesis, insulin resistance, oxidative stress, mitochondrial dysfunction, gut dysbiosis and hepatic inflammation, can provide a more accurate explanation of the pathogenesis of NAFLD. Betaine plays important roles in regulating the genes associated with NAFLD through anti-inflammatory effects, increased free fatty oxidation, anti-lipogenic effects and improved insulin resistance and mitochondrial function; however, the mechanism of betaine remains elusive.
Collapse
|
54
|
Villard A, Boursier J, Andriantsitohaina R. Bacterial and eukaryotic extracellular vesicles and nonalcoholic fatty liver disease: new players in the gut-liver axis? Am J Physiol Gastrointest Liver Physiol 2021; 320:G485-G495. [PMID: 33471632 DOI: 10.1152/ajpgi.00362.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The liver and intestine communicate in a bidirectional way through the biliary tract, portal vein, and other components of the gut-liver axis. The gut microbiota is one of the major contributors to the production of several proteins and bile acids. Imbalance in the gut bacterial community, called dysbiosis, participates in the development and progression of several chronic liver diseases, such as nonalcoholic fatty liver disease (NAFLD). NAFLD is currently considered the main chronic liver disease worldwide. Dysbiosis contributes to NAFLD development and progression, notably by a greater translocation of pathogen-associated molecular patterns (PAMPs) in the blood. Lipopolysaccharide (LPS) is a PAMP that activates Toll-like receptor 4 (TLR4), induces liver inflammation, and participates in the development of fibrogenesis. LPS can be transported by bacterial extracellular vesicles (EVs). EVs are spherical structures produced by eukaryotic and prokaryotic cells that transfer information to distant cells and may represent new players in NAFLD development and progression. The present review summarizes the role of eukaryotic EVs, either circulating or tissue-derived, in NAFLD features, such as liver inflammation, angiogenesis, and fibrosis. Circulating EV levels are dynamic and correlate with disease stage and severity. However, scarce information is available concerning the involvement of bacterial EVs in liver disease. The present review highlights a potential role of bacterial EVs in insulin resistance and liver inflammation, although the mechanism involved has not been elucidated. In addition, because of their distinct signatures, eukaryotic and prokaryotic EVs may also represent a promising NAFLD diagnostic tool as a "liquid biopsy" in the future.
Collapse
Affiliation(s)
- Alexandre Villard
- INSERM UMR1063, Stress Oxydant et Pathologies Métaboliques, Faculté de Santé, Université d'Angers, Université Bretagne Loire, Angers, France.,EA 3859, Hémodynamique, Interaction Fibrose et Invasivité Tumorales Hépatiques (HIFIH), Angers, France
| | - Jérôme Boursier
- EA 3859, Hémodynamique, Interaction Fibrose et Invasivité Tumorales Hépatiques (HIFIH), Angers, France
| | - Ramaroson Andriantsitohaina
- INSERM UMR1063, Stress Oxydant et Pathologies Métaboliques, Faculté de Santé, Université d'Angers, Université Bretagne Loire, Angers, France
| |
Collapse
|
55
|
Jiang H, Zhang Y, Xu D, Wang Q. Probiotics ameliorates glycemic control of patients with diabetic nephropathy: A randomized clinical study. J Clin Lab Anal 2021; 35:e23650. [PMID: 33666270 PMCID: PMC8059722 DOI: 10.1002/jcla.23650] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE This research aimed to explore the effects of probiotic administration on glycemic control and renal function in patients with diabetic nephropathy (DN). METHODS The 101 participants were randomly divided into two treatment groups and 76 patients were included in the final analysis. In 76 patients with diabetic nephropathy of type 2 diabetes, a randomized double-blind and placebo-controlled clinical trial was conducted to evaluate the administration of 3.2 × 109 CFU probiotic supplements per day (Bifidobacterium bifidum, 1.2 × 109 CFU, Lactobacillus acidophilus 4.2 × 109 CFU, Streptococcus thermophilus 4.3 × 109 CFU) for 12 weeks on glycemic control of patients, including fasting blood glucose, 2 h postprandial blood glucose, glycosylated hemoglobin (HbA1c), microalbuminuria/creatinine (mAlb/Cr) and estimated glomerular filtration rate (eGFR) levels. The placebo group daily received empty capsules filled with starch. RESULTS After 12 weeks, the administration of probiotics demonstrated a significant reduction in fasting blood glucose (10.68 ± 3.24 mmol/L before vs. 7.81 ± 2.77 mmol/L after, p < 0.05), HbA1c (8.19 ± 1.60% before vs. 7.32 ± 1.20% after, p < 0.05) and mAlb/Cr (101.60 ± 22.17 mg/g before vs. 67.53 ± 20.11 mg/g after, p < 0.05), while only mAlb/Cr level was significantly lower in the probiotic group than in the placebo group after intervention (67.53 ± 20.11 mg/g vs. 87.71 ± 23.01, p < 0.05). Meanwhile, there was no significant reduction of 2 h postprandial blood glucose level (18.95 ± 5.23 mmol/L vs. 17.35 ± 6.28 mmol/L, p = 0.24) and eGFR (84.34 ± 6.97 ml/min vs. 82.8 ± 8.72 ml/min, p = 0.45) in patients before and after probiotic intake. In addition, the placebo group failed to show any significant change of these parameters. CONCLUSION This clinical study revealed probiotic administration could ameliorate glycemic control of patients with diabetic nephropathy, potentiating its therapeutic potential in clinical application.
Collapse
Affiliation(s)
- Hongyang Jiang
- China‐Japan Union Hospital Affiliated Jilin UniversityChangchunChina
| | - Yan Zhang
- China‐Japan Union Hospital Affiliated Jilin UniversityChangchunChina
| | - Dongyan Xu
- China‐Japan Union Hospital Affiliated Jilin UniversityChangchunChina
| | - Qing Wang
- China‐Japan Union Hospital Affiliated Jilin UniversityChangchunChina
| |
Collapse
|
56
|
Oh JH, Lee JH, Cho MS, Kim H, Chun J, Lee JH, Yoon Y, Kang W. Characterization of Gut Microbiome in Korean Patients with Metabolic Associated Fatty Liver Disease. Nutrients 2021; 13:nu13031013. [PMID: 33801023 PMCID: PMC8004024 DOI: 10.3390/nu13031013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic associated fatty liver disease (MAFLD) is a new concept where the presence of both fatty liver and metabolic abnormality are necessary for diagnosis. Several studies have reported that altered gut microbiome is closely associated with metabolic diseases and non-alcoholic fatty liver disease. However, the studies on MAFLD population are scarce. This prospective study aimed to identify differences in gut microbiome between patients with MAFLD and healthy controls in Korean population. In this study, patients with MAFLD and age, sex-matched healthy controls were included, and their stool samples were collected. Taxonomic composition of gut microbiota was analyzed using 16S ribosomal ribonucleic acid pyrosequencing. Twenty-two MAFLD patients and 44 healthy controls were included. Taxonomic diversity was lower in patients with MAFLD in the aspect of alpha and beta diversity. The differences were also found at phylum, class, family, and genus levels between the two groups. Phylum Proteobacteria, family Enterobactereriaceae, genus Citrobacter abundance was significantly increased and genus Faecalibacterium was significantly decreased in patients with MAFLD. In addition, butyrate-producing bacteria were decreased and ethanol-producing bacteria were increased in patients with MAFLD. The composition of gut microbiome was different between MAFLD and healthy controls in Korean population. This could offer potential targets for therapeutic intervention in MAFLD.
Collapse
Affiliation(s)
- Joo Hyun Oh
- Samsung Medical Center, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.H.O.); (J.H.L.)
- Department of Medicine, Eulji General Hospital, Eulji University School of Medicine, Seoul 01830, Korea
| | - Je Hee Lee
- ChunLab, Inc., Seoul 06194, Korea; (J.H.L.); (M.S.C.); (J.C.)
| | - Min Seok Cho
- ChunLab, Inc., Seoul 06194, Korea; (J.H.L.); (M.S.C.); (J.C.)
| | - Hyeree Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea;
- Samsung Medical Center, Institute for Future Medicine, Seoul 06351, Korea
| | - Jongsik Chun
- ChunLab, Inc., Seoul 06194, Korea; (J.H.L.); (M.S.C.); (J.C.)
| | - Joon Hyeok Lee
- Samsung Medical Center, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.H.O.); (J.H.L.)
| | - Yeup Yoon
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea;
- Samsung Medical Center, Institute for Future Medicine, Seoul 06351, Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (Y.Y.); (W.K.)
| | - Wonseok Kang
- Samsung Medical Center, Department of Medicine, Sungkyunkwan University School of Medicine, Seoul 06351, Korea; (J.H.O.); (J.H.L.)
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University School of Medicine, Seoul 06355, Korea;
- Samsung Medical Center, Institute for Future Medicine, Seoul 06351, Korea
- Correspondence: (Y.Y.); (W.K.)
| |
Collapse
|
57
|
Bhat M, Usmani SE, Azhie A, Woo M. Metabolic Consequences of Solid Organ Transplantation. Endocr Rev 2021; 42:171-197. [PMID: 33247713 DOI: 10.1210/endrev/bnaa030] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/12/2022]
Abstract
Metabolic complications affect over 50% of solid organ transplant recipients. These include posttransplant diabetes, nonalcoholic fatty liver disease, dyslipidemia, and obesity. Preexisting metabolic disease is further exacerbated with immunosuppression and posttransplant weight gain. Patients transition from a state of cachexia induced by end-organ disease to a pro-anabolic state after transplant due to weight gain, sedentary lifestyle, and suboptimal dietary habits in the setting of immunosuppression. Specific immunosuppressants have different metabolic effects, although all the foundation/maintenance immunosuppressants (calcineurin inhibitors, mTOR inhibitors) increase the risk of metabolic disease. In this comprehensive review, we summarize the emerging knowledge of the molecular pathogenesis of these different metabolic complications, and the potential genetic contribution (recipient +/- donor) to these conditions. These metabolic complications impact both graft and patient survival, particularly increasing the risk of cardiovascular and cancer-associated mortality. The current evidence for prevention and therapeutic management of posttransplant metabolic conditions is provided while highlighting gaps for future avenues in translational research.
Collapse
Affiliation(s)
- Mamatha Bhat
- Multi Organ Transplant program and Division of Gastroenterology & Hepatology, University Health Network, Ontario M5G 2N2, Department of Medicine, University of Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shirine E Usmani
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, Ontario, and Sinai Health System, Ontario, University of Toronto, Toronto, Ontario, Canada
| | - Amirhossein Azhie
- Multi Organ Transplant program and Division of Gastroenterology & Hepatology, University Health Network, Ontario M5G 2N2, Department of Medicine, University of Toronto, Ontario, Canada.,Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, Ontario, and Sinai Health System, Ontario, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
58
|
Wang T, Chen L, Huang P, Yang T, Zhang S, Zhao L, Chen L, Ye Z, Luo L, Qin J. Association of maternal gut microbiota and plasma metabolism with congenital heart disease in offspring: a multi-omic analysis. Sci Rep 2021; 11:5339. [PMID: 33674681 PMCID: PMC7935922 DOI: 10.1038/s41598-021-84901-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/22/2021] [Indexed: 12/22/2022] Open
Abstract
Congenital heart disease (CHD) is the most common congenital disorder diagnosed in newborns. Although lots of related studies have been published, yet the pathogenesis has not been fully elucidated. A growing body of evidence indicates perturbations of the gut microbiota may contribute in a significant way to the development of obesity and diabetes. Given that maternal obesity and diabetes are well-known risk factors for CHD, maternal gut microbiota may be considered as one of the environmental factors involved in the pathogenesis of CHD. The object of this study is to explore the association between maternal gut microbiota and risk of congenital heart disease (CHD) in offspring, as well as the possible mechanisms linking gut microbiota and disease risk. A case-control study was conducted in mothers of infants with CHD (n = 101) and mothers of infants without CHD (n = 95). By applying 16S rRNA gene sequencing and metabolic approaches to 196 stool and plasma samples, we determined microbiome and metabolome profiles in mothers of infants with CHD and controls, and their association with risk of CHD in offspring. The gut microbiome of mothers of infants with CHD was characterized with lower alpha-diversity and distinct overall microbial composition compared with mothers of infants without CHD. A distinct different metabolic profile was found between mothers of infants with CHD and controls. After controlling for the possible confounders, thirty-four bacterial genera and fifty-three plasma metabolites showed distinct abundances between the two groups. The results of the Spearman correlation analyses revealed a great number of significant correlations between the abundant bacterial genera and differentially expressed metabolites. In particular, the genus Bifidobacterium and Streptococcus showed comparable moderate positive correlations with a range of metabolites that involved in lipid metabolism pathway. Our findings suggest that perturbations of maternal gut microbiota and plasma metabolites may be associated with risk of CHD in offspring, and co-variation between microbiota and metabolites may play a part in the linkage between gut microbiota and risk of CHD in offspring.
Collapse
Affiliation(s)
- Tingting Wang
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Lizhang Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, Hunan, China
| | - Peng Huang
- Department of Thoracic Cardiac Surgery, Hunan Children's Hospital, Changsha, Hunan, China
| | - Tubao Yang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Senmao Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Lijuan Zhao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Letao Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Ziwei Ye
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Liu Luo
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China.
| |
Collapse
|
59
|
Zhou H, Ma C, Wang C, Gong L, Zhang Y, Li Y. Research progress in use of traditional Chinese medicine monomer for treatment of non-alcoholic fatty liver disease. Eur J Pharmacol 2021; 898:173976. [PMID: 33639194 DOI: 10.1016/j.ejphar.2021.173976] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
With the improvement of people's living standards and the change of eating habits, non-alcoholic fatty liver disease (NAFLD) has gradually become one of the most common chronic liver diseases in the world. However, there are no effective drugs for the treatment of NAFLD. Therefore, it is urgent to find safe, efficient, and economical anti-NAFLD drugs. Compared with western medicines that possess fast lipid-lowering effect, traditional Chinese medicines (TCM) have attracted increasing attention for the treatment of NAFLD due to their unique advantages such as multi-targets and multi-channel mechanisms of action. TCM monomers have been proved to treat NAFLD through regulating various pathways, including inflammation, lipid production, insulin sensitivity, mitochondrial dysfunction, autophagy, and intestinal microbiota. In particular, peroxisome proliferator-activated receptor α (PPAR-α), sterol regulatory element-binding protein 1c (SREBP-1c), nuclear transcription factor kappa (NF-κB), phosphoinositide 3-kinase (PI3K), sirtuin1 (SIRT1), AMP-activated protein kinase (AMPK), p53 and nuclear factor erythroid 2-related factor 2 (Nrf2) are considered as important molecular targets for ameliorating NAFLD by TCM monomers. Therefore, by searching PubMed, Web of Science and SciFinder databases, this paper updates and summarizes the experimental and clinical evidence of TCM monomers for the treatment of NAFLD in the past six years (2015-2020), thus providing thoughts and prospects for further exploring the pathogenesis of NAFLD and TCM monomer therapies.
Collapse
Affiliation(s)
- Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
60
|
Li A, Wang N, Li N, Li B, Yan F, Song Y, Hou J, Huo G. Modulation effect of chenpi extract on gut microbiota in high-fat diet-induced obese C57BL/6 mice. J Food Biochem 2021; 45:e13541. [PMID: 33570789 DOI: 10.1111/jfbc.13541] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/06/2020] [Accepted: 10/02/2020] [Indexed: 01/11/2023]
Abstract
The current study employed high-fat diet (HFD) induced murine model to assess the relationship between the lipid-lowering effect of aged citrus peel (chenpi) extract and the alterations of gut microbiota. The results showed that intake of chenpi extract for 12 week dose-dependently suppressed HFD-induced body weight, food intake, Lee's index, together with decreased the level of fasting blood glucose, total cholesterol, triglyceride, and low-density lipoprotein cholesterol. Moreover, chenpi extract administration up-regulated the abundance and diversity of fecal microbiota and down-regulated the ratio of Firmicutes-to-Bacteroidetes, which was characterized by the lower family of Lachnospiraceae, Helicobacteraceae, and Desulfovibrionaceae, and higher family of Bacteroidales_S24-7, Bacteroidaceae, Rikenellaceae, and Ruminococcaceae. Consistently, at the genus levels, chenpi extract treatment reversed the expansions of Helicobacter, Lachnospiraceae_UCG-006, and Desulfovibrio, while increased the abundance of Bacteroides, Rikenellaceae_RC9_gut_group, and Alistipes (belonging to Rikenellaceae family), Anaerotruncus and Odoribacter (belonging to Ruminococcaceae family), which were significantly negatively correlated with the levels of the serum lipid parameters. In conclusion, our findings indicated that anti-obesity ability of chenpi extract might be related to the improvement of gut microbiota imbalance. PRACTICAL APPLICATIONS: With the improvement of living standards, the incidence of metabolic diseases such as obesity, hypertension, and diabetes has increased significantly, and it has become a public health problem that seriously affects the health of the people. Chenpi contains a large amount of active ingredients, flavonoids, and other compounds, which can promote the absorption of the digestive system and have good effects on diseases such as the cardiovascular system. Our previous study has confirmed that the chenpi extract effectively regulated the glucose and lipid metabolism disorder induced by high-fat diet. However, it is not clear whether the effect is closely related to the improvement of gut microbiota. Accordingly, our result would provide a theoretical basis for future research on the relationship between obesity, chenpi extract, and gut microbiota, and support additional understanding of its potential anti-obesity effects.
Collapse
Affiliation(s)
- Aili Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Nana Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Na Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Fenfen Yan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Yue Song
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Juncai Hou
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, China
| |
Collapse
|
61
|
Gut Microbiota Functional Dysbiosis Relates to Individual Diet in Subclinical Carotid Atherosclerosis. Nutrients 2021; 13:nu13020304. [PMID: 33494335 PMCID: PMC7911134 DOI: 10.3390/nu13020304] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Gut Microbiota (GM) dysbiosis associates with Atherosclerotic Cardiovascular Diseases (ACVD), but whether this also holds true in subjects without clinically manifest ACVD represents a challenge of personalized prevention. We connected exposure to diet (self-reported by food diaries) and markers of Subclinical Carotid Atherosclerosis (SCA) with individual taxonomic and functional GM profiles (from fecal metagenomic DNA) of 345 subjects without previous clinically manifest ACVD. Subjects without SCA reported consuming higher amounts of cereals, starchy vegetables, milky products, yoghurts and bakery products versus those with SCA (who reported to consume more mechanically separated meats). The variety of dietary sources significantly overlapped with the separations in GM composition between subjects without SCA and those with SCA (RV coefficient between nutrients quantities and microbial relative abundances at genus level = 0.65, p-value = 0.047). Additionally, specific bacterial species (Faecalibacterium prausnitzii in the absence of SCA and Escherichia coli in the presence of SCA) are directly related to over-representation of metagenomic pathways linked to different dietary sources (sulfur oxidation and starch degradation in absence of SCA, and metabolism of amino acids, syntheses of palmitate, choline, carnitines and Trimethylamine n-oxide in presence of SCA). These findings might contribute to hypothesize future strategies of personalized dietary intervention for primary CVD prevention setting.
Collapse
|
62
|
Wei L, Yue F, Xing L, Wu S, Shi Y, Li J, Xiang X, Lam SM, Shui G, Russell R, Zhang D. Constant Light Exposure Alters Gut Microbiota and Promotes the Progression of Steatohepatitis in High Fat Diet Rats. Front Microbiol 2020; 11:1975. [PMID: 32973715 PMCID: PMC7472380 DOI: 10.3389/fmicb.2020.01975] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Background Non-alcoholic fatty liver disease (NAFLD) poses a significant health concern worldwide. With the progression of urbanization, light pollution may be a previously unrecognized risk factor for NAFLD/NASH development. However, the role of light pollution on NAFLD is insufficiently understood, and the underlying mechanism remains unclear. Interestingly, recent studies indicate the gut microbiota affects NAFLD/NASH development. Therefore, the present study explored effects of constant light exposure on NAFLD and its related microbiotic mechanisms. Materials and Methods Twenty-eight SD male rats were divided into four groups (n = 7 each): rats fed a normal chow diet, and exposed to standard light-dark cycle (ND-LD); rats fed a normal chow diet, and exposed to constant light (ND-LL); rats fed a high fat diet, and exposed to standard light-dark cycle (HFD-LD); and rats on a high fat diet, and exposed to constant light (HFD-LL). Body weight, hepatic pathophysiology, gut microbiota, and short/medium chain fatty acids in colon contents, serum lipopolysaccharide (LPS), and liver LPS-binding protein (LBP) mRNA expression were documented post intervention and compared among groups. Result In normal chow fed groups, rats exposed to constant light displayed glucose abnormalities and dyslipidemia. In HFD-fed rats, constant light exposure exacerbated glucose abnormalities, insulin resistance, inflammation, and liver steatohepatitis. Constant light exposure altered composition of gut microbiota in both normal chow and HFD fed rats. Compared with HFD-LD group, HFD-LL rats displayed less Butyricicoccus, Clostridium, and Turicibacter, butyrate levels in colon contents, decreased colon expression of occludin-1 and zonula occluden−1 (ZO-1), and increased serum LPS and liver LBP mRNA expression. Conclusion Constant light exposure impacts gut microbiota and its metabolic products, impairs gut barrier function and gut-liver axis, promotes NAFLD/NASH progression in HFD rats.
Collapse
Affiliation(s)
- Lin Wei
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Fangzhi Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Xing
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Shanyu Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Jinchen Li
- Department of Geriatrics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xingwei Xiang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Ryan Russell
- Cardiomatabolic Exercise Lab Director, Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
63
|
Transcriptional Regulation in Non-Alcoholic Fatty Liver Disease. Metabolites 2020; 10:metabo10070283. [PMID: 32660130 PMCID: PMC7408131 DOI: 10.3390/metabo10070283] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/01/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity is the primary risk factor for the pathogenesis of non-alcoholic fatty liver disease (NAFLD), the worldwide prevalence of which continues to increase dramatically. The liver plays a pivotal role in the maintenance of whole-body lipid and glucose homeostasis. This is mainly mediated by the transcriptional activation of hepatic pathways that promote glucose and lipid production or utilization in response to the nutritional state of the body. However, in the setting of chronic excessive nutrition, the dysregulation of hepatic transcriptional machinery promotes lipid accumulation, inflammation, metabolic stress, and fibrosis, which culminate in NAFLD. In this review, we provide our current understanding of the transcription factors that have been linked to the pathogenesis and progression of NAFLD. Using publicly available transcriptomic data, we outline the altered activity of transcription factors among humans with NAFLD. By expanding this analysis to common experimental mouse models of NAFLD, we outline the relevance of mouse models to the human pathophysiology at the transcriptional level.
Collapse
|
64
|
Song K, Kwon H, Han C, Chen W, Zhang J, Ma W, Dash S, Gandhi CR, Wu T. Yes-Associated Protein in Kupffer Cells Enhances the Production of Proinflammatory Cytokines and Promotes the Development of Nonalcoholic Steatohepatitis. Hepatology 2020; 72:72-87. [PMID: 31610032 PMCID: PMC7153981 DOI: 10.1002/hep.30990] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 10/07/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Yes-associated protein (YAP) plays an important role in hepatocarcinogenesis, although the potential role of YAP in non-neoplastic liver diseases remains largely unknown. We report herein that YAP in Kupffer cells (KCs) enhances the production of proinflammatory cytokines and promotes the development of nonalcoholic steatohepatitis (NASH). Our data show that the expression of YAP is significantly increased in KCs of wild-type mice fed a high-fat diet (HFD). APPROACH AND RESULTS We generated mice with macrophage/monocyte-specific deletion of YAP (YAPϕKO ) or Toll-like receptor 4 (TLR4; TLR4ϕKO ), and animals were fed an HFD or treated with lipopolysaccharide (LPS). Our data showed that YAPϕKO mice fed an HFD exhibited lower serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) levels and less hepatic inflammation when compared to their littermate controls. LPS treatment induced accumulation of YAP in KCs in vitro and in mice, which was prevented by macrophage/monocyte-specific deletion of TLR4 (TLR4ϕKO ). LPS transcriptionally activates YAP through activator protein 1 in macrophages/KCs. LPS-induced YAP further enhances expression of proinflammatory cytokines (including monocyte chemoattractant protein 1, tumor necrosis factor alpha, and interleukin 6) through YAP association with the TEA domain-binding motif in the promoter region of inflammatory cytokines. Forced overexpression of active YAP (YAP5SA) in KCs enhanced the production of proinflammatory cytokines. Treatment of HFD-fed mice with verteporfin inhibited KC activation, reduced liver inflammation, and decreased serum ALT/AST levels. Analyses of liver tissues from NASH patients reveal that YAP is increased in KCs and that level of YAP in human liver tissues is positively correlated with expression of proinflammatory cytokines. CONCLUSIONS This study describes an important role of YAP in KCs for regulation of liver inflammation in NASH. Our findings suggest that inhibition of YAP may represent an effective therapeutic strategy for NASH treatment.
Collapse
Affiliation(s)
- Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Hyunjoo Kwon
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Chang Han
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Janqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Wenbo Ma
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| | - Chandrashekhar R. Gandhi
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center and Department of Surgery, University of Cincinnati, Cincinnati, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, USA
| |
Collapse
|
65
|
Sivamaruthi BS, Fern LA, Rashidah Pg Hj Ismail DSN, Chaiyasut C. The influence of probiotics on bile acids in diseases and aging. Biomed Pharmacother 2020; 128:110310. [PMID: 32504921 DOI: 10.1016/j.biopha.2020.110310] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023] Open
Abstract
Recent evidence indicates the use of probiotics in the prevention and treatment of diseases. Probiotics are capable of changing the gut microbiota composition and bile acid synthesis to elicit health benefits such as cholesterol-lowering, weight reduction, and improving insulin sensitivity. The aging population is prone to develop diseases because of their decreased physiological and biological systems. Probiotics are one of the promising supplements that may potentially counteract these detrimental effects. This review will discuss the influence of probiotics on bile acids in different populations-the elderly, obese individuals, and those with hypercholesterolemia, type 2 diabetes, hypertension, and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Lim Ai Fern
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link BE1410, Brunei
| | | | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
66
|
Hori S, Abe T, Lee DG, Fukiya S, Yokota A, Aso N, Shirouchi B, Sato M, Ishizuka S. Association between 12α-hydroxylated bile acids and hepatic steatosis in rats fed a high-fat diet. J Nutr Biochem 2020; 83:108412. [PMID: 32534424 DOI: 10.1016/j.jnutbio.2020.108412] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/04/2020] [Accepted: 05/04/2020] [Indexed: 12/24/2022]
Abstract
High-fat (HF) diet induces hepatic steatosis that is a risk factor for noncommunicable diseases such as obesity, type 2 diabetes and cardiovascular disease. Previously, we found that HF feeding in rats increases the excretion of fecal bile acids (BAs), specifically 12α-hydroxylated (12αOH) BAs. Although the liver is the metabolic center in our body, the association between hepatic steatosis and 12αOH BAs in HF-fed rats is unclear. Thus, we investigated extensively BA composition in HF-fed rats and evaluated the association between hepatic steatosis and 12αOH BAs. Acclimated male inbred WKAH/HkmSlc rats were divided into two groups and fed either control or HF diet for 8 weeks. Feeding HF diet increased hepatic triglyceride and total cholesterol concentrations, which correlated positively with 12αOH BAs concentrations but not with non-12αOH BAs in the feces, portal plasma and liver. Accompanied by the increase in 12αOH BAs, the rats fed HF diet showed increased fat absorption and higher mRNA expression of liver Cidea. The enhancement of 12αOH BA secretion may contribute to hepatic steatosis by the promotion of dietary fat absorption and hepatic Cidea mRNA expression. The increase in 12αOH BAs was associated with enhanced liver cholesterol 7α-hydroxylase (Cyp7a1) and sterol 12α-hydroxylase (Cyp8b1) mRNA expression. There was a significant increase in 7α-hydroxycholesterol, a precursor of BAs, in the liver of HF-fed rats. Altogether, these data suggest that the HF diet increases preferentially 12αOH BAs synthesis by utilizing the accumulated hepatic cholesterol and enhancing mRNA expression of Cyp7a1 and Cyp8b1 in the liver.
Collapse
Affiliation(s)
- Shota Hori
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Takayuki Abe
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Dong Geun Lee
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Satoru Fukiya
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Atsushi Yokota
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Nao Aso
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Bungo Shirouchi
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Masao Sato
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Satoshi Ishizuka
- Division of Fundamental Agriscience Research, Research Faculty of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan.
| |
Collapse
|
67
|
Hepatic lipid droplet homeostasis and fatty liver disease. Semin Cell Dev Biol 2020; 108:72-81. [PMID: 32444289 DOI: 10.1016/j.semcdb.2020.04.011] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/17/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
In cells, lipids are stored in lipid droplets, dynamic organelles that adapt their size, abundance, lipid and protein composition and organelle interactions to metabolic changes. Lipid droplet accumulation in the liver is the hallmark of non-alcoholic fatty liver disease (NAFLD). Due to the prevalence of obesity, the strongest risk factor for steatosis, NAFLD and its associated complications are currently affecting more than 1 billion people worldwide. Here, we review how triglyceride and phospholipid homeostasis are regulated in hepatocytes and how imbalances between lipid storage, degradation and lipoprotein secretion lead to NAFLD. We discuss how organelle interactions are altered in NAFLD and provide insights how NAFLD progression is associated with changes in hepatocellular signaling and organ-crosstalk. Finally, we highlight unsolved questions in hepatic LD and lipoprotein biology and give an outlook on therapeutic options counteracting hepatic lipid accumulation.
Collapse
|
68
|
Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol 2020; 17:279-297. [PMID: 32152478 DOI: 10.1038/s41575-020-0269-9] [Citation(s) in RCA: 630] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/21/2020] [Indexed: 02/07/2023]
Abstract
Gut microbiota dysbiosis has been repeatedly observed in obesity and type 2 diabetes mellitus, two metabolic diseases strongly intertwined with non-alcoholic fatty liver disease (NAFLD). Animal studies have demonstrated a potential causal role of gut microbiota in NAFLD. Human studies have started to describe microbiota alterations in NAFLD and have found a few consistent microbiome signatures discriminating healthy individuals from those with NAFLD, non-alcoholic steatohepatitis or cirrhosis. However, patients with NAFLD often present with obesity and/or insulin resistance and type 2 diabetes mellitus, and these metabolic confounding factors for dysbiosis have not always been considered. Patients with different NAFLD severity stages often present with heterogeneous lesions and variable demographic characteristics (including age, sex and ethnicity), which are known to affect the gut microbiome and have been overlooked in most studies. Finally, multiple gut microbiome sequencing tools and NAFLD diagnostic methods have been used across studies that could account for discrepant microbiome signatures. This Review provides a broad insight into microbiome signatures for human NAFLD and explores issues with disentangling these signatures from underlying metabolic disorders. More advanced metagenomics and multi-omics studies using system biology approaches are needed to improve microbiome biomarkers.
Collapse
|
69
|
Aron-Wisnewsky J, Warmbrunn MV, Nieuwdorp M, Clément K. Nonalcoholic Fatty Liver Disease: Modulating Gut Microbiota to Improve Severity? Gastroenterology 2020; 158:1881-1898. [PMID: 32044317 DOI: 10.1053/j.gastro.2020.01.049] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/13/2020] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Gut microbiota plays a role in the pathophysiology of metabolic diseases, which include nonalcoholic fatty liver diseases, through the gut-liver axis. To date, clinical guidelines recommend a weight loss goal of 7%-10% to improve features of nonalcoholic fatty liver diseases. Because this target is not easily achieved by all patients, alternative therapeutic options are currently being evaluated. This review focuses on therapeutics that aim to modulate the gut microbiota and the gut-liver axis. We discuss how probiotics, prebiotics, synbiotic, fecal microbiota transfer, polyphenols, specific diets, and exercise interventions have been found to modify gut microbiota signatures; improve nonalcoholic fatty liver disease outcomes; and detail, when available, the different mechanisms by which these beneficial outcomes might occur. Apart from probiotics that have already been tested in human randomized controlled trials, most of these potential therapeutics have been studied in animals. Their efficacy still warrants confirmation in humans using appropriate design.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Sorbonne Université, INSERM, UMRS U1269, Nutriomics Research Unit, Paris, France; Nutrition Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centre de Recherche en Nutrition Humaine d'Ile de France, Paris, France; Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands.
| | - Moritz V Warmbrunn
- Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands
| | - Max Nieuwdorp
- Department of Vascular Medicine, University of Amsterdam Medical Center, Amsterdam, The Netherlands; Department of Internal Medicine, University of Amsterdam Medical Center, Free University, Amsterdam, The Netherlands
| | - Karine Clément
- Sorbonne Université, INSERM, UMRS U1269, Nutriomics Research Unit, Paris, France; Nutrition Department, Assistance Publique Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Centre de Recherche en Nutrition Humaine d'Ile de France, Paris, France.
| |
Collapse
|
70
|
Martynov V, Havryliuk V, Skliar T, Sokolova I. Comparative analysis of the composition of intestinal microbiome in patients with liver diseases. SCIENCERISE: BIOLOGICAL SCIENCE 2020. [DOI: 10.15587/2519-8025.2020.192721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
71
|
Li T, Chiang JYL. Bile acid-based therapies for non-alcoholic steatohepatitis and alcoholic liver disease. Hepatobiliary Surg Nutr 2020; 9:152-169. [PMID: 32355674 PMCID: PMC7188552 DOI: 10.21037/hbsn.2019.09.03] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022]
Abstract
Bile acids are synthesized from cholesterol only in hepatocytes. Bile acids circulating in the enterohepatic system act as physiological detergent molecules to help solubilize biliary cholesterol and emulsify dietary lipids and fat-soluble vitamins in small intestine. Bile acids are signaling molecules that activate nuclear receptor farnesoid X receptor (FXR) and cell surface G protein-coupled receptor TGR5. FXR critically regulates bile acid homeostasis by mediating bile acid feedback inhibition of hepatic bile acid synthesis. In addition, bile acid-activated cellular signaling pathways regulate metabolic homeostasis, immunity, and cell proliferation in various metabolically active organs. In the small and large intestine, gut bacterial enzymes modify primary bile acids to generate secondary bile acids to help shape the bile acid pool composition and subsequent biological effects. In turn, bile acids exhibit anti-microbial properties and modulate gut microbiota to influence host metabolism and immunity. Currently, bile acid-based therapies including systemic and intestine-restricted FXR agonists, TGR5 agonists, fibroblast growth factor 19 analogue, intestine FXR antagonists, and intestine apical sodium-bile acid transporter (ASBT) inhibitors have been developed as promising treatments for non-alcoholic steatohepatitis (NASH). These pharmacological agents improved metabolic and inflammatory disorders via distinct mechanisms of action that are subjects of extensive research interest. More recently, human and experimental alcoholic liver disease (ALD) has been associated with disrupted bile acid homeostasis. In additional, new findings showed that targeting bile acid metabolism and signaling may be promising therapeutic approaches for treating ALD.
Collapse
Affiliation(s)
- Tiangang Li
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, USA
| | - John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
72
|
Chong CYL, Orr D, Plank LD, Vatanen T, O’Sullivan JM, Murphy R. Randomised Double-Blind Placebo-Controlled Trial of Inulin with Metronidazole in Non-Alcoholic Fatty Liver Disease (NAFLD). Nutrients 2020; 12:nu12040937. [PMID: 32230987 PMCID: PMC7230525 DOI: 10.3390/nu12040937] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
Background: Non-alcoholic fatty liver disease (NAFLD) can be ameliorated by weight loss although difficult to maintain. Emerging evidence indicates that prebiotics and antibiotics improve NAFLD. Aim: To determine whether inulin supplementation after brief metronidazole therapy is effective in reducing alanine aminotransferase (ALT) and maintaining weight loss achieved through a very-low-calorie diet (VLCD) among people with NAFLD. Methods: Sixty-two people with NAFLD commenced 4-week VLCD using Optifast meal replacements (600 kcal/day). Sixty were then randomised into a 12-week double-blind, placebo-controlled, parallel three-arm trial: (1) 400 mg metronidazole twice daily in Week 1 then inulin 4 g twice daily OR (2) placebo twice daily in week one then inulin OR (3) placebo-placebo. Main outcomes were ALT and body weight at 12 weeks. Fecal microbiota changes were also evaluated. Results: Mean body mass index (BMI) and ALT reduced after VLCD by 2.4 kg/m2 and 11 U/L, respectively. ALT further decreased after metronidazole-inulin compared to after placebo-placebo (mean ALT change -19.6 vs. -0.2 U/L, respectively; p = 0.026); however, weight loss maintenance did not differ. VLCD treatment decreased the ratio of Firmicutes/Bacteroidetes (p = 0.002). Conclusion: Brief metronidazole followed by inulin supplementation can reduce ALT beyond that achieved after VLCD in patients with NAFLD.
Collapse
Affiliation(s)
- Clara Yieh Lin Chong
- Liggins Institute, The University of Auckland, Auckland 1142, New Zealand; (C.Y.L.C.); (T.V.); (J.M.O.)
| | - David Orr
- New Zealand Liver Transplant Unit, Auckland City Hospital, Auckland 1023, New Zealand
- Correspondence: (D.O.); (R.M.); Tel.: +64-9-923-6313
| | - Lindsay D. Plank
- Department of Surgery, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand;
| | - Tommi Vatanen
- Liggins Institute, The University of Auckland, Auckland 1142, New Zealand; (C.Y.L.C.); (T.V.); (J.M.O.)
| | - Justin M. O’Sullivan
- Liggins Institute, The University of Auckland, Auckland 1142, New Zealand; (C.Y.L.C.); (T.V.); (J.M.O.)
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, The University of Auckland, Auckland 1142, New Zealand
- Correspondence: (D.O.); (R.M.); Tel.: +64-9-923-6313
| |
Collapse
|
73
|
Akkermansia muciniphila Prevents Fatty Liver Disease, Decreases Serum Triglycerides, and Maintains Gut Homeostasis. Appl Environ Microbiol 2020; 86:AEM.03004-19. [PMID: 31953338 DOI: 10.1128/aem.03004-19] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 01/12/2020] [Indexed: 12/19/2022] Open
Abstract
The objective of this study was to elucidate the effect of intestinal Akkermansia muciniphila bacteria on fatty liver disease. Five-week-old C57BL/6N mice were administered either phosphate-buffered saline (PBS; control) or A. muciniphila at 108 to 109 CFU/ml, and were fed either a 45% fat diet (high-fat diet [HFD]) or a 10% fat diet (normal diet [ND]) for 10 weeks. After 10 weeks, the mice were euthanized, and blood and tissue samples, including adipose tissue, cecum, liver, and brain, were immediately collected. Biochemical and histological analyses were conducted, and the expression levels of related factors were compared to determine the antiobesity effects of Akkermansia muciniphila The gut microbiome was analyzed in fecal samples. Oral administration of A. muciniphila significantly (P < 0.05) lowered serum triglyceride (TG) and alanine aminotransferase (ALT) levels in obese mice. Compared to the non-A. muciniphila-treated group, the expression of SREBP (regulator of TG synthesis in liver tissue) was decreased in the A. muciniphila-treated group. The expression of IL-6 in the liver of obese mice was decreased following the administration of A. muciniphila Furthermore, alterations in the ratio of Firmicutes to Bacteroidetes and the decrease in bacterial diversity caused by the HFD were restored upon the administration of A. muciniphila These results indicate that A. muciniphila prevents fatty liver disease in obese mice by regulating TG synthesis in the liver and maintaining gut homeostasis.IMPORTANCE This study investigated the effect of Akkermansia muciniphila on fatty liver disease. Although some research about the effects of A. muciniphila on host health has been published, study of the relationship between A. muciniphila administration and fatty liver, as well as changes in the gut microbiota, has not been conducted. In this study, we demonstrated that A. muciniphila prevented fatty liver disease by regulation of the expression of genes that regulate fat synthesis and inflammation in the liver.
Collapse
|
74
|
Xu Y, Wang N, Tan HY, Li S, Zhang C, Feng Y. Function of Akkermansia muciniphila in Obesity: Interactions With Lipid Metabolism, Immune Response and Gut Systems. Front Microbiol 2020; 11:219. [PMID: 32153527 PMCID: PMC7046546 DOI: 10.3389/fmicb.2020.00219] [Citation(s) in RCA: 266] [Impact Index Per Article: 53.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/30/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity and its metabolic syndrome, including liver disorders and type 2 diabetes, are a worldwide epidemic and are intimately linked to diet. The gut microbiota interaction has been pointed to as a hot topic of research in the treatment of obesity and related metabolic diseases by influencing energy metabolism and the immune system. In terms of the novel beneficial microbes identified, Akkermansia muciniphila (A. muciniphila) colonizes the mucosa layer of the gut and modulates basal metabolism. A. muciniphila is consistently correlated with obesity. The causal beneficial impact of A. muciniphila treatment on obesity is coming to light, having been proved by a variety of animal models and human studies. A. muciniphila has been characterized as a beneficial player in body metabolism and has great prospects for treatments of the metabolic disorders associated with obesity, as well as being considered for next-generation therapeutic agents. This paper aimed to investigate the basic mechanism underlying the relation of A. muciniphila to obesity and its host interactions, as identified in recent discoveries, facilitating the establishment of the causal relationship in A. muciniphila-associated therapeutic supplement in humans.
Collapse
Affiliation(s)
- Yu Xu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Hor-Yue Tan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
75
|
Du H, Zhao A, Wang Q, Yang X, Ren D. Supplementation of Inulin with Various Degree of Polymerization Ameliorates Liver Injury and Gut Microbiota Dysbiosis in High Fat-Fed Obese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:779-787. [PMID: 31894986 DOI: 10.1021/acs.jafc.9b06571] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The chain length of fructan determines its different physiological effects. This study is to explore the effects of low-performance inulin [LPI, degree of polymerization (DP) ≤ 9] and high-performance inulin (HPI, DP ≥ 23) on obesity-associated liver injury of high-fat diet (HFD) feeding mice and its underlying mechanism. Eight weeks of supplementation of C57BL/6J mice with HPI, relative to LPI (p < 0.05), caused the more efficient improvement against the HFD-induced liver insulin resistance through activating IRS1/PI3K/Akt pathway and reduced protein expressions of inflammatory factors nuclear factor-kappaB (NF-κB) and interleukin-6 (IL-6) in the liver. HPI exhibited the more positive effects on liver steatosis by inhibiting acetyl-CoA carboxylase (ACC), fatty acid synthase (FAS), and sterol regulatory element binding protein 1 (SREBP1) in comparison with LPI (p < 0.05). HPI also increased acetic acid, propionic acid, and butyric acid levels in the colon of HFD-fed mice (p < 0.05). Compared to LPI, HPI feeding of HFD-fed mice led to the more effective decrease in the Firmicutes abundance from 72.1% to 34.5%, but a more significant increase in the Bacteroidetes population from 19.8 to 57.1% at the phyla level, and increased the abundance of Barnesiella, Bacteroides, and Parabacteroides at the genus level (p < 0.05). Depending on DP, HPI exerts the more positive regulation on liver injury and gut microbiota dysfunction than LPI.
Collapse
Affiliation(s)
- Haiping Du
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science , Shaanxi Normal University , Xi'an 710119 , China
| | - Aiqing Zhao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science , Shaanxi Normal University , Xi'an 710119 , China
| | - Qi Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science , Shaanxi Normal University , Xi'an 710119 , China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science , Shaanxi Normal University , Xi'an 710119 , China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science , Shaanxi Normal University , Xi'an 710119 , China
| |
Collapse
|
76
|
Quesada-Vázquez S, Aragonès G, Del Bas JM, Escoté X. Diet, Gut Microbiota and Non-Alcoholic Fatty Liver Disease: Three Parts of the Same Axis. Cells 2020; 9:E176. [PMID: 31936799 PMCID: PMC7016763 DOI: 10.3390/cells9010176] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/04/2020] [Accepted: 01/07/2020] [Indexed: 01/30/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most common liver disease in the world. NAFLD is principally characterized by an excessive fat accumulation in the hepatocytes. Diet is considered as one of the main drivers to modulate the composition of gut microbiota, which participate in different processes, affecting human metabolism. A disruption in the homeostasis of gut microbiota may lead to dysbiosis, which is commonly reflected by a reduction of the beneficial species and an increment in pathogenic microbiota. Gut and liver are in close relation due to the anatomical and functional interactions led by the portal vein, thus altered intestinal microbiota might affect liver functions, promoting inflammation, insulin resistance and steatosis, which is translated into NAFLD. This review will highlight the association between diet, gut microbiota and liver, and how this axis may promote the development of NAFLD progression, discussing potential mechanisms and alterations due to the dysbiosis of gut microbiota. Finally, it will revise the variations in gut microbiota composition in NAFLD, and it will focus in specific species, which directly affect NAFLD progression.
Collapse
Affiliation(s)
- Sergio Quesada-Vázquez
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (J.M.D.B.)
| | - Gerard Aragonès
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Nutrigenomics Research Group, 43007 Tarragona, Spain;
| | - Josep M Del Bas
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (J.M.D.B.)
| | - Xavier Escoté
- Unitat de Nutrició i Salut, Centre Tecnològic de Catalunya, Eurecat, 43204 Reus, Spain; (S.Q.-V.); (J.M.D.B.)
| |
Collapse
|
77
|
Yong HY, Larrouy-Maumus G, Zloh M, Smyth R, Ataya R, Benton CM, Munday MR. Early detection of metabolic changes in drug-induced steatosis using metabolomics approaches. RSC Adv 2020; 10:41047-41057. [PMID: 35519189 PMCID: PMC9057704 DOI: 10.1039/d0ra06577c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/23/2020] [Indexed: 12/26/2022] Open
Abstract
Steatosis is the accumulation of triglycerides in hepatic cells wherein fats exceed 5% of the entire liver weight. Although steatotic liver damage is reversible due to the liver's regenerative capability, protracted damage often and typically leads to irreversible conditions such as cirrhosis and hepatocellular carcinoma (HCC). Therefore, early steatotic detection is critical for preventing progression to advanced liver diseases. This also becomes particularly important given the higher prevalence of drug usage, as drugs are a frequent cause of liver damage. Currently, the recommendation to diagnose steatosis is using liver enzymes and performing a liver biopsy. Liver biopsy remains the gold standard method of detection, but the procedure is invasive and an unreliable diagnostic tool. Non-invasive, specific and sensitive diagnostic solutions such as biomarkers are therefore needed for the early detection of steatosis. Our aim is to identify changes in urinary metabolites in tetracycline-induced hepatic steatotic rats at different stages of the diseases using metabolomic-based techniques. Sprague Dawley male rats are treated by intraperitoneal injection (I.P.) with either 62.5 mg kg−1 or 125 mg kg−1 tetracycline, an antibiotic previously known to induce steatosis. We analyse the metabolic profile of the urinary tetracycline induced hepatic steatotic rats using 1H nuclear magnetic resonance (NMR), 2D 1H–1H TOCSY (total correlation spectroscopy) and electrospray liquid chromatography-mass spectrometry (ESI-LC-MS/MS) based metabolomics. The combined analysis of haematoxylin & eosin (H&E), oil red O (ORO) and direct measurement of triglyceride content in the liver tissues of the control samples against 125 mg kg−1 and 62.5 mg kg−1 treated samples, reveals that 125 mg kg−1 tetracycline exposure potentially induces steatosis. The combination of 1H NMR, 2D 1H–1H TOCSY and ESI-LC-MS/MS alongside multivariate statistical analysis, detected a total of 6 urinary metabolites changes, across 6 metabolic pathways. Furthermore, lysine concentration correlates with liver damage as tetracycline dose concentration increases, whilst both H&E and ORO fail to detect hepatocellular damage at the lowest dose concentration. We conclude that the combination of 1H NMR and ESI-LC-MS/MS suggests that these are suitable platforms for studying the pathogenesis of steatosis development, prior to morphological alterations observed in staining techniques and offer a more detailed description of the severity of the steatotic disease. Urinary metabolic profiling of tetracycline induced hepatic steatotic rats were investigated using 1H nuclear magnetic resonance, 2D 1H–1H total correlation spectroscopy and electrospray liquid chromatography-mass spectrometry based metabolomics.![]()
Collapse
Affiliation(s)
- Helena Y Yong
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Gerald Larrouy-Maumus
- MRC Centre for Molecular Bacteriology and Infection
- Department of Life Science
- Faculty of Natural Sciences
- Imperial College London
- UK
| | - Mire Zloh
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Rosemary Smyth
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | - Rayan Ataya
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| | | | - Michael R. Munday
- Department of Pharmaceutical and Biological Chemistry
- University of London
- UK
| |
Collapse
|
78
|
Fukui H. Role of Gut Dysbiosis in Liver Diseases: What Have We Learned So Far? Diseases 2019; 7:diseases7040058. [PMID: 31726747 PMCID: PMC6956030 DOI: 10.3390/diseases7040058] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence supports that gut dysbiosis may relate to various liver diseases. Alcoholics with high intestinal permeability had a decrease in the abundance of Ruminnococcus. Intestinal dysmotility, increased gastric pH, and altered immune responses in addition to environmental and genetic factors are likely to cause alcohol-associated gut microbial changes. Alcohol-induced dysbiosis may be associated with gut barrier dysfunction, as microbiota and their products modulate barrier function by affecting epithelial pro-inflammatory responses and mucosal repair functions. High levels of plasma endotoxin are detected in alcoholics, in moderate fatty liver to advanced cirrhosis. Decreased abundance of Faecalibacterium prausnitzii, an anti-inflammatory commensal, stimulating IL-10 secretion and inhibiting IL-12 and interferon-γ expression. Proteobacteria, Enterobacteriaceae, and Escherichia were reported to be increased in NAFLD (nonalcoholic fatty liver disease) patients. Increased abundance of fecal Escherichia to elevated blood alcohol levels in these patients and gut microbiota enriched in alcohol-producing bacteria produce more alcohol (alcohol hypothesis). Some undetermined pathological sequences related to gut dysbiosis may facilitate energy-producing and proinflammatory conditions for the progression of NAFLD. A shortage of autochthonous non-pathogenic bacteria and an overgrowth of potentially pathogenic bacteria are common findings in cirrhotic patients. The ratio of the amounts of beneficial autochthonous taxa (Lachnospiraceae + Ruminococaceae + Veillonellaceae + Clostridiales Incertae Sedis XIV) to those of potentially pathogenic taxa (Enterobacteriaceae + Bacteroidaceae) was low in those with early death and organ failure. Cirrhotic patients with decreased microbial diversity before liver transplantation were more likely to develop post-transplant infections and cognitive impairment related to residual dysbiosis. Patients with PSC had marked reduction of bacterial diversity. Enterococcus and Lactobacillus were increased in PSC patients (without liver cirrhosis.) Treatment-naive PBC patients were associated with altered composition and function of gut microbiota, as well as a lower level of diversity. As serum anti-gp210 antibody has been considered as an index of disease progression, relatively lower species richness and lower abundance of Faecalibacterium spp. in gp210-positive patients are interesting. The dysbiosis-induced altered bacterial metabolites such as a hepatocarcinogenesis promotor DCA, together with a leaky gut and bacterial translocation. Gut protective Akkermansia and butyrate-producing genera were decreased, while genera producing-lipopolysaccharide were increased in early hepatocellular carcinoma (HCC) patients.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Nara Medical University, Kashihara 634-8522, Japan
| |
Collapse
|
79
|
Wang T, Wei X, Chen T, Wang W, Xia X, Miao J, Yin S. Studies of the mechanism of fatty liver formation in Takifugu fasciatus following copper exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 181:353-361. [PMID: 31207574 DOI: 10.1016/j.ecoenv.2019.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/03/2019] [Accepted: 06/05/2019] [Indexed: 06/09/2023]
Abstract
Fatty liver is widely observed during Takifugu fasciatus production, but the mechanisms underlying fatty liver formation remain unknown. The present study was conducted to determine the potential effects of copper (Cu) on hepatic lipid deposition and metabolism in T. fasciatus after 21 days of exposure to Cu (levels: 0, 20 and 100 μg/L). Copper exposure decreased the weight gain rate (WG) in T. fasciatus, but increased the values of the viscerosomatic index (VSI) and hepatosomatic index (HSI) compared with the control. The time-dependent Cu accumulation in tissues increased as the Cu concentration increased. The order of Cu accumulation was liver > intestine > muscle. The lipid content, triglyceride (TG) content and lipoprotein lipase (LPL) activity increased after Cu exposure compared with the control. In addition, more lipid droplets and greater vacuolization were observed in the liver after exposure to 20 μg/L Cu than after 100 μg/L Cu. The expression of genes involved in lipogenesis (g6pd, 6pgd, lpl, fas and acc), lipolysis (hsl and cpt 1) and transcription (ppar α and ppar ©) was dependent on Cu. An analysis of the intestinal microbiome community showed that the highest values of the Chao 1 index, ACE, Shannon index and Simpson index were obtained in fish exposed to 20 μg/L Cu, whereas the lowest values were obtained after the 100 μg/L Cu treatment. The Principal Coordinates Analysis (PCoA) plots of the data revealed structural differences in the groups treated with Cu compared with the control group. At the phylum level, the intestinal microbiota in the Cu-treated and control fish were dominated by Proteobacteria and Bacteroidetes. The higher Firmicutes to Bacteroidetes ratio was observed in fish treated with 20 μg/L Cu compared with other groups, while the lowest ratio was observed in fish exposed to 100 μg/L Cu. Our study revealed the mechanisms by which Cu exposure altered (i) lipid deposition in the body and (ii) the intestinal microbiome, which may contribute to maintain the health status of T. fasciatus for the aquaculture.
Collapse
Affiliation(s)
- Tao Wang
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu, 222005, China
| | - Xiaozhen Wei
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu, 222005, China
| | - Tongqing Chen
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China
| | - Wei Wang
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China
| | - Xiaoyu Xia
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China
| | - Jinhan Miao
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China
| | - Shaowu Yin
- College of Marine Science and Engineering, College of Life Science, Nanjing Normal University, Nanjing, Jiangsu, 210023, China; Co-Innovation Center for Marine Bio-Industry Technology of Jiangsu Province, Lianyungang, Jiangsu, 222005, China.
| |
Collapse
|
80
|
An X, Bao Q, Di S, Zhao Y, Zhao S, Zhang H, Lian F, Tong X. The interaction between the gut Microbiota and herbal medicines. Biomed Pharmacother 2019; 118:109252. [DOI: 10.1016/j.biopha.2019.109252] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
|
81
|
Alcohol or Gut Microbiota: Who Is the Guilty? Int J Mol Sci 2019; 20:ijms20184568. [PMID: 31540133 PMCID: PMC6770333 DOI: 10.3390/ijms20184568] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD), a disorder caused by excessive alcohol intake represents a global health care burden. ALD encompasses a broad spectrum of hepatic injuries including asymptomatic steatosis, alcoholic steatohepatitis (ASH), fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). The susceptibility of alcoholic patients to develop ALD is highly variable and its progression to more advanced stages is strongly influenced by several hits (i.e., amount and duration of alcohol abuse). Among them, the intestinal microbiota and its metabolites have been recently identified as paramount in ALD pathophysiology. Ethanol abuse triggers qualitative and quantitative modifications in intestinal flora taxonomic composition, mucosal inflammation, and intestinal barrier derangement. Intestinal hypermeability results in the translocation of viable pathogenic bacteria, Gram-negative microbial products, and pro-inflammatory luminal metabolites into the bloodstream, further corroborating the alcohol-induced liver damage. Thus, the premise of this review is to discuss the beneficial effect of gut microbiota modulation as a novel therapeutic approach in ALD management.
Collapse
|
82
|
Liu B, Zhang J, Sun P, Yi R, Han X, Zhao X. Raw Bowl Tea (Tuocha) Polyphenol Prevention of Nonalcoholic Fatty Liver Disease by Regulating Intestinal Function in Mice. Biomolecules 2019; 9:biom9090435. [PMID: 31480575 PMCID: PMC6770140 DOI: 10.3390/biom9090435] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
A high-fat diet-induced C57BL/6N mouse model of non-alcoholic fatty liver disease (NAFLD) was established. The effect and mechanism of Raw Bowl Tea polyphenols (RBTP) on preventing NAFLD via regulating intestinal function were observed. The serum, liver, epididymis, small intestine tissues, and feces of mice were examined by biochemical and molecular biological methods, and the composition of RBTP was analyzed by HPLC assay. The results showed that RBTP could effectively reduce the body weight, liver weight, and liver index of NAFLD mice. The serum effects of RBTP were: (1) decreases in alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (AKP), total cholesterol (TC), triglyceride (TG), low density lipoprotein cholesterol (LDL-C), D-lactate (D-LA), diamine oxidase (DAO), lipopolysaccharide (LPS), and an increase of high density lipoprotein cholesterol (HDL-C) levels; (2) a decrease of inflammatory cytokines such as interleukin 1 beta (IL-1β), interleukin 4 (IL-4), interleukin 6 (IL-6), interleukin 10 (IL-10), tumor necrosis factor alpha (TNF-α), and interferon gamma (INF-γ); (3) a decrease the reactive oxygen species (ROS) level in liver tissue; and (4) alleviation of pathological injuries of liver, epididymis, and small intestinal tissues caused by NAFLD and protection of body tissues. qPCR and Western blot results showed that RBTP could up-regulate the mRNA and protein expressions of LPL, PPAR-α, CYP7A1, and CPT1, and down-regulate PPAR-γ and C/EBP-α in the liver of NAFLD mice. In addition, RBTP up-regulated the expression of occludin and ZO-1, and down-regulated the expression of CD36 and TNF-α in the small intestines of NAFLD mice. Studies on mice feces showed that RBTP reduced the level of Firmicutes and increased the minimum levels of Bacteroides and Akkermansia, as well as reduced the proportion of Firmicutes/Bacteroides in the feces of NAFLD mice, which play a role in regulating intestinal microecology. Component analysis showed that RBTP contained seven polyphenolic compounds: Gallic acid, (-)-epigallocatechin, catechin, L-epicatechin, (-)-epigallocatechin gallate, (-)-gallocatechin gallate, and (-)-epicatechin gallate (ECG), and high levels of caffeine, (-)-epigallocatechin (EGC), and ECG. RBTP improved the intestinal environment of NAFLD mice with the contained active ingredients, thus playing a role in preventing NAFLD. The effect was positively correlated with the dose of 100 mg/kg, which was even better than that of the clinical drug bezafibrate.
Collapse
Affiliation(s)
- Bihui Liu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China
- College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| | - Jing Zhang
- Environment and Quality Inspection College, Chongqing Chemical Industry Vocational College, Chongqing 401228, China
| | - Peng Sun
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China
- College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| | - Ruokun Yi
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China
- Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China
| | - Xiaoyan Han
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China
- College of Biological and Chemical Engineering, Chongqing University of Education, Chongqing 400067, China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China.
- Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China.
| |
Collapse
|
83
|
Schwenger KJ, Clermont-Dejean N, Allard JP. The role of the gut microbiome in chronic liver disease: the clinical evidence revised. JHEP Rep 2019; 1:214-226. [PMID: 32039372 PMCID: PMC7001555 DOI: 10.1016/j.jhepr.2019.04.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/08/2019] [Accepted: 04/27/2019] [Indexed: 02/07/2023] Open
Abstract
Recent research has suggested a role for the intestinal microbiota in the pathogenesis and potential treatment of a wide range of liver diseases. The intestinal microbiota and bacterial products may contribute to the development of liver diseases through multiple mechanisms including increased intestinal permeability, chronic systemic inflammation, production of short-chain fatty acids and changes in metabolism. This suggests a potential role for pre-, pro- and synbiotic products in the prevention or treatment of some liver diseases. In addition, there is emerging evidence on the effects of faecal microbial transplant. Herein, we discuss the relationship between the intestinal microbiota and liver diseases, as well as reviewing intestinal microbiota-based treatment options that are currently being investigated.
Collapse
Affiliation(s)
- Katherine Jp Schwenger
- Toronto General Hospital, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | | | - Johane P Allard
- Toronto General Hospital, University Health Network, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada.,Department of Nutritional Sciences, University of Toronto, Toronto, Canada
| |
Collapse
|
84
|
Kim HN, Joo EJ, Cheong HS, Kim Y, Kim HL, Shin H, Chang Y, Ryu S. Gut Microbiota and Risk of Persistent Nonalcoholic Fatty Liver Diseases. J Clin Med 2019; 8:E1089. [PMID: 31344854 PMCID: PMC6722749 DOI: 10.3390/jcm8081089] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/12/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022] Open
Abstract
Gut dysbiosis is regarded as a pathogenetic factor of nonalcoholic fatty liver disease (NAFLD), but its role in NAFLD persistence is unknown. We investigated the influence of the gut microbiota on persistent NAFLD. This cohort study included 766 subjects with 16S ribosomal RNA (rRNA) gene sequencing data from fecal samples at baseline who underwent repeated health check-up examinations. Fatty liver was determined using ultrasound at baseline and follow-up. Participants were categorized into four groups: none (control), developed, regressed, or persistent NAFLD. The persistent NAFLD group had lower richness compared with the control group. Significant differences were also found in both non-phylogenic and phylogenic beta diversity measures according to NAFLD persistence. Pairwise comparisons indicated that taxa abundance mainly differed between the control and persistent NAFLD groups. A relative high abundance of Fusobacteria and low abundance of genera Oscillospira and Ruminococcus of the family Ruminococcaceae and genus Coprococcus of the family Lachnospiraceae were found in the persistent NAFLD group. Based on the functional predictions, pathways related to primary and secondary bile acid biosynthesis were highly detected in the persistent NAFLD group compared with the control group. These findings support that the composition of the gut microbiome associated with dysregulation of bile acid biosynthetic pathways may contribute to the persistence of NAFLD. This is the first cohort study to demonstrate the influence of microbiota on persistent NAFLD. Our findings may help identify potential targets for therapeutic intervention in NAFLD.
Collapse
Affiliation(s)
- Han-Na Kim
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 03181, Korea
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul 06351, Korea
| | - Eun-Jeong Joo
- Division of Infectious Diseases, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 03181, Korea
| | - Hae Suk Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 03181, Korea
| | - Yejin Kim
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea
| | - Hyung-Lae Kim
- Department of Biochemistry, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Hocheol Shin
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea
- Department of Family Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, Korea
| | - Yoosoo Chang
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea.
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea.
| | - Seungho Ryu
- Center for Cohort Studies, Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea.
- Department of Clinical Research Design & Evaluation, SAIHST, Sungkyunkwan University, Seoul 06351, Korea.
- Department of Occupational and Environmental Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, School of Medicine, Seoul 04514, Korea.
| |
Collapse
|
85
|
Jayakumar S, Loomba R. Review article: emerging role of the gut microbiome in the progression of nonalcoholic fatty liver disease and potential therapeutic implications. Aliment Pharmacol Ther 2019; 50:144-158. [PMID: 31149745 PMCID: PMC6771496 DOI: 10.1111/apt.15314] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/24/2018] [Accepted: 04/30/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent disorder associated with obesity and diabetes. Few treatment options are effective for patients with NAFLD, but connections between the gut microbiome and NAFLD and NAFLD-associated conditions suggest that modulation of the gut microbiota could be a novel therapeutic option. AIM To examine the effect of the gut microbiota on pathophysiologic causes of NAFLD and assess the potential of microbiota-targeting therapies for NAFLD. METHODS A PubMed search of the literature was performed; relevant articles were included. RESULTS The composition of bacteria in the gastrointestinal tract can enhance fat deposition, modulate energy metabolism and alter inflammatory processes. Emerging evidence suggests a role for the gut microbiome in obesity and metabolic syndrome. NAFLD is often considered the hepatic manifestation of metabolic syndrome, and there has been tremendous progress in understanding the association of gut microbiome composition with NAFLD disease severity. We discuss the role of the gut microbiome in NAFLD pathophysiology and whether the microbiome composition can differentiate the two categories of NAFLD: nonalcoholic fatty liver (NAFL, the non-progressive form) vs nonalcoholic steatohepatitis (NASH, the progressive form). The association between gut microbiome and fibrosis progression in NAFLD is also discussed. Finally, we review whether modulation of the gut microbiome plays a role in improving treatment outcomes for patients with NAFLD. CONCLUSIONS Multiple pathophysiologic pathways connect the gut microbiome with the pathophysiology of NAFLD. Therefore, therapeutics that effectively target the gut microbiome may be beneficial for the treatment of patients with NAFLD.
Collapse
Affiliation(s)
- Saumya Jayakumar
- Division of Gastroenterology and Hepatology, Department of MedicineNAFLD Research Center, University of California at San DiegoLa JollaCalifornia
| | - Rohit Loomba
- Division of Gastroenterology and Hepatology, Department of MedicineNAFLD Research Center, University of California at San DiegoLa JollaCalifornia,Division of Epidemiology, Department of Family Medicine and Public HealthUniversity of California at San DiegoLa JollaCalifornia
| |
Collapse
|
86
|
Ni J, Huang R, Zhou H, Xu X, Li Y, Cao P, Zhong K, Ge M, Chen X, Hou B, Yu M, Peng B, Li Q, Zhang P, Gao Y. Analysis of the Relationship Between the Degree of Dysbiosis in Gut Microbiota and Prognosis at Different Stages of Primary Hepatocellular Carcinoma. Front Microbiol 2019; 10:1458. [PMID: 31293562 PMCID: PMC6603198 DOI: 10.3389/fmicb.2019.01458] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022] Open
Abstract
Gut microbiota dysbiosis is closely associated with primary hepatocellular carcinoma (HCC). Recent studies have evaluated the early diagnosis of primary HCC through analysis of gut microbiota dysbiosis. However, the relationship between the degree of dysbiosis and the prognosis of primary HCC remains unclear. Because primary HCC is accompanied by dysbiosis and dysbiosis usually increases the circulatory concentrations of endotoxin and other harmful bacterial substances, which further increases liver damage, we hypothesized that level of dysbiosis associated with primary HCC increases with the stage of cancer progression. To test this hypothesis, we introduced a more integrated index referred to as the degree of dysbiosis (Ddys ); and we investigated Ddys of the gut microbiota with the development of primary HCC through high-throughput sequencing of 16S rRNA gene amplicons. Our results showed that compared with healthy individuals, patients with primary HCC showed increased pro-inflammatory bacteria in their fecal microbiota. The Ddys increased significantly in patients with primary HCC compared with that in healthy controls. Moreover, there was a tendency for the Ddys to increase with the development of primary HCC, although no significant difference was detected between different stages of primary HCC. Our findings provide important insights into the use of gut microbiota analysis during the treatment of primary HCC.
Collapse
Affiliation(s)
- Jiajia Ni
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Rong Huang
- Department of Neonatal Surgery, Guangdong Women and Children Hospital, Guangzhou, China
| | - Huifang Zhou
- Department of Clinical Laboratory, First People's Hospital of Kashi, Kashgar, China
| | - Xiaoping Xu
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yang Li
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Peihua Cao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Kebo Zhong
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Mei Ge
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xiaoxia Chen
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Baohua Hou
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Min Yu
- Department of General Surgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Baogang Peng
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiao Li
- Department of Hepatic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peng Zhang
- Department of Organ Transplantation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center of Artificial Organ and Tissue Engineering, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
87
|
El Golli-Bennour E, Timoumi R, Annaibi E, Mokni M, Omezzine A, Bacha H, Abid-Essefi S. Protective effects of kefir against deltamethrin-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:18856-18865. [PMID: 31062243 DOI: 10.1007/s11356-019-05253-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/22/2019] [Indexed: 06/09/2023]
Abstract
Deltamethrine (DLM) is a synthetic pyrethroid with broad spectrum activities against acaricides and insects. Widely used for agricultural and veterinary purposes, its human and animal exposure occurs by ingestion of contaminated water and food and leads to serious health problems. Kefir is fermented milk with numerous health favors counting restorative properties of bacterial flora, immune system stimulation, cholesterol reduction, as well as anti-mutagenic and anti-tumor properties. The present study was undertaken to examine the hepatoprotective and antioxidant potential of kefir against DLM toxicity in male Wistar albino rats. DLM-treated animals revealed a significant increase in serum biochemical parameters as well as hepatic protein and lipid oxidations but caused an inhibition in antioxidant enzymes. Additionally, we have observed an increase in hepatocyte DNA damages. This toxic effect was confirmed by histological study. Kefir administration normalized the elevated serum levels of aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (T bilirubin), and cholesterol. It also reduced DLM-induced protein carbonyl (PC) and malondialdehyde (MDA) formations. Furthermore, Kefir treatment restored catalase (CAT) and superoxide dismutase (SOD) activities. The co-treatment as well as the pre-treatment by kefir showed an improvement of oxidative status as well as suppressed inflammation and DNA damages. However, the pre-treatment seems to be the most efficient. Therefore, it could be concluded that kefir is a natural product able to protect against the hepatotoxic effects of DLM by its free radical-scavenging and potent antioxidant activity.
Collapse
Affiliation(s)
- Emna El Golli-Bennour
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Rue Avicenne, Monastir, Tunisia
| | - Rim Timoumi
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Rue Avicenne, Monastir, Tunisia
| | - Emna Annaibi
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Rue Avicenne, Monastir, Tunisia
| | - Moncef Mokni
- Department of Anatomic Pathology and Histology, University Hospital Farhat Hached, Sousse, Tunisia
| | - Asma Omezzine
- LR12SP11, Biochemistry Department, Sahloul University Hospital, Sousse, Tunisia
| | - Hassen Bacha
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Rue Avicenne, Monastir, Tunisia
| | - Salwa Abid-Essefi
- Laboratory for Research on Biologically Compatible Compounds, Faculty of Dental Medicine, Rue Avicenne, Monastir, Tunisia.
| |
Collapse
|
88
|
Sun J, Qi C, Zhu H, Zhou Q, Xiao H, Le G, Chen D, Yu R. IgA-Targeted Lactobacillus jensenii Modulated Gut Barrier and Microbiota in High-Fat Diet-Fed Mice. Front Microbiol 2019; 10:1179. [PMID: 31178854 PMCID: PMC6542990 DOI: 10.3389/fmicb.2019.01179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/08/2019] [Indexed: 12/14/2022] Open
Abstract
IgA-coated Lactobacillus live in the mucous layer of the human or mammalian intestine in close proximity to epithelial cells. They act as potential probiotics for functional food development, but their physiological regulation has not yet been studied. We isolated IgA-targeted (Lactobacillus jensenii IgA21) and lumen lactic acid bacterial strains (Pediococcus acidilactici FS1) from the fecal microbiota of a healthy woman. C57BL/6 mice were fed a normal (CON) or high fat diet (HFD) for 6 weeks and then treated with IgA21 or FS1 for 4 weeks. HFD caused dyslipidemia, mucosal barrier damage, and intestinal microbiota abnormalities. Only IgA21 significantly inhibited dyslipidemia and gut barrier damage. This was related to significant up-regulation of mucin-2, PIgR mRNA expression, and colonic butyrate production (P < 0.05 vs. HFD). Unlike IgA21, FS1 caused a more pronounced gut dybiosis than did HFD, and, in particular, it induced a significant decrease in the Bacteroidales S24-7 group and an increase in Desulfovibrionaceae (P < 0.05 vs. CON). In conclusion, IgA-coated and non-coated lactic acid bacteria of gut have been demonstrated to differentially affect the intestinal barrier and serum lipids. This indicates that IgA-bound bacteria possess the potential to more easily interact with the host gut to regulate homeostasis.
Collapse
Affiliation(s)
- Jin Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ce Qi
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hualing Zhu
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qin Zhou
- The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA, United States
| | - Guowei Le
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Daozhen Chen
- The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Renqiang Yu
- The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
89
|
Szanto KB, Li J, Cordero P, Oben JA. Ethnic differences and heterogeneity in genetic and metabolic makeup contributing to nonalcoholic fatty liver disease. Diabetes Metab Syndr Obes 2019; 12:357-367. [PMID: 30936733 PMCID: PMC6430068 DOI: 10.2147/dmso.s182331] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obesity is the most prevalent noncommunicable disease in the 21st century, associated with triglyceride deposition in hepatocytes leading to nonalcoholic fatty liver disease (NAFLD). NAFLD is now present in around a third of the world's population. Epidemiological studies have concluded that ethnicity plays a role in complications and treatment response. However, definitive correlations of ethnicity with NAFLD are thoroughly under-reported. A comprehensive review was conducted on ethnic variation in NAFLD patients and its potential role as a crucial effector in complications and treatment response. The highest NAFLD prevalence is observed in Hispanic populations, exhibiting a worse disease progression. In contrast, African-Caribbeans exhibit the lowest risk, with less severe steatosis and inflammation, lower levels of triglycerides, and less metabolic derangement, but conversely higher prevalence of insulin resistance. The prevalence of NAFLD in Asian cohorts is under-reported, although reaching epidemic proportions in these populations. The most well-documented NAFLD patient population is that of Caucasian ethnicity, especially from the US. The relative paucity of available literature suggests there is a vital need for more large-scale multi-ethnic clinical cohort studies to determine the incidence of NAFLD within ethnic groups. This would improve therapy and drug development, as well as help identify candidate gene mutations which may differ within the population based on ethnic background.
Collapse
Affiliation(s)
- Krisztina B Szanto
- Faculty of Life Sciences and Medicine, School of Medicine, King's College London, London, UK,
- Institute for Liver and Digestive Health, University College London, London, UK,
| | - Jiawei Li
- Institute for Liver and Digestive Health, University College London, London, UK,
- Institute of Child Health, University College London, London, UK
| | - Paul Cordero
- Institute for Liver and Digestive Health, University College London, London, UK,
| | - Jude A Oben
- Institute for Liver and Digestive Health, University College London, London, UK,
- Department of Gastroenterology and Hepatology, Guy's and St Thomas' Hospital, NHS Foundation Trust, London, UK
| |
Collapse
|
90
|
Ma Y, Chen K, Lv L, Wu S, Guo Z. Ferulic acid ameliorates nonalcoholic fatty liver disease and modulates the gut microbiota composition in high-fat diet fed ApoE -/- mice. Biomed Pharmacother 2019; 113:108753. [PMID: 30856537 DOI: 10.1016/j.biopha.2019.108753] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/21/2022] Open
Abstract
The objective of this study was to investigate the effects of ferulic acid (FA) on nonalcoholic fatty liver disease (NAFLD) and gut microbiota, and its regulation mechanism in ApoE-/- mice fed on a high-fat diet (HFD). Liver morphology, blood lipids, gut microbiota and their metabolite indole-3-acetic acid (I3A) were determined in ApoE-/- mice. We also examined the hepatic expression of aryl hydrocarbon receptor (AHR), which inhibits the expression of fatty acid synthase (FASN) and sterol regulatory element-binding protein 1c (SREBP-1c), and ultimately reduces the deposition of triglycerides (TG) and total cholesterol (TC) in the liver. The results of the animal experiment showed that oral administration of FA markedly alleviated the formation of NAFLD and decreased the levels of serum TC, TG and low-density lipoprotein cholesterol (LDL-C). Furthermore, FA supplementation altered the composition of gut microbiota, in particular, modulating the ratio of Firmicutes to Bacteroidetes, and decreased the generation of I3A. Additionally, FA could increase the expression of hepatic AHR and inhibit the expression of FASN and SREBP-1c in the liver. Finally, we found that FA did not have hepatorenal toxicity. The findings above illustrate that FA has the potential to ameliorate NAFLD, some of which are closely related to the modulation of specific gut microbiota and the regulation of genes involved in TG and TC metabolism.
Collapse
Affiliation(s)
- Yunci Ma
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kai Chen
- College of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lin Lv
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, China
| | - Shaoyu Wu
- School of Pharmaceutical Science, Southern Medical University, Guangzhou, China.
| | - Zhijian Guo
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
91
|
Gut Microbiota-Derived Mediators as Potential Markers in Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8507583. [PMID: 30719448 PMCID: PMC6334327 DOI: 10.1155/2019/8507583] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common, multifactorial, and poorly understood liver disease whose incidence is globally rising. During the past decade, several lines of evidence suggest that dysbiosis of intestinal microbiome represents an important factor contributing to NAFLD occurrence and its progression into NASH. The mechanisms that associate dysbiosis with NAFLD include changes in microbiota-derived mediators, deregulation of the gut endothelial barrier, translocation of mediators of dysbiosis, and hepatic inflammation. Changes in short chain fatty acids, bile acids, bacterial components, choline, and ethanol are the result of altered intestinal microbiota. We perform a narrative review of the previously published evidence and discuss the use of gut microbiota-derived mediators as potential markers in NAFLD.
Collapse
|
92
|
Arunima A, Das JK, Suar M. Gut Microbes in Liver Diseases. DIETARY INTERVENTIONS IN GASTROINTESTINAL DISEASES 2019:117-131. [DOI: 10.1016/b978-0-12-814468-8.00010-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
93
|
Dam B, Misra A, Banerjee S. Role of Gut Microbiota in Combating Oxidative Stress. OXIDATIVE STRESS IN MICROBIAL DISEASES 2019:43-82. [DOI: 10.1007/978-981-13-8763-0_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
94
|
Current Models of Fatty Liver Disease; New Insights, Therapeutic Targets and Interventions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:33-58. [PMID: 30919331 DOI: 10.1007/978-3-030-12668-1_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) encompasses a spectrum of disorders ranging from simple steatosis to steatosis with inflammation and fibrosis. NAFLD is currently the most prevalent chronic liver disease worldwide, with a global prevalence of 25%, and is soon projected to be the leading cause for liver transplantation in the US. Alarmingly, few effective pharmacotherapeutic approaches are currently available to block or attenuate development and progression of NAFLD. Preclinical models are critical for unraveling the complex and multi-factorial etiology of NAFLD and for testing potential therapeutics. Here we review preclinical models that have been instrumental in highlighting molecular and cellular mechanisms underlying the pathogenesis of NAFLD and in facilitating early proof-of-concept investigations into novel intervention strategies.
Collapse
|
95
|
Porras D, Nistal E, Martínez-Flórez S, González-Gallego J, García-Mediavilla MV, Sánchez-Campos S. Intestinal Microbiota Modulation in Obesity-Related Non-alcoholic Fatty Liver Disease. Front Physiol 2018; 9:1813. [PMID: 30618824 PMCID: PMC6305464 DOI: 10.3389/fphys.2018.01813] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/05/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity and associated comorbidities, including non-alcoholic fatty liver disease (NAFLD), are a major concern to public well-being worldwide due to their high prevalence among the population, and its tendency on the rise point to as important threats in the future. Therapeutic approaches for obesity-associated disorders have been circumscribed to lifestyle modifications and pharmacological therapies have demonstrated limited efficacy. Over the last few years, different studies have shown a significant role of intestinal microbiota (IM) on obesity establishment and NAFLD development. Therefore, modulation of IM emerges as a promising therapeutic strategy for obesity-associated diseases. Administration of prebiotic and probiotic compounds, fecal microbiota transplantation (FMT) and exercise protocols have shown a modulatory action over the IM. In this review we provide an overview of current approaches targeting IM which have shown their capacity to counteract NAFLD and metabolic syndrome features in human patients and animal models.
Collapse
Affiliation(s)
- David Porras
- Institute of Biomedicine, University of León, León, Spain
| | - Esther Nistal
- Institute of Biomedicine, University of León, León, Spain.,Department of Gastroenterology, Complejo Asistencial Universitario de León, León, Spain
| | | | - Javier González-Gallego
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - María Victoria García-Mediavilla
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| | - Sonia Sánchez-Campos
- Institute of Biomedicine, University of León, León, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Madrid, Spain
| |
Collapse
|
96
|
Schwenger KJP, Bolzon CM, Li C, Allard JP. Non-alcoholic fatty liver disease and obesity: the role of the gut bacteria. Eur J Nutr 2018; 58:1771-1784. [PMID: 30306296 DOI: 10.1007/s00394-018-1844-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty-liver disease (NAFLD) is now considered one of the leading causes of liver disease worldwide and is associated with metabolic syndrome and obesity. There are several factors contributing to the disease state. Recent research suggests that the intestinal microbiota (IM) and bacterial products may play a role through several mechanisms which include increased energy uptake, intestinal permeability and chronic inflammation. In addition to diet and exercise, treatment options targeting the IM are being investigated and include the use of pre-, pro- and synbiotics as well as the possibility of fecal microbial transfers. This literature review explores the relationship between NAFLD and the IM as well as highlight new IM treatment options that may become available in the near future.
Collapse
Affiliation(s)
- Katherine J P Schwenger
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Colin M Bolzon
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Carrie Li
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada
| | - Johane P Allard
- Toronto General Hospital, University Health Network, 585 University Avenue, 9-973, Toronto, ON, M5G 2C4, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,Department of Nutritional Sciences, University of Toronto, Toronto, Canada.
| |
Collapse
|
97
|
Li Y, Liu T, Yan C, Xie R, Guo Z, Wang S, Zhang Y, Li Z, Wang B, Cao H. Diammonium Glycyrrhizinate Protects against Nonalcoholic Fatty Liver Disease in Mice through Modulation of Gut Microbiota and Restoration of Intestinal Barrier. Mol Pharm 2018; 15:3860-3870. [PMID: 30036479 DOI: 10.1021/acs.molpharmaceut.8b00347] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD), as a common chronic liver disorder, is prevalent in the world. Recent evidence demonstrates that the "gut-liver axis" is related well to the progression of NAFLD, which regards gut microbiota and the intestinal barrier as two critical factors correlated with NAFLD. Diammonium glycyrrhizinate (DG), a compound of the natural bioactive pentacyclic triterpenoid glycoside, is the main component of licorice root extracts. The anti-inflammatory and liver protection effects of DG have already been reported, but to date, the mechanism has not been fully elucidated. In this research, we observed that DG reduced body weight, liver steatosis, as well as hepatic inflammation in NAFLD model mice induced by a high-fat diet. Illumina sequencing of the 16S rRNA revealed that DG intervention notably altered the composition of the gut microbiota in NAFLD mice. The richness of gut microbiota was significantly increased by DG. Specifically, DG reduced the Firmicutes-to- Bacteroidetes ratio and the endotoxin-producing bacteria such as Desulfovibrio and elevated the abundance of probiotics such as Proteobacteria and Lactobacillus. DG could augment the levels of short-chain fatty acid (SCFA)-producing bacteria such as Ruminococcaceae and Lachnospiraceae and promote SCFA production. In addition, DG supplementation dramatically alleviated the intestinal low-grade inflammation. Meanwhile, DG improved the expression of tight junction proteins, the goblet cell number, and mucin secretion and sequentially enhanced the function of intestinal barrier. Collectively, the prevention of NAFLD by DG might be mediated by modulating gut microbiota and restoring the intestinal barrier.
Collapse
|
98
|
Schwenger KJP, Chen L, Chelliah A, Da Silva HE, Teterina A, Comelli EM, Taibi A, Arendt BM, Fischer S, Allard JP. Markers of activated inflammatory cells are associated with disease severity and intestinal microbiota in adults with non‑alcoholic fatty liver disease. Int J Mol Med 2018; 42:2229-2237. [PMID: 30085339 DOI: 10.3892/ijmm.2018.3800] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/15/2018] [Indexed: 01/18/2023] Open
Abstract
Several mechanisms contribute to the pathogenesis of non‑alcoholic fatty liver disease (NAFLD). The intestinal microbiota (IM) and liver immune cells (LIC) may serve a role, but there has been no previous study assessing potential associations between IM and LIC. The aim of the present study was to investigate whether there are differences in LIC markers between patients with NAFLD and healthy controls (HC), and to determine whether these markers are associated with specific IM. The present prospective, cross‑sectional study examined a cohort of adults with liver biopsy‑confirmed NAFLD and HC. Clinical and laboratory data were collected. Fecal IM was assessed by quantitative polymerase chain reaction and LIC, by immunohistochemistry. NAFLD activity score (NAS) was used for disease severity. Liver immune cell counts were increased in patients with NAFLD (n=34) vs. HC (n=8) and this was associated with disease severity. Hematopoietic cell marker cluster of differentiation (CD)45+ and Kupffer cell marker CD163+ were higher in NAFLD compared with HC, and those with an NAS ≥5 had higher levels of CD20+ cells, a marker of B cells, vs. a NAS of 0 or 1‑4. Additionally, from those patients (5 HC, 34 NAFLD), IM was measured. Specific immune cells in portal or lobular areas correlated with specific fecal IM, suggesting a potential association between IM and liver inflammation in patients with NAFLD. Specifically, Faecalibacterium prausnitzii was negatively correlated with CD45+ (r= ‑0.394; P=0.015) and CD163+ (r= ‑0.371; P=0.022) cells in the portal tract and Prevotella was negatively correlated with CD20+ (r= ‑0.353; P=0.028) cells in the liver lobule. Other taxa exhibited no correlation. In conclusion, the present study demonstrated a potential association between IM and liver inflammation in NAFLD.
Collapse
Affiliation(s)
| | - Lina Chen
- Department of Medicine, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada
| | | | - Hannah E Da Silva
- Department of Nutrition, Sunnybrook Health Sciences Center, Toronto, ON M4N 3M5, Canada
| | - Anastasia Teterina
- Department of Medicine, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Elena M Comelli
- Department of Nutritional Sciences, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Amel Taibi
- Department of Nutritional Sciences, University of Toronto, Toronto, ON M5S 3E2, Canada
| | - Bianca M Arendt
- Department of Medicine, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Sandra Fischer
- Department of Pathology, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Johane P Allard
- Department of Medicine, University of Toronto, Toronto, ON M5S 3E2, Canada
| |
Collapse
|
99
|
Ye H, Li Q, Zhang Z, Sun M, Zhao C, Zhang T. Effect of a novel potential probiotic Lactobacillus paracasei Jlus66 isolated from fermented milk on nonalcoholic fatty liver in rats. Food Funct 2018; 8:4539-4546. [PMID: 29106426 DOI: 10.1039/c7fo01108c] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the main cause of chronic liver disease worldwide. Previous evidence indicates that probiotics can be applied as a therapeutic agent for NAFLD. In this study, the potential probiotic strain Lactobacillus paracasei Jlus66 was isolated from natural fermented milk by a culture-dependent method, and its probiotic potentials were tested by established in vitro tests. In addition, the protective effect of Lactobacillus paracasei Jlus66 against NAFLD was evaluated in rat models. Compared with the high-fat-diet (HFD) group, the rats administered with 4 × 1010 cfu Jlus66 had significantly lower body weight gain, serum triglyceride (TG), low-density lipoprotein (LDL) as well as aminotransferase (ALT). Histopathological analysis showed Jlus66 also reduced the level of hepatic triglycerides and steatosis. From the above we conclude that L. paracasei Jlus66 has great potential as a probiotic in protecting from NAFLD.
Collapse
Affiliation(s)
- Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| | | | | | | | | | | |
Collapse
|
100
|
Shibata M, Iwane T, Higuchi R, Suwa K, Nakajima K. Potential common factors associated with predisposition to common cold in middle-aged and elderly Japanese: A community-based cross-sectional study. Medicine (Baltimore) 2018; 97:e10729. [PMID: 29768343 PMCID: PMC5976333 DOI: 10.1097/md.0000000000010729] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 04/23/2018] [Indexed: 02/05/2023] Open
Abstract
People worldwide frequently catch a common cold, which occasionally develops into secondary severe conditions such as pneumonia. However, it is unclear whether predisposition to the common cold is associated with the individual's characteristics including age, body weight, lifestyles, diets, and intestinal functions, besides exposure to a responsible pathogen. We addressed this issue epidemiologically considering many relevant clinical factors.We reviewed data from a cross-sectional study consisting of 39,524 apparently healthy Japanese aged 40 to 79 years (26,975 men and 12,549 women) who underwent a checkup in 2007. Self-reported predisposition to common cold (SPCC) and relevant clinical conditions and parameters were considered.We observed no significant difference in most clinical parameters including age, body mass index (BMI), glycated hemoglobin (HbA1c), and prevalence of men and current smokers between subjects with and without SPCC. In univariate analysis, circulating white blood cell (WBC) count and serum alanine-aminotransferase (ALT) were significantly higher in subjects with SPCC than in those without, whereas serum high-density lipoprotein cholesterol (HDL-C) and duration of sleep were lower. In logistic regression analysis after full adjustment for relevant confounding factors, BMI categories except BMI of ≥27.0 kg/m were significantly associated with SPCC compared with BMI of 23.0 to 24.9 kg/m. Short duration of sleep (≤5 hours), occasional alcohol drinking, and no-exercise were significantly associated with SPCC compared with 7 hours sleep duration, no-drinking alcohol, and low frequent exercise (twice per month), respectively. All gastrointestinal disorders (gastric complaints, constipation, and diarrhea) were independently associated with SPCC. Imbalanced diet and taking a snack were also associated with SPCC in a degree dependent manner. Furthermore, WBC count, serum ALT, and HDL-C (as continuous variables) were associated with SPCC (HDL-C was inversely), whereas no significant association was observed between SPCC and age, smoking, HbA1c, and pharmacotherapy for diabetes, hypertension, and dyslipidemia.Our results demonstrated that multifactorial conditions and parameters might be simultaneously associated with the predisposition to common cold. Prospective studies including detailed common cold questionnaire and measurements are needed to confirm currently suspected causative and protective factors.
Collapse
Affiliation(s)
- Michi Shibata
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services
- Department of Nutrition, St. Marianna University School of Medicine Hospital, Kawasaki, Kanagawa
| | - Taizo Iwane
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services
| | - Ryoko Higuchi
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services
| | - Kaname Suwa
- Saitama Health Promotion Corporation, Yoshimimachi, Hikigun
| | - Kei Nakajima
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services
- Department of Endocrinology and Diabetes, Saitama Medical Center, Saitama Medical University, Kawagoe, Saitama, Japan
| |
Collapse
|