101
|
Fan L, Zhao X, Tong Q, Zhou X, Chen J, Xiong W, Fang J, Wang W, Shi C. Interactions of Dihydromyricetin, a Flavonoid from Vine Tea (Ampelopsis grossedentata) with Gut Microbiota. J Food Sci 2018; 83:1444-1453. [PMID: 29660761 DOI: 10.1111/1750-3841.14128] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/02/2018] [Accepted: 02/22/2018] [Indexed: 01/20/2023]
Abstract
Dihydromyricetin (DMY) is the main bioactive constituent in vine tea (Ampelopsis grossedentata), which was predominantly distributed in the gastrointestinal tract and showed poor oral bioavailability. Our aim was to systematically investigate the interactions of DMY with gut microbiota. Through the metabolism study of DMY by fecal microflora in vitro, it was found that DMY could be metabolized into three metabolites by fecal microflora via reduction and dehydroxylation pathways, and the dehydroxylation metabolite was the dominant one. Meanwhile, in order to consider the influence of gut microbiota metabolism on the pharmacokinetics of DMY, the pharmacokinetics of DMY in control and pseudo-germ-free rats were compared. It was shown that area under the curve (AUC) could only slightly increase, however, peak concentration (Cmax ) could significantly increase in the pseudo-germ-free rats compared with the control rats, which indicated the gut microbiota metabolism played an important role in the pharmacokinetics of DMY. In addition, the long-term influence of DMY on gut microbiota composition by using 16S rRNA pyrosequencing was further investigated. And it was found that DMY could markedly alter the richness and diversity of the gut microbiota and modulate the gut microbiota composition. The present findings will be helpful for the future development and clinical application of DMY. PRACTICAL APPLICATION The gut microbiota plays an important role in the pharmacokinetics of flavonoids. As well, the long-term supplements of flavonoids could alter the gut microbiota composition in turn. The study aims to clarify the mutual interaction of DMY with gut microbiota, which may lead to new information with respect to the mechanism study and clinical application of DMY.
Collapse
Affiliation(s)
- Li Fan
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Xinyuan Zhao
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Qing Tong
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Xiya Zhou
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Jing Chen
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Wei Xiong
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Jianguo Fang
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Wenqing Wang
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| | - Chunyang Shi
- Dept. of Pharmacy, Tongji Hospital Affiliated with Tongji Medical College, Huazhong Univ. of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
102
|
Effects of shenling baizhu powder herbal formula on intestinal microbiota in high-fat diet-induced NAFLD rats. Biomed Pharmacother 2018; 102:1025-1036. [PMID: 29710519 DOI: 10.1016/j.biopha.2018.03.158] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/26/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Worldwide, non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease closely associated with obesity, diabetes and other metabolic diseases. Shenling Baizhu powder (SLBZP), a formulation of a variety of natural medicinal plants, has hepatoprotective properties and clinical efficacy in treating non-infectious intestinal disease. SLBZP has improved NAFLD symptoms; however, its mechanism of action is unknown. METHODS We established an NAFLD model in rats given a high-fat diet (HFD), administered different interventions and measured serum biochemical indices and inflammatory factors. Liver tissues were stained with hematoxylin and eosin (HE) and oil red O, and colon tissues were analyzed by immunohistochemistry. The expression profiles of liver TLR4 pathway related protein was confirmed by western blotting. Changes in intestinal microbiota composition were analyzed using a 16S rDNA sequencing technique. RESULTS Of note, SLBZP effectively reduced body weight in HFD-fed rats (p < 0.05). Serum biochemical analysis indicated that SLBZP decreased the serum level of total cholesterol (TC) and improved liver function. Additionally, SLBZP decreased the serum level of endotoxin, tumor necrosis factor α (TNF-α), interleukin-1β (IL-β) (p < 0.05), and decreased the expression of TLR4 pathway related protein. Pathological examination showed that SLBZP alleviates hepatic steatosis and repairs colon mucosa. Microbiome analysis revealed that SLBZP improved the abundance of intestinal microbiota. In taxonomy-based analysis, compared with control rats, SLBZP-treated rats showed obvious changes in intestinal microbiota composition. Moreover, SLBZP increased the relative abundance of short-chain fatty acid (SCFA)-producing bacteria, including Bifidobacterium and Anaerostipes. CONCLUSION Taken together, these results suggest that the effects of SLBZP against NAFLD may be related to the increased abundance of beneficial gut microbiota and decreased levels of LPS in the portal vein.
Collapse
|
103
|
Chu H, Williams B, Schnabl B. Gut microbiota, fatty liver disease, and hepatocellular carcinoma. LIVER RESEARCH 2018; 2:43-51. [PMID: 30416839 PMCID: PMC6223644 DOI: 10.1016/j.livres.2017.11.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intestinal bacteria contribute to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Recently developed microbial profiling techniques are beginning to shed light on the nature of the changes in the gut microbiota that accompany NAFLD and non-alcoholic steatohepatitis (NASH). In this review, we summarize the role of gut microbiota in the development of NAFLD, NASH, and hepatocellular carcinoma (HCC). We highlight the mechanisms by which gut microbiota contribute to NAFLD/NASH, including through alterations in gut epithelial permeability, choline metabolism, endogenous alcohol production, release of inflammatory cytokines, regulation of hepatic Toll-like receptor (TLR), and bile acid metabolism. In addition, we analyze possible mechanisms for enhanced hepatic carcinogenesis, including alterations in bile acid metabolism, release of inflammatory cytokines, and expression of TLR-4. Finally, we describe therapeutic approaches for NAFLD/NASH and preventive strategies for HCC involving modulation of the intestinal microbiota or affected host pathways. Although recent studies have provided useful information, large-scale prospective studies are required to better characterize the intestinal microbiota and metabolome, in order to demonstrate a causative role for changes in the gut microbiota in the etiology of NAFLD/NASH, to identify new therapeutic strategies for NAFLD/NASH, and to develop more effective methods of preventing HCC.
Collapse
Affiliation(s)
- Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA,epartment of Medicine, VA San Diego Healthcare System, San Diego, CA, USA,Corresponding author. Department of Medicine, University of California San Diego, Biomedical Research Facility 2 (BRF2), La Jolla, CA, USA. (B. Schnabl)
| |
Collapse
|
104
|
Emerging metabolic risk factors in hepatocellular carcinoma and their influence on the liver microenvironment. Biochim Biophys Acta Mol Basis Dis 2018; 1864:607-617. [PMID: 29197664 DOI: 10.1016/j.bbadis.2017.11.026] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 11/14/2017] [Accepted: 11/28/2017] [Indexed: 12/14/2022]
|
105
|
Saltzman ET, Palacios T, Thomsen M, Vitetta L. Intestinal Microbiome Shifts, Dysbiosis, Inflammation, and Non-alcoholic Fatty Liver Disease. Front Microbiol 2018; 9:61. [PMID: 29441049 PMCID: PMC5797576 DOI: 10.3389/fmicb.2018.00061] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/10/2018] [Indexed: 12/16/2022] Open
Abstract
Adverse fluctuations in the distribution of the intestinal microbiome cohort has been associated with the onset of intra- and extra-intestinal inflammatory conditions, like the metabolic syndrome (MetS) and it's hepatic manifestation, non-alcoholic fatty liver disease (NAFLD). The intestinal microbial community of obese compared to lean subjects has been shown to undergo configurational shifts in various genera, including but not limited to increased abundances of Prevotella, Escherichia, Peptoniphilus, and Parabacteroides and decreased levels of Bifidobacteria, Roseburia, and Eubacteria genera. At the phylum level, decreased Bacteroidetes and increased Firmicutes have been reported. The intestinal microbiota therefore presents an important target for designing novel therapeutic modalities that target extra-intestinal inflammatory disorders, such as NAFLD. This review hypothesizes that disruption of the intestinal-mucosal macrophage interface is a key factor in intestinal-liver axis disturbances. Intestinal immune responses implicated in the manifestation, maintenance and progression of NAFLD provide insights into the dialogue between the intestinal microbiome, the epithelia and mucosal immunity. The pro-inflammatory activity and immune imbalances implicated in NAFLD pathophysiology are reported to stem from dysbiosis of the intestinal epithelia which can serve as a source of hepatoxic effects. We posit that the hepatotoxic consequences of intestinal dysbiosis are compounded through intestinal microbiota-mediated inflammation of the local mucosa that encourages mucosal immune dysfunction, thus contributing important plausible insight in NAFLD pathogenesis. The administration of probiotics and prebiotics as a cure-all remedy for all chronic diseases is not advocated, instead, the incorporation of evidence based probiotic/prebiotic formulations as adjunctive modalities may enhance lifestyle modification management strategies for the amelioration of NAFLD.
Collapse
Affiliation(s)
- Emma T. Saltzman
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Talia Palacios
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Michael Thomsen
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| | - Luis Vitetta
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Medlab Clinical, Sydney, NSW, Australia
| |
Collapse
|
106
|
Zhou YZ, Yan ML, Gao L, Zhang JQ, Qin XM, Zhang X, Du GH. Metabonomics approach to assessing the metabolism variation and gender gap of Drosophila melanogaster in aging process. Exp Gerontol 2017; 98:110-119. [DOI: 10.1016/j.exger.2017.07.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 07/25/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023]
|
107
|
Gutiérrez-Grobe Y, Juárez-Hernández E, Sánchez-Jiménez B, Uribe-Ramos M, Ramos-Ostos M, Uribe M, Chávez-Tapia N. Less liver fibrosis in metabolically healthy compared with metabolically unhealthy obese patients with non-alcoholic fatty liver disease. DIABETES & METABOLISM 2017; 43:332-337. [DOI: 10.1016/j.diabet.2017.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 02/07/2017] [Accepted: 02/15/2017] [Indexed: 12/14/2022]
|
108
|
Shen F, Zheng RD, Sun XQ, Ding WJ, Wang XY, Fan JG. Gut microbiota dysbiosis in patients with non-alcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2017; 16:375-381. [PMID: 28823367 DOI: 10.1016/s1499-3872(17)60019-5] [Citation(s) in RCA: 390] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 11/30/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gut microbiota plays a significant role in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). This study aimed to assess the contribution of gut microbiota dysbiosis to the pathogenesis of NAFLD. METHODS Forty-seven human feces samples (25 NAFLD patients and 22 healthy subjects) were collected and 16S rDNA amplicon sequencing was conducted on Hiseq 2000 platform. Discrepancy of species composition between controls and NAFLD group was defined by Metastats analysis under P value <0.01. RESULTS NAFLD patients harbored lower gut microbiota diversity than healthy subjects did. In comparison to the control group, the Proteobacteria (13.50%) and Fusobacteria (2.76%) phyla were more abundant in NAFLD patients. Additionally, the Lachnospiraceae (21.90%), Enterobacteriaceae (12.02%), Erysipelotrichaceae (3.83%), and Streptococcaceae (1.39%) families, as well as the Escherichia_Shigella (10.84%), Lachnospiraceae_Incertae_Sedis (7.79%), and Blautia (4.95%) genera were enriched in the NAFLD group. However, there was a lower abundance of Prevotella in the NAFLD group than that in the control group (5.83% vs 27.56%, P<0.01). The phylum Bacteroidetes (44.63%) also tended to be more abundant in healthy subjects, and the families Prevotellaceae (28.66%) and Ruminococcaceae (26.44%) followed the same trend. Compared to those without non-alcoholic steatohepatitis (NASH), patients with NASH had higher abundance of genus Blautia (5.82% vs 2.25%; P=0.01) and the corresponding Lachnospiraceae family (24.33% vs 14.21%; P<0.01). Patients with significant fibrosis had a higher abundance of genus Escherichia_Shigella (12.53% vs 1.97%; P<0.01) and the corresponding Enterobacteriaceae family (13.92% vs 2.07%; P<0.01) compared to those with F0/F1 fibrosis. CONCLUSIONS NAFLD patients and healthy subjects harbor varying gut microbiota. In contrast to the results of previous research on children, decreased levels of Prevotella might be detrimental for adults with NAFLD. The increased level of the genus Blautia, the family Lachnospiraceae, the genus Escherichia_Shigella, and the family Enterobacteriaceae may be a primary contributor to NAFLD progression.
Collapse
Affiliation(s)
- Feng Shen
- Department of Gastroenterology, Shanghai, China
| | - Rui-Dan Zheng
- Research and Therapy Centre for Liver Disease, Zhengxing Hospital, Zhangzhou 363000, China
| | - Xing-Qiang Sun
- Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Department of Microbial Genomics Research, BGI Shenzhen, Shenzhen 518083, China
| | | | | | - Jian-Gao Fan
- Department of Gastroenterology, Shanghai, China.
| |
Collapse
|
109
|
Diet-induced obesity and weight loss alter bile acid concentrations and bile acid-sensitive gene expression in insulin target tissues of C57BL/6J mice. Nutr Res 2017; 46:11-21. [PMID: 29173647 DOI: 10.1016/j.nutres.2017.07.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/13/2017] [Accepted: 07/20/2017] [Indexed: 12/31/2022]
Abstract
Bile acids (BAs) influence the metabolism of glucose, lipids, and energy expenditure. We hypothesized that BA concentrations and related gene expression would be altered in lean (low-fat diet fed; LFD) vs diet-induced obese (high-fat diet fed; HFD) groups of mice and that some detected changes would remain after weight loss in an HFD group switched to the LFD (SW). Taurine conjugates dominated the bile acid composition of the liver, epididymal white adipose tissue (eWAT), and hypothalamus, with the latter having lower levels (~95%, ~95%, and ~80%, respectively; P<.05). Plasma conjugated bile acids were elevated in the HFD relative to the LFD and SW animals. Total hepatic BA concentrations decreased in obese mice fed HFD, and levels returned to preobese levels in the SW group. Subtle changes in unconjugated bile acids were detected in the eWAT, hypothalamus, and muscle. Liver expression of a variety of enzymes involved in BA synthesis (eg, Cyp27a1, Acox2), BA transport (eg, Slc22a8), and BA-sensitive receptors (Fxr, Tgr5) were unchanged by HFD feeding but decreased with SW. Other hepatic enzymes were induced in the SW group (eg, Amacr and Bal). In eWAT, Cyp27a1 and Acox2 also declined in the SW group, whereas the HFD group showed reduced expression of BA transporters (eg, Abcc3), and changes in Fxr and Tgr5 were unclear. Therefore, although most detectable changes in BA metabolism associated with diet-induced obesity are reversed by diet-induced weight loss, some effects on BA composition, concentrations, and gene expression can persist after weight loss.
Collapse
|
110
|
New insight into inter-organ crosstalk contributing to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Protein Cell 2017. [PMID: 28643267 PMCID: PMC5818366 DOI: 10.1007/s13238-017-0436-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver dysfunction and a significant global health problem with substantial rise in prevalence over the last decades. It is becoming increasingly clear that NALFD is not only predominantly a hepatic manifestation of metabolic syndrome, but also involves extra-hepatic organs and regulatory pathways. Therapeutic options are limited for the treatment of NAFLD. Accordingly, a better understanding of the pathogenesis of NAFLD is critical for gaining new insight into the regulatory network of NAFLD and for identifying new targets for the prevention and treatment of NAFLD. In this review, we emphasize on the current understanding of the inter-organ crosstalk between the liver and peripheral organs that contributing to the pathogenesis of NAFLD.
Collapse
|
111
|
Behrouz V, Jazayeri S, Aryaeian N, Zahedi MJ, Hosseini F. Effects of Probiotic and Prebiotic Supplementation on Leptin, Adiponectin, and Glycemic Parameters in Non-alcoholic Fatty Liver Disease: A Randomized Clinical Trial. Middle East J Dig Dis 2017; 9:150-157. [PMID: 28894517 PMCID: PMC5585907 DOI: 10.15171/mejdd.2017.66] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND
According to previous studies, probiotic and prebiotic supplementation have desirable effects
on glycemic parameters. Thus far, the effect of supplementation on the glycemic parameters and
adipokines in non-alcoholic fatty liver disease (NAFLD) has not been assessed. Therefore, the
aim of this study was to determine the effects of supplementation with probiotic and prebiotic on
adiokines and glycemic parameters in the patients with NAFLD.
METHODS
In the present randomized, double-blind, placebo-controlled trial, 89 patients with NAFLD
were randomly divided into three groups to receive one probiotic capsule + 16 g/d maltodextrin
(probiotic group) or 16 g/d oligofructose powder + one placebo capsule (prebiotic group), and
one placebo capsule + 16 g/d maltodextrin (control group) for 12 weeks. All the subjects in the
study were advised to follow the weight loss diet and physical activity recommendations during
the intervention. Fasting blood samples were taken at baseline and after the intervention to measure
leptin, adiponectin, insulin, and fasting blood sugar.
RESULTS
At the end of the study, serum concentrations of leptin, insulin, and HOMA-IR decreased
significantly in the probiotic and prebiotic groups compared with the control group. Despite the
changes within the groups, serum concentrations of adiponectin did not change significantly between
the three groups. Also, fasting blood sugar did not change between the groups, but decreased in the
prebiotic group. Quantitative insulin-sensitivity check index (QUICKI) increased significantly in
probiotic and prebiotic groups compared with the control group.
CONCLUSION
Probiotic and prebiotic supplementation along with lifestyle intervention has a favorable impact
on glycemic parameters and leptin levels compared with lifestyle intervention alone.
Collapse
Affiliation(s)
- Vahideh Behrouz
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Shima Jazayeri
- Research Center for Prevention of Cardiovascular Disease, Iran University of Medical Sciences, Tehran, Iran
| | - Naheed Aryaeian
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Hosseini
- Department of Epidemiology and Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
112
|
Sun LY, Yang YS, Qu W, Zhu ZJ, Wei L, Ye ZS, Zhang JR, Sun XY, Zeng ZG. Gut microbiota of liver transplantation recipients. Sci Rep 2017. [PMID: 28630433 PMCID: PMC5476624 DOI: 10.1038/s41598-017-03476-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The characteristics of intestinal microbial communities may be affected by changes in the pathophysiology of patients with end-stage liver disease. Here, we focused on the characteristics of intestinal fecal microbial communities in post-liver transplantation (LT) patients in comparison with those in the same individuals pre-LT and in healthy individuals. The fecal microbial communities were analyzed via MiSeq-PE250 sequencing of the V4 region of 16S ribosomal RNA and were then compared between groups. We found that the gut microbiota of patients with severe liver disease who were awaiting LT was significantly different from that of healthy controls, as represented by the first principal component (p = 0.0066). Additionally, the second principal component represented a significant difference in the gut microbiota of patients between pre-LT and post-LT surgery (p = 0.03125). After LT, there was a significant decrease in the abundance of certain microbial species, such as Actinobacillus, Escherichia, and Shigella, and a significant increase in the abundance of other microbial species, such as Micromonosporaceae, Desulfobacterales, the Sarcina genus of Eubacteriaceae, and Akkermansia. Based on KEGG profiles, 15 functional modules were enriched and 21 functional modules were less represented in the post-LT samples compared with the pre-LT samples. Our study demonstrates that fecal microbial communities were significantly altered by LT.
Collapse
Affiliation(s)
- Li-Ying Sun
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yun-Sheng Yang
- Institute of Digestive Diseases, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, China.
| | - Wei Qu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhi-Jun Zhu
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lin Wei
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhi-Sheng Ye
- Institute of Developmental Biology and Molecular Medicine, Fudan University, Shanghai, China
| | - Jian-Rui Zhang
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiao-Ye Sun
- Tianjin First Central Hospital, Tianjin, China
| | - Zhi-Gui Zeng
- Liver Transplantation Center, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
113
|
Panasevich MR, Peppler WT, Oerther DB, Wright DC, Rector RS. Microbiome and NAFLD: potential influence of aerobic fitness and lifestyle modification. Physiol Genomics 2017; 49:385-399. [PMID: 28600319 DOI: 10.1152/physiolgenomics.00012.2017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease with prevalence rates that are on the rise in the US and worldwide. NAFLD encompasses a spectrum of liver pathologies including simple steatosis to nonalcoholic steatohepatitis (NASH) with inflammation and fibrosis. The gut microbiome has emerged as a potential therapeutic target in combating metabolic diseases including obesity, Type 2 diabetes, and NAFLD/NASH. Diet-induced obesity/Western style diet feeding causes severe microbial dysbiosis initiating a microbiome signature that promotes metabolite production that directly impacts hepatic metabolism. Changes in lifestyle (i.e., diet, exercise, and aerobic fitness) improve NAFLD outcomes and can significantly influence the microbiome. However, directly linking lifestyle-induced remodeling of the microbiome to NAFLD pathogenesis is not well understood. Understanding the reshaping of the microbiome and the metabolites produced and their subsequent actions on hepatic metabolism are vital in understanding the gut-liver axis. In this review, we 1) discuss microbiome-derived metabolites that significantly contribute to the gut-liver axis and are directly linked to NAFLD/NASH and 2) present evidence on lifestyle modifications reshaping the microbiome and the potential therapeutic aspects in combating the disease.
Collapse
Affiliation(s)
- Matthew R Panasevich
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri.,Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology; University of Missouri, Columbia, Missouri
| | - Willem T Peppler
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Daniel B Oerther
- Department of Civil, Architectural, and Environmental Engineering, Missouri University of Science and Technology, Rolla, Missouri; and
| | - David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - R Scott Rector
- Research Service, Harry S Truman Memorial Veterans Medical Center, Columbia, Missouri; .,Department of Medicine, Division of Gastroenterology and Hepatology, University of Missouri, Columbia, Missouri.,Department of Nutrition and Exercise Physiology; University of Missouri, Columbia, Missouri
| |
Collapse
|
114
|
Abstract
Bile acids play a critical role in the regulation of glucose, lipid, and energy metabolism through activation of the nuclear bile acid receptor farnesoid X receptor (FXR) and membrane G protein-coupled bile acid receptor-1 (Gpbar-1, aka TGR5). Agonist activation of FXR and TGR5 improves insulin and glucose sensitivity and stimulates energy metabolism to prevent diabetes, obesity, and non-alcoholic fatty liver disease (NAFLD). Bile acids have both pro- and anti-inflammatory actions through FXR and TGR5 in the intestine and liver. In the intestine, bile acids activate FXR and TGR5 to stimulate stimulate fibroblast growth factor 15 and glucagon-like peptide-1 secretion. FXR and TGR5 agonists may have therapeutic potential for treating liver-related metabolic diseases, such as diabetes and NAFLD.
Collapse
Affiliation(s)
- John Y. L. Chiang
- Corresponding author. Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical, University, Rootstown, OH, USA, (John Y. L. Chiang)
| |
Collapse
|
115
|
Kim DH, Kim H, Jeong D, Kang IB, Chon JW, Kim HS, Song KY, Seo KH. Kefir alleviates obesity and hepatic steatosis in high-fat diet-fed mice by modulation of gut microbiota and mycobiota: targeted and untargeted community analysis with correlation of biomarkers. J Nutr Biochem 2017; 44:35-43. [DOI: 10.1016/j.jnutbio.2017.02.014] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/30/2016] [Accepted: 02/16/2017] [Indexed: 02/08/2023]
|
116
|
Ma Y, Liu J, Rhodes C, Nie Y, Zhang F. Ethical Issues in Fecal Microbiota Transplantation in Practice. THE AMERICAN JOURNAL OF BIOETHICS : AJOB 2017; 17:34-45. [PMID: 28430065 DOI: 10.1080/15265161.2017.1299240] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Fecal microbiota transplantation (FMT) has demonstrated efficacy and is increasingly being used in the treatment of patients with recurrent Clostridium difficile infection. Despite a lack of high-quality trials to provide more information on the long-term effects of FMT, there has been great enthusiasm about the potential for expanding its applications. However, FMT presents many serious ethical and social challenges that must be addressed as part of a successful regulatory policy response. In this article, we draw on a sample of the scientific and bioethics literatures to examine clusters of ethical and social issues arising in five main areas: (1) informed consent and the vulnerability of patients; (2) determining what a "suitable healthy donor" is; (3) safety and risk; (4) commercialization and potential exploitation of vulnerable patients; and (5) public health implications. We find that these issues are complex and worthy of careful consideration by health care professionals. Desperation of a patient should not be the basis for selecting treatment with FMT, and the patient's interests should always be of paramount concern. Authorities must prioritize development of appropriate and effective regulation of FMT to safeguard patients and donors, promote further research into safety and efficacy, and avoid abuse of the treatment.
Collapse
Affiliation(s)
| | | | | | | | - Faming Zhang
- e Second Affiliated Hospital of Nanjing Medical University
| |
Collapse
|
117
|
Dahiya DK, Renuka, Puniya M, Shandilya UK, Dhewa T, Kumar N, Kumar S, Puniya AK, Shukla P. Gut Microbiota Modulation and Its Relationship with Obesity Using Prebiotic Fibers and Probiotics: A Review. Front Microbiol 2017; 8:563. [PMID: 28421057 PMCID: PMC5378938 DOI: 10.3389/fmicb.2017.00563] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/20/2017] [Indexed: 12/12/2022] Open
Abstract
In the present world scenario, obesity has almost attained the level of a pandemic and is progressing at a rapid rate. This disease is the mother of all other metabolic disorders, which apart from placing an added financial burden on the concerned patient also has a negative impact on his/her well-being and health in the society. Among the various plausible factors for the development of obesity, the role of gut microbiota is very crucial. In general, the gut of an individual is inhabited by trillions of microbes that play a significant role in host energy homeostasis by their symbiotic interactions. Dysbiosis in gut microbiota causes disequilibrium in energy homeostasis that ultimately leads to obesity. Numerous mechanisms have been reported by which gut microbiota induces obesity in experimental models. However, which microbial community is directly linked to obesity is still unknown due to the complex nature of gut microbiota. Prebiotics and probiotics are the safer and effective dietary substances available, which can therapeutically alter the gut microbiota of the host. In this review, an effort was made to discuss the current mechanisms through which gut microbiota interacts with host energy metabolism in the context of obesity. Further, the therapeutic approaches (prebiotics/probiotics) that helped in positively altering the gut microbiota were discussed by taking experimental evidence from animal and human studies. In the closing statement, the challenges and future tasks within the field were discussed.
Collapse
Affiliation(s)
- Dinesh K Dahiya
- Advanced Milk Testing Research Laboratory, Post Graduate Institute of Veterinary Education and Research - Rajasthan University of Veterinary and Animal Sciences at BikanerJaipur, India
| | - Renuka
- Department of Biochemistry, Basic Medical Science, South Campus, Panjab UniversityChandigarh, India
| | - Monica Puniya
- Food Safety Management System Division, Food Safety and Standards Authority of IndiaNew Delhi, India
| | - Umesh K Shandilya
- Animal Biotechnology Division, National Bureau of Animal Genetic ResourcesKarnal, India
| | - Tejpal Dhewa
- Department of Nutrition Biology, Central University of HaryanaMahendergarh, India
| | - Nikhil Kumar
- Department of Life Sciences, Shri Venkateshwara UniversityJP Nagar, India
| | - Sanjeev Kumar
- Department of Life Science, Central Assam UniversitySilchar, India
| | - Anil K Puniya
- College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences UniversityLudhiana, India.,Dairy Microbiology Division, ICAR-National Dairy Research InstituteKarnal, India
| | - Pratyoosh Shukla
- Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand UniversityRohtak, India
| |
Collapse
|
118
|
Xue L, He J, Gao N, Lu X, Li M, Wu X, Liu Z, Jin Y, Liu J, Xu J, Geng Y. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci Rep 2017; 7:45176. [PMID: 28349964 PMCID: PMC5368635 DOI: 10.1038/srep45176] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Gut-derived bacterial lipopolysaccharide (LPS) and subsequent hepatic toll-like receptor 4 (TLR4) activation have been recognized to be involved in the onset of diet-induced nonalcoholic fatty liver disease (NAFLD), but little is known about the variation of LPS and TLR4 during the progression of NAFLD. Probiotics were able to inhibit proliferation of harmful bacteria and improve gastrointestinal barrier function. However, it's unclear whether LPS/TLR4 is involved in the protection effect of probiotics on NAFLD. In this study, we described characteristic of gut microbiota structure in the progression of NAFLD, and we also analyzed the relationship between gut microbiota and LPS/TLR4 in this process. Furthermore, we applied probiotics intervention to investigate the effect of probiotics on gut flora structure, intestinal integrity, serum LPS, liver TLR4 and liver pathology. Our results showed that serum LPS and liver TLR4 were highly increased during progression of NAFLD, with gut flora diversity and gut mircobiological colonization resistance (B/E) declining. Furthermore, probiotics could improve gut microbiota structure and liver pathology. Probiotics could also downregulate serum LPS and liver TLR4. Our results suggested that both gut flora alteration and endotoxemia may be involved in the progression of NAFLD. Probiotics may delay the progression of NAFLD via LPS/TLR4 signaling.
Collapse
Affiliation(s)
- Li Xue
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Juntao He
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ning Gao
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaolan Lu
- Department of Gastroenterology, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ming Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaokang Wu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zeshi Liu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Yaofeng Jin
- Department of Pathology, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiali Liu
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Jiru Xu
- Department of Immunology and Pathogenic Biology, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yan Geng
- Department of Laboratory, The Second Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
119
|
Del Chierico F, Nobili V, Vernocchi P, Russo A, De Stefanis C, Gnani D, Furlanello C, Zandonà A, Paci P, Capuani G, Dallapiccola B, Miccheli A, Alisi A, Putignani L. Gut microbiota profiling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology 2017; 65:451-464. [PMID: 27028797 DOI: 10.1002/hep.28572] [Citation(s) in RCA: 514] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
Abstract
UNLABELLED There is evidence that nonalcoholic fatty liver disease (NAFLD) is affected by gut microbiota. Therefore, we investigated its modifications in pediatric NAFLD patients using targeted metagenomics and metabolomics. Stools were collected from 61 consecutive patients diagnosed with nonalcoholic fatty liver (NAFL), nonalcoholic steatohepatitis (NASH), or obesity and 54 healthy controls (CTRLs), matched in a case-control fashion. Operational taxonomic units were pyrosequenced targeting 16S ribosomal RNA and volatile organic compounds determined by solid-phase microextraction gas chromatography-mass spectrometry. The α-diversity was highest in CTRLs, followed by obese, NASH, and NAFL patients; and β-diversity distinguished between patients and CTRLs but not NAFL and NASH. Compared to CTRLs, in NAFLD patients Actinobacteria were significantly increased and Bacteroidetes reduced. There were no significant differences among the NAFL, NASH, and obese groups. Overall NAFLD patients had increased levels of Bradyrhizobium, Anaerococcus, Peptoniphilus, Propionibacterium acnes, Dorea, and Ruminococcus and reduced proportions of Oscillospira and Rikenellaceae compared to CTRLs. After reducing metagenomics and metabolomics data dimensionality, multivariate analyses indicated a decrease of Oscillospira in NAFL and NASH groups and increases of Ruminococcus, Blautia, and Dorea in NASH patients compared to CTRLs. Of the 292 volatile organic compounds, 26 were up-regulated and 2 down-regulated in NAFLD patients. Multivariate analyses found that combination of Oscillospira, Rickenellaceae, Parabacteroides, Bacteroides fragilis, Sutterella, Lachnospiraceae, 4-methyl-2-pentanone, 1-butanol, and 2-butanone could discriminate NAFLD patients from CTRLs. Univariate analyses found significantly lower levels of Oscillospira and higher levels of 1-pentanol and 2-butanone in NAFL patients compared to CTRLs. In NASH, lower levels of Oscillospira were associated with higher abundance of Dorea and Ruminococcus and higher levels of 2-butanone and 4-methyl-2-pentanone compared to CTRLs. CONCLUSION An Oscillospira decrease coupled to a 2-butanone up-regulation and increases in Ruminococcus and Dorea were identified as gut microbiota signatures of NAFL onset and NAFL-NASH progression, respectively. (Hepatology 2017;65:451-464).
Collapse
Affiliation(s)
| | - Valerio Nobili
- Hepato-Metabolic Disease Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy.,Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - Pamela Vernocchi
- Human Microbiome Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - Alessandra Russo
- Human Microbiome Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | | | - Daniela Gnani
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - Cesare Furlanello
- Predictive Models for Biomedicine and Environment Unit, Fondazione Bruno Kessler, Trento, Italy
| | - Alessandro Zandonà
- Predictive Models for Biomedicine and Environment Unit, Fondazione Bruno Kessler, Trento, Italy
| | - Paola Paci
- Institute for Systems Analysis and Computer Science "Antonio Ruberti", National Research Council, 00185, Rome, Italy.,SysBio Centre for Systems Biology, 00185, Rome, Italy
| | | | - Bruno Dallapiccola
- Scientific Directorate, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | | | - Anna Alisi
- Liver Research Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy.,Parasitology Unit, "Bambino Gesù" Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
120
|
Porras D, Nistal E, Martínez-Flórez S, Pisonero-Vaquero S, Olcoz JL, Jover R, González-Gallego J, García-Mediavilla MV, Sánchez-Campos S. Protective effect of quercetin on high-fat diet-induced non-alcoholic fatty liver disease in mice is mediated by modulating intestinal microbiota imbalance and related gut-liver axis activation. Free Radic Biol Med 2017; 102:188-202. [PMID: 27890642 DOI: 10.1016/j.freeradbiomed.2016.11.037] [Citation(s) in RCA: 357] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 11/18/2016] [Accepted: 11/23/2016] [Indexed: 02/08/2023]
Abstract
Gut microbiota is involved in obesity, metabolic syndrome and the progression of nonalcoholic fatty liver disease (NAFLD). It has been recently suggested that the flavonoid quercetin may have the ability to modulate the intestinal microbiota composition, suggesting a prebiotic capacity which highlights a great therapeutic potential in NAFLD. The present study aims to investigate benefits of experimental treatment with quercetin on gut microbial balance and related gut-liver axis activation in a nutritional animal model of NAFLD associated to obesity. C57BL/6J mice were challenged with high fat diet (HFD) supplemented or not with quercetin for 16 weeks. HFD induced obesity, metabolic syndrome and the development of hepatic steatosis as main hepatic histological finding. Increased accumulation of intrahepatic lipids was associated with altered gene expression related to lipid metabolism, as a result of deregulation of their major modulators. Quercetin supplementation decreased insulin resistance and NAFLD activity score, by reducing the intrahepatic lipid accumulation through its ability to modulate lipid metabolism gene expression, cytochrome P450 2E1 (CYP2E1)-dependent lipoperoxidation and related lipotoxicity. Microbiota composition was determined via 16S ribosomal RNA Illumina next-generation sequencing. Metagenomic studies revealed HFD-dependent differences at phylum, class and genus levels leading to dysbiosis, characterized by an increase in Firmicutes/Bacteroidetes ratio and in Gram-negative bacteria, and a dramatically increased detection of Helicobacter genus. Dysbiosis was accompanied by endotoxemia, intestinal barrier dysfunction and gut-liver axis alteration and subsequent inflammatory gene overexpression. Dysbiosis-mediated toll-like receptor 4 (TLR-4)-NF-κB signaling pathway activation was associated with inflammasome initiation response and reticulum stress pathway induction. Quercetin reverted gut microbiota imbalance and related endotoxemia-mediated TLR-4 pathway induction, with subsequent inhibition of inflammasome response and reticulum stress pathway activation, leading to the blockage of lipid metabolism gene expression deregulation. Our results support the suitability of quercetin as a therapeutic approach for obesity-associated NAFLD via its anti-inflammatory, antioxidant and prebiotic integrative response.
Collapse
Affiliation(s)
- David Porras
- Institute of Biomedicine (IBIOMED), University of León, León, Spain.
| | - Esther Nistal
- Institute of Biomedicine (IBIOMED), University of León, León, Spain.
| | | | | | - José Luis Olcoz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Department of Gastroenterology, Complejo Asistencial Universitario de León, León, Spain.
| | - Ramiro Jover
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Experimental Hepatology Unit, IIS Hospital La Fe, Valencia, Spain; Department of Biochemistry and Molecular Biology, University of Valencia, Valencia, Spain.
| | - Javier González-Gallego
- Institute of Biomedicine (IBIOMED), University of León, León, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| | - María Victoria García-Mediavilla
- Institute of Biomedicine (IBIOMED), University of León, León, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| | - Sonia Sánchez-Campos
- Institute of Biomedicine (IBIOMED), University of León, León, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
121
|
Alwahsh SM, Gebhardt R. Dietary fructose as a risk factor for non-alcoholic fatty liver disease (NAFLD). Arch Toxicol 2016; 91:1545-1563. [PMID: 27995280 DOI: 10.1007/s00204-016-1892-7] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 11/08/2016] [Indexed: 12/16/2022]
Abstract
Glucose is a major energy source for the entire body, while fructose metabolism occurs mainly in the liver. Fructose consumption has increased over the last decade globally and is suspected to contribute to the increased incidence of non-alcoholic fatty liver disease (NAFLD). NAFLD is a manifestation of metabolic syndrome affecting about one-third of the population worldwide and has progressive pathological potential for liver cirrhosis and cancer through non-alcoholic steatohepatitis (NASH). Here we have reviewed the possible contribution of fructose to the pathophysiology of NAFLD. We critically summarize the current findings about several regulators, and their potential mechanisms, that have been studied in humans and animal models in response to fructose exposure. A novel hypothesis on fructose-dependent perturbation of liver regeneration and metabolism is advanced. Fructose intake could affect inflammatory and metabolic processes, liver function, gut microbiota, and portal endotoxin influx. The role of the brain in controlling fructose ingestion and the subsequent development of NAFLD is highlighted. Although the importance for fructose (over)consumption for NAFLD in humans is still debated and comprehensive intervention studies are invited, understanding of how fructose intake can favor these pathological processes is crucial for the development of appropriate noninvasive diagnostic and therapeutic approaches to detect and treat these metabolic effects. Still, lifestyle modification, to lessen the consumption of fructose-containing products, and physical exercise are major measures against NAFLD. Finally, promising drugs against fructose-induced insulin resistance and hepatic dysfunction that are emerging from studies in rodents are reviewed, but need further validation in human patients.
Collapse
Affiliation(s)
- Salamah Mohammad Alwahsh
- Faculty of Medicine, Institute of Biochemistry, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany. .,MCR Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Dr, EH16 4UU Edinburgh, UK.
| | - Rolf Gebhardt
- Faculty of Medicine, Institute of Biochemistry, University of Leipzig, Johannisallee 30, 04103, Leipzig, Germany.
| |
Collapse
|
122
|
Abstract
Objective: This review aimed to summarize the relationship between intestinal microbiota metabolism and cardiovascular disease (CVD) and to propose a novel CVD therapeutic target. Data Sources: This study was based on data obtained from PubMed and EMBASE up to June 30, 2015. Articles were selected using the following search terms: “Intestinal microbiota”, “trimethylamine N-oxide (TMAO)”, “trimethylamine (TMA)”, “cardiovascular”, and “atherosclerosis”. Study Selection: Studies were eligible if they present information on intestinal microbiota metabolism and atherosclerosis. Studies on TMA-containing nutrients were also included. Results: A new CVD risk factor, TMAO, was recently identified. It has been observed that several TMA-containing compounds may be catabolized by specific intestinal microbiota, resulting in TMA release. TMA is subsequently converted to TMAO in the liver. Several preliminary studies have linked TMAO to CVD, particularly atherosclerosis; however, the details of this relationship remain unclear. Conclusions: Intestinal microbiota metabolism is associated with atherosclerosis and may represent a promising therapeutic target with respect to CVD management.
Collapse
Affiliation(s)
| | | | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
123
|
Kaska L, Sledzinski T, Chomiczewska A, Dettlaff-Pokora A, Swierczynski J. Improved glucose metabolism following bariatric surgery is associated with increased circulating bile acid concentrations and remodeling of the gut microbiome. World J Gastroenterol 2016; 22:8698-8719. [PMID: 27818587 PMCID: PMC5075546 DOI: 10.3748/wjg.v22.i39.8698] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/23/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
Clinical studies have indicated that circulating bile acid (BA) concentrations increase following bariatric surgery, especially following malabsorptive procedures such as Roux-en-Y gastric bypasses (RYGB). Moreover, total circulating BA concentrations in patients following RYGB are positively correlated with serum glucagon-like peptide-1 concentrations and inversely correlated with postprandial glucose concentrations. Overall, these data suggest that the increased circulating BA concentrations following bariatric surgery - independently of calorie restriction and body-weight loss - could contribute, at least in part, to improvements in insulin sensitivity, incretin hormone secretion, and postprandial glycemia, leading to the remission of type-2 diabetes (T2DM). In humans, the primary and secondary BA pool size is dependent on the rate of biosynthesis and the enterohepatic circulation of BAs, as well as on the gut microbiota, which play a crucial role in BA biotransformation. Moreover, BAs and gut microbiota are closely integrated and affect each other. Thus, the alterations in bile flow that result from anatomical changes caused by bariatric surgery and changes in gut microbiome may influence circulating BA concentrations and could subsequently contribute to T2DM remission following RYGB. Research data coming largely from animal and cell culture models suggest that BAs can contribute, via nuclear farnezoid X receptor (FXR) and membrane G-protein-receptor (TGR-5), to beneficial effects on glucose metabolism. It is therefore likely that FXR, TGR-5, and BAs play a similar role in glucose metabolism following bariatric surgery in humans. The objective of this review is to discuss in detail the results of published studies that show how bariatric surgery affects glucose metabolism and subsequently T2DM remission.
Collapse
|
124
|
Abstract
Low testosterone levels increase the risk for cardiovascular disease in men and lead to shorter life spans. Our recent study showed that androgen deprivation via castration altered fecal microbiota and exacerbated risk factors for cardiovascular disease, including obesity, impaired fasting glucose, excess hepatic triglyceride accumulation, and thigh muscle weight loss only in high-fat diet (HFD)-fed male mice. However, when mice were administered antibiotics that disrupted the gut microbiota, castration did not increase cardiovascular risks or decrease the ratio of dried feces to food intake. Here, we show that changes in cecal microbiota (e.g., an increased Firmicutes/Bacteroidetes ratio and number of Lactobacillus species) were consistent with changes in feces and that there was a decreased cecal content secondary to castration in HFD mice. Castration increased rectal body temperature and plasma adiponectin, irrespective of diet. Changes in the gut microbiome may provide novel insight into hypogonadism-induced cardiovascular diseases.
Collapse
Affiliation(s)
- Naoki Harada
- Division of Applied Life Sciences, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University,
Sakai, Osaka, Japan,CONTACT Naoki Harada, Ph.D. ,
Division of Applied Life Sciences, Graduate School of Life and
Environmental Sciences, Osaka Prefecture University, 1-1
Gakuen-cho, Naka-ku, Sakai Osaka 599-8531,
Japan
| | - Ryo Hanaoka
- Division of Applied Life Sciences, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University,
Sakai, Osaka, Japan
| | - Kazuki Hanada
- Division of Applied Life Sciences, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University,
Sakai, Osaka, Japan
| | - Takeshi Izawa
- Division of Veterinary Science, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University,
Izumisano, Osaka, Japan
| | - Hiroshi Inui
- Division of Clinical Nutrition, Graduate
School of Comprehensive Rehabilitation, Osaka Prefecture University,
Habikino, Osaka, Japan
| | - Ryoichi Yamaji
- Division of Applied Life Sciences, Graduate
School of Life and Environmental Sciences, Osaka Prefecture University,
Sakai, Osaka, Japan
| |
Collapse
|
125
|
Aly AM, Adel A, El-Gendy AO, Essam TM, Aziz RK. Gut microbiome alterations in patients with stage 4 hepatitis C. Gut Pathog 2016; 8:42. [PMID: 27625705 PMCID: PMC5020480 DOI: 10.1186/s13099-016-0124-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023] Open
Abstract
Background Hepatitis C virus (HCV) causes debilitating liver diseases, which may progress to cirrhosis and cancer, and claims 500,000 annual lives worldwide. While HCV epidemiology, pathophysiology, and therapy are being deeply studied, rare attention is given to reciprocal interactions between HCV infection , HCV-induced chronic liver diseases, and the human gut microbiome. As Egypt has the world’s highest prevalence of HCV infections, we launched this study to monitor differences in the gut microbial community composition of Egyptian HCV patients that may affect, or result from, the patients’ liver state. Results To this end, we analyzed stool samples from six stage 4-HCV patients and eight healthy individuals by high-throughput 16S rRNA gene sequencing using Illumina MiSeq. Overall, the alpha-diversity of the healthy persons’ gut microbiomes was higher than those of the HCV patients. Whereas members of phylum Bacteroidetes were more abundant in HCV patients, healthy individuals had higher abundance of Firmicutes, Proteobacteria, and Actinobacteria. Genus-level analysis showed differential abundance of Prevotella and Faecalibacterium (higher in HCV patients) vs. Ruminococcus and Clostridium (healthy group), indicating that the higher abundance of Bacteroidetes in HCV patients is most likely due to Prevotella overabundance. The probiotic genus, Bifidobacterium, was only observed in the microbiotas of healthy individuals. Conclusions To the best of our knowledge, this study provides a first overview of major phyla and genera differentiating stage 4-HCV patients from healthy individuals and suggests possible microbiome remodeling in chronic hepatitis C, possibly shaped by bacterial translocation as well as the liver’s impaired role in digestion and protein synthesis. Future studies will investigate the microbiome composition and functional capabilities in more patients while tracing some potential biomarker taxa (e.g., Prevotella, Faecalibacterium vs. Bifidobacterium). Electronic supplementary material The online version of this article (doi:10.1186/s13099-016-0124-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- AbdelRahman Mahmoud Aly
- Faculty of Post Graduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511 Egypt
| | - AbdelReheem Adel
- Faculty of Post Graduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, 62511 Egypt
| | - Ahmed Osama El-Gendy
- Department of Microbiology and Immunology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62511 Egypt
| | - Tamer M Essam
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562 Egypt
| | - Ramy K Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562 Egypt
| |
Collapse
|
126
|
Abstract
New-onset diabetes is a frequent complication after solid organ transplantation. Although a number of common factors are associated with the disease, including recipient age, body mass index, hepatitis C infection, and use of immunosuppressive drugs, new-onset diabetes after liver transplantation (NODALT) has the following unique aspects and thus needs to be considered its own entity. First, a liver graft becomes the patient's primary metabolic regulator after liver transplantation, but this would not be the case for kidney or other grafts. The metabolic states, as well as the genetics of the graft, play crucial roles in the development of NODALT. Second, dysfunction of the islets of Langerhans is common in cirrhotic patients and would be exacerbated by immunosuppressive agents, particularly calcineurin inhibitors. On the other hand, minimized immunosuppressive protocols have been widely advocated in liver transplantation because of liver tolerance (immune privilege). Third and last, through the "gut-liver axis," graft function is closely linked to gut microbiota, which is now considered an important metabolic organ and known to independently influence the host's metabolic homeostasis. Liver transplant recipients present with specific gut microbiota that may be prone to trigger metabolic disorders. In this review, we proposed 3 possible sites for the origin of NODALT, which are liver, islets, and gut, to help elucidate the underlying mechanism of NODALT.
Collapse
|
127
|
Aron-Wisnewsky J, Clement K, Pépin JL. Nonalcoholic fatty liver disease and obstructive sleep apnea. Metabolism 2016; 65:1124-35. [PMID: 27324067 DOI: 10.1016/j.metabol.2016.05.004] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/03/2016] [Accepted: 05/07/2016] [Indexed: 12/18/2022]
Abstract
Obstructive sleep apnea (OSA) and more importantly its hallmark, chronic intermittent hypoxia (CIH), are established factors in the pathogenesis and exacerbation of nonalcoholic fatty liver disease (NAFLD). This has been clearly demonstrated in rodent models exposed to intermittent hypoxia, and strong evidence now also exists in both paediatric and adult human populations. OSA and CIH induce insulin-resistance and dyslipidemia which are involved in NAFLD physiopathogenesis. CIH increases the expression of the hypoxia inducible transcription factor HIF1α and that of downstream genes involved in lipogenesis, thereby increasing β-oxidation and consequently exacerbating liver oxidative stress. OSA also disrupts the gut liver axis, increasing intestinal permeability and with a possible role of gut microbiota in the link between OSA and NAFLD. OSA patients should be screened for NAFLD and vice versa those with NAFLD for OSA. To date there is no evidence that treating OSA with continuous positive airway pressure (CPAP) will improve NAFLD but it might at least stabilize and slow its progression. Nevertheless, these multimorbid patients should be efficiently treated for all their metabolic co-morbidities and be encouraged to follow weight stabilization or weight loss programs and physical activity life style interventions.
Collapse
Affiliation(s)
- Judith Aron-Wisnewsky
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR_S U1166, Nutriomics, 75013 Paris, France; INSERM, UMR_S U1166, Nutriomics, 75013 Paris, France.
| | - Karine Clement
- Institute of Cardiometabolism and Nutrition, ICAN, Assistance Pitié-Salpêtrière Hospital, Paris, France; Sorbonne Universités, Université Pierre et Marie Curie-Paris 6, UMR_S U1166, Nutriomics, 75013 Paris, France; INSERM, UMR_S U1166, Nutriomics, 75013 Paris, France
| | - Jean-Louis Pépin
- Institut National de la Santé et de la Recherche Médicale (INSERM), U 1042, HP2 Laboratory (Hypoxia: Pathophysiology), Grenoble Alpes Univ., Grenoble, F-38000, France;; Grenoble Alpes University Hospital, Pole Thorax et Vaisseaux, F-38000, France.
| |
Collapse
|
128
|
Vespasiani-Gentilucci U, Gallo P, Porcari A, Carotti S, Galati G, Piccioni L, De Vincentis A, Dell'Unto C, Vorini F, Morini S, Riva E, Picardi A. The PNPLA3 rs738409 C > G polymorphism is associated with the risk of progression to cirrhosis in NAFLD patients. Scand J Gastroenterol 2016; 51:967-73. [PMID: 27150500 DOI: 10.3109/00365521.2016.1161066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS The patatin-like phospholipase domain-containing 3 (PNPLA3) rs738409 C > G single nucleotide polymorphism (SNP) has been associated with steatosis and fibrosis in previous NAFLD populations in which cirrhotic patients were very poorly represented. Since not all NAFLD with fibrosis evolve to cirrhosis, we investigated the specific risk of cirrhosis conferred in NAFLD patients by carrying this SNP. METHODS Three groups were studied: patients with NASH-cirrhosis; patients with biopsy-proven non-cirrhotic NAFLD; healthy subjects undergoing medicine check-ups. Epidemiological, anthropometric, and clinical data were collected, and the SNP was analyzed by pyrosequencing. RESULTS Sixty-one patients with NASH-cirrhosis, 60 with non-cirrhotic NAFLD, and 125 healthy controls were included. Frequency of the PNPLA3 minor (G) allele was increased in patients with NASH-cirrhosis compared with non-cirrhotic NAFLD and controls (allele frequency: 0.598 versus 0.367 versus 0.2, respectively, p < 0.001), and different between the latter two groups (p < 0.001). Three-quarters (74%) of NASH cirrhotics carried at least one G allele, and almost half of them (46%) were GG homozygous. By multivariate analysis in the NAFLD population, each copy of the G allele was associated with an almost doubling of the risk of cirrhosis [OR 1.8 (1.02-3.2)], while being GG homozygous with a tripled risk compared with being CC homozygous [3.01 (1.03-10.8)]. CONCLUSIONS In NAFLD patients, carriage of the PNPLA3G allele, and particularly of the GG genotype, is significantly associated with the risk of cirrhotic evolution. If confirmed in larger series, these results would suggest that most of NASH cases require the contribution of an altered PNPLA3 function to progress until cirrhosis.
Collapse
Affiliation(s)
| | - Paolo Gallo
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Aldostefano Porcari
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Simone Carotti
- b Laboratory of Microscopic and Ultrastructural Anatomy, CIR , University Campus Bio-Medico , Rome , Italy
| | - Giovanni Galati
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Livia Piccioni
- c Virology Unit , University Campus Bio-Medico , Rome , Italy
| | - Antonio De Vincentis
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Chiara Dell'Unto
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Ferruccio Vorini
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| | - Sergio Morini
- b Laboratory of Microscopic and Ultrastructural Anatomy, CIR , University Campus Bio-Medico , Rome , Italy
| | - Elisabetta Riva
- c Virology Unit , University Campus Bio-Medico , Rome , Italy
| | - Antonio Picardi
- a Internal Medicine and Hepatology Unit , University Campus Bio-Medico , Rome , Italy
| |
Collapse
|
129
|
Matsushita N, Osaka T, Haruta I, Ueshiba H, Yanagisawa N, Omori-Miyake M, Hashimoto E, Shibata N, Tokushige K, Saito K, Tsuneda S, Yagi J. Effect of Lipopolysaccharide on the Progression of Non-Alcoholic Fatty Liver Disease in High Caloric Diet-Fed Mice. Scand J Immunol 2016; 83:109-18. [PMID: 26524607 DOI: 10.1111/sji.12397] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/22/2015] [Indexed: 12/17/2022]
Abstract
The incidence of non-alcoholic steatohepatitis (NASH) is increasing. Because gut microbiota have been highlighted as one of the key factors in the pathogenesis of metabolic syndrome, we investigated the involvement of the bacterial component in the progression of non-alcoholic fatty liver (NAFL) to NASH. C57BL/6 mice were fed with maintenance food (MF, groups A and B) or a high caloric diet (HCD, groups C and D) for 1 month. Mice were then divided into four groups: Groups A and C were inoculated with PBS, while groups B and D were inoculated with lipopolysaccharide (LPS) plus complete Freund's adjuvant (CFA). The inoculations were performed a total of 3 times over 3 months. At 6 months, while hepatic steatosis was observed in groups C and D, cellular infiltration and fibrosis were less evident in group C than in group D. Inflammatory cytokines were upregulated in groups B and D. 16S rRNA pyrosequencing of whole colon homogenates containing faeces showed that certain bacterial groups, such as Bacteroidaceae, Peptostreptococcaceae and Erysipelotrichaceae, were increased in groups C and D. Although loading of bacterial components (LPS) resulted in hepatic inflammation in both MF- and HCD-fed mice, HCD feeding was more crucial in the progression of NAFL during the triggering phase.
Collapse
Affiliation(s)
- N Matsushita
- Support Center for Women Health Care Professionals and Researchers, Tokyo Women's Medical University, Tokyo, Japan.,Department of Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - T Osaka
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| | - I Haruta
- Department of Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan.,Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| | - H Ueshiba
- Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - N Yanagisawa
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| | - M Omori-Miyake
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| | - E Hashimoto
- Department of Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - N Shibata
- Department of Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - K Tokushige
- Department of Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - K Saito
- Support Center for Women Health Care Professionals and Researchers, Tokyo Women's Medical University, Tokyo, Japan
| | - S Tsuneda
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - J Yagi
- Department of Microbiology and Immunology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
130
|
Hepatocellular Carcinoma in Obesity, Type 2 Diabetes, and NAFLD. Dig Dis Sci 2016; 61:1234-45. [PMID: 26921078 DOI: 10.1007/s10620-016-4085-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the second commonest cause of cancer death worldwide. Rather than falling as a result of prevention and treatments for viral hepatitis, an increase is evident in developed nations consequent to the rising prevalence of obesity and type 2 diabetes mellitus (T2DM)-the two major risk factors for nonalcoholic fatty liver disease (NAFLD). The majority of patients with HCC complicating these conditions present with advanced disease as the tools for surveillance are inadequate, and the "at-risk" population is not well characterized. This review will summarize the epidemiological evidence linking obesity, T2DM, and NAFLD with HCC, what is known about the pathogenic mechanisms involved, as well as their relevance for clinicians managing patients at risk. There will also be an overview of the "unmet needs" surrounding this topic, with suggestions for the direction translational research should take in order to prevent progression of NAFLD to HCC, to improve early detection of HCC in those with NAFLD, as well as to improve outcomes for those affected.
Collapse
|
131
|
Chen J, Xie P, Lin J, He J, Zeng C, Chen J. Effects of microcystin-LR on gut microflora in different gut regions of mice. J Toxicol Sci 2016; 40:485-94. [PMID: 26165645 DOI: 10.2131/jts.40.485] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To reveal the toxicological effects of the hepatotoxic microcystin-leucine arginine (MC-LR) on gut microbial community composition in different gut regions, we conducted a subchronic exposure of BALB/c mice to MC-LR via intragastric administration. Denaturing gradient gel electrophoresis (DGGE) was employed to profile the shifts of microbes after MC-LR treatment in the jejuno-ileum, caecum and colon. DGGE profiles analysis showed that MC-LR increased the microbial species richness (number of microbial bands) in the caecum and colon as well as microbial diversity (Shannon-Wiener index) in the caecum. The cluster analysis of DGGE profiles indicated that the microbial structures in the caecum and colon shifted significantly after MC-LR treatment, while that in the jejuno-ileum did not. All the relatively decreased gut microbes belonged to Clostridia in the Firmicutes phylum, and most of them were Lachnospiraceae. The increased ones derived from a variety of microbes including species from Porphyromonadaceae and Prevotellaceae in the Bacteroidetes phylum, as well as Lachnospiraceae and Ruminococcaceae in the Firmicutes phylum, and among which, the increase of Barnesiella in Porphyromonadaceae was most remarkable. In conclusion, subchronic exposure to MC-LR could disturb the balance of gut microbes in mice, and its toxicological effects varied between the jejuno-ileum and the other two gut regions.
Collapse
Affiliation(s)
- Jing Chen
- Donghu Experimental Station of Lake Ecosystems, State Key Laboratory of Freshwater Ecology and Biotechnology of China, Institute of Hydrobiology, Chinese Academy of Sciences, China
| | | | | | | | | | | |
Collapse
|
132
|
NAFLD-Associated Hepatocellular Carcinoma: a Threat to Patients with Metabolic Disorders. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s11901-016-0297-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
133
|
Gut Microbiota and Lifestyle Interventions in NAFLD. Int J Mol Sci 2016; 17:447. [PMID: 27023533 PMCID: PMC4848903 DOI: 10.3390/ijms17040447] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 02/07/2023] Open
Abstract
The human digestive system harbors a diverse and complex community of microorganisms that work in a symbiotic fashion with the host, contributing to metabolism, immune response and intestinal architecture. However, disruption of a stable and diverse community, termed "dysbiosis", has been shown to have a profound impact upon health and disease. Emerging data demonstrate dysbiosis of the gut microbiota to be linked with non-alcoholic fatty liver disease (NAFLD). Although the exact mechanism(s) remain unknown, inflammation, damage to the intestinal membrane, and translocation of bacteria have all been suggested. Lifestyle intervention is undoubtedly effective at improving NAFLD, however, not all patients respond to these in the same manner. Furthermore, studies investigating the effects of lifestyle interventions on the gut microbiota in NAFLD patients are lacking. A deeper understanding of how different aspects of lifestyle (diet/nutrition/exercise) affect the host-microbiome interaction may allow for a more tailored approach to lifestyle intervention. With gut microbiota representing a key element of personalized medicine and nutrition, we review the effects of lifestyle interventions (diet and physical activity/exercise) on gut microbiota and how this impacts upon NAFLD prognosis.
Collapse
|
134
|
Kim B, Park KY, Ji Y, Park S, Holzapfel W, Hyun CK. Protective effects of Lactobacillus rhamnosus GG against dyslipidemia in high-fat diet-induced obese mice. Biochem Biophys Res Commun 2016; 473:530-6. [PMID: 27018382 DOI: 10.1016/j.bbrc.2016.03.107] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 12/20/2022]
Abstract
Recent reports suggest that gut microbiota can be a major determinant of dyslipidemia and non-alcoholic fatty liver disease (NAFLD) and its modulation by treating probiotics is a valid strategy to exert a protective effect. In this study, high-fat diet (HFD)-fed mice were orally administrated with Lactobacillus rhamnosus GG (LGG) for 13 weeks. Significant reductions in the weights of the liver, mesenteric and subcutaneous adipose tissues were observed in LGG-treated HFD-fed mice compared to LGG-non-treated controls. The serum levels of triglyceride and cholesterol were also significantly reduced in LGG-treated mice. Gut microbial composition analysis showed that shifts in the diversity of dominant gut bacteria were caused by HFD and restored by LGG treatment. A remarkable decrease of hepatic fat content was also observed in LGG-treated mice, accompanied by downregulated expressions of lipogenic and pro-inflammatory genes in the liver. LGG-treated mice had lower expression levels of genes involved in cholesterol synthesis, but conversely, higher expression levels of cholesterol efflux-related genes compared to LGG-non-treated controls. The cholesterol-lowering effect of LGG was also found to be mediated by suppression of FXR and FGF15 signaling, resulting in the upregulation of hepatic CYP7A1. Our findings confirm a therapeutic potential of probiotics for ameliorating dyslipidemia and NAFLD.
Collapse
Affiliation(s)
- Bobae Kim
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Kun-Young Park
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Yosep Ji
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Soyoung Park
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Wilhelm Holzapfel
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea
| | - Chang-Kee Hyun
- School of Life Science, Handong Global University, Pohang, Gyungbuk 791-708, Republic of Korea.
| |
Collapse
|
135
|
He X, Ji G, Jia W, Li H. Gut Microbiota and Nonalcoholic Fatty Liver Disease: Insights on Mechanism and Application of Metabolomics. Int J Mol Sci 2016; 17:300. [PMID: 26999104 PMCID: PMC4813164 DOI: 10.3390/ijms17030300] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota are intricately involved in the development of obesity-related metabolic diseases such as nonalcoholic fatty liver disease (NAFLD), type 2 diabetes, and insulin resistance. In the current review, we discuss the role of gut microbiota in the development of NAFLD by focusing on the mechanisms of gut microbiota-mediated host energy metabolism, insulin resistance, regulation of bile acids and choline metabolism, as well as gut microbiota-targeted therapy. We also discuss the application of a metabolomic approach to characterize gut microbial metabotypes in NAFLD.
Collapse
Affiliation(s)
- Xuyun He
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Wei Jia
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
- Center for Translational Medicine, and Shanghai Key Laboratory of Diabetes Mellitus, Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Houkai Li
- Center for Chinese Medical Therapy and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
136
|
Khan MJ, Gerasimidis K, Edwards CA, Shaikh MG. Role of Gut Microbiota in the Aetiology of Obesity: Proposed Mechanisms and Review of the Literature. J Obes 2016; 2016:7353642. [PMID: 27703805 PMCID: PMC5040794 DOI: 10.1155/2016/7353642] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/21/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023] Open
Abstract
The aetiology of obesity has been attributed to several factors (environmental, dietary, lifestyle, host, and genetic factors); however none of these fully explain the increase in the prevalence of obesity worldwide. Gut microbiota located at the interface of host and environment in the gut are a new area of research being explored to explain the excess accumulation of energy in obese individuals and may be a potential target for therapeutic manipulation to reduce host energy storage. Several mechanisms have been suggested to explain the role of gut microbiota in the aetiology of obesity such as short chain fatty acid production, stimulation of hormones, chronic low-grade inflammation, lipoprotein and bile acid metabolism, and increased endocannabinoid receptor system tone. However, evidence from animal and human studies clearly indicates controversies in determining the cause or effect relationship between the gut microbiota and obesity. Metagenomics based studies indicate that functionality rather than the composition of gut microbiota may be important. Further mechanistic studies controlling for environmental and epigenetic factors are therefore required to help unravel obesity pathogenesis.
Collapse
Affiliation(s)
- Muhammad Jaffar Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Phase V Hayatabad, Peshawar, Khyber Pakhtunkhwa, Pakistan
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
- *Muhammad Jaffar Khan:
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| | - Christine Ann Edwards
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical Veterinary and Life Sciences, University of Glasgow, Level 3, New Lister Building, Glasgow Royal Infirmary, 10-16 Alexandra Parade, Glasgow G31 2ER, UK
| | - M. Guftar Shaikh
- Department of Endocrinology, Royal Hospital for Children, 1345 Govan Rd, Govan, Glasgow G51 4TF, UK
| |
Collapse
|
137
|
Wieland A, Frank DN, Harnke B, Bambha K. Systematic review: microbial dysbiosis and nonalcoholic fatty liver disease. Aliment Pharmacol Ther 2015; 42:1051-63. [PMID: 26304302 DOI: 10.1111/apt.13376] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/11/2015] [Accepted: 07/29/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND The human intestinal microbiota is a key regulator of host metabolic and immune functions and alterations in the microbiome ('dysbiosis') have been implicated in several human diseases. Because of the anatomical links between the intestines and the liver, dysbiosis may also disrupt hepatic function and thereby contribute to the pathogenesis of nonalcoholic fatty liver disease (NAFLD). AIM To perform a comprehensive review of the medical literature investigating associations between intestinal dysbiosis and NAFLD, with a particular emphasis on studies that characterise the microbiome in NAFLD. METHODS We conducted a search of PubMed, Embase, and Web of Science using multiple search terms including: 'NAFLD, NASH, fatty liver, steatohepatitis' combined with 'metagenome, microbiom*, microbiota*, fecal flora, intestinal flora, gut bacteria'. Results were manually reviewed and studies selected based on relevance to intestinal microbiota and NAFLD. We also included studies that addressed potential mechanistic models of pathways linking the dysbiosis to NAFLD. RESULTS Nine studies (five human and four animal models) were identified in our search that assessed associations between specific intestinal microbiota composition and NAFLD. We reviewed and summarised the results of additional investigations that more broadly addressed the mechanisms by which the microbiome may impact NAFLD pathogenesis. CONCLUSIONS Investigations in humans and animals demonstrate associations between intestinal dysbiosis and NAFLD; however, causality has not been proven and mechanistic links require further delineation. As the field of microbiome research matures in techniques and study design, more detailed insights into NAFLD pathogenesis and its associations with the intestinal microbiota will be elucidated.
Collapse
Affiliation(s)
- A Wieland
- Division of Gastroenterology and Hepatology, University of Colorado Denver, Aurora, CO, USA
| | - D N Frank
- Division of Infectious Diseases and Microbiome Research Consortium, University of Colorado Denver, Aurora, CO, USA
| | - B Harnke
- University of Colorado Health Science Library, Aurora, CO, USA
| | - K Bambha
- Division of Gastroenterology and Hepatology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
138
|
Gut Microbiota and Host Reaction in Liver Diseases. Microorganisms 2015; 3:759-91. [PMID: 27682116 PMCID: PMC5023261 DOI: 10.3390/microorganisms3040759] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/08/2015] [Accepted: 10/21/2015] [Indexed: 02/07/2023] Open
Abstract
Although alcohol feeding produces evident intestinal microbial changes in animals, only some alcoholics show evident intestinal dysbiosis, a decrease in Bacteroidetes and an increase in Proteobacteria. Gut dysbiosis is related to intestinal hyperpermeability and endotoxemia in alcoholic patients. Alcoholics further exhibit reduced numbers of the beneficial Lactobacillus and Bifidobacterium. Large amounts of endotoxins translocated from the gut strongly activate Toll-like receptor 4 in the liver and play an important role in the progression of alcoholic liver disease (ALD), especially in severe alcoholic liver injury. Gut microbiota and bacterial endotoxins are further involved in some of the mechanisms of nonalcoholic fatty liver disease (NAFLD) and its progression to nonalcoholic steatohepatitis (NASH). There is experimental evidence that a high-fat diet causes characteristic dysbiosis of NAFLD, with a decrease in Bacteroidetes and increases in Firmicutes and Proteobacteria, and gut dysbiosis itself can induce hepatic steatosis and metabolic syndrome. Clinical data support the above dysbiosis, but the details are variable. Intestinal dysbiosis and endotoxemia greatly affect the cirrhotics in relation to major complications and prognosis. Metagenomic approaches to dysbiosis may be promising for the analysis of deranged host metabolism in NASH and cirrhosis. Management of dysbiosis may become a cornerstone for the future treatment of liver diseases.
Collapse
|
139
|
Fotschki B, Jurgoński A, Juśkiewicz J, Zduńczyk Z. Dietary Supplementation with Raspberry Seed Oil Modulates Liver Functions, Inflammatory State, and Lipid Metabolism in Rats. J Nutr 2015; 145:1793-9. [PMID: 26108544 DOI: 10.3945/jn.115.212407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/29/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Although raspberry seed oil (RO) is rich in essential fatty acids, there is a lack of experiments assessing benefits of its consumption. OBJECTIVE We investigated the effects of dietary supplementation with RO on healthy rats and rats with low-grade systemic inflammation, liver disorders, and dyslipidemia induced by a high-fat/low-fiber (HF/LF) diet. METHODS Thirty-two rats were allocated into 4 groups of 8 rats each and fed for 8 wk a control (C; 7% lard and 5% cellulose) or HF/LF (21% lard and 2% cellulose) diet or modifications of these diets in which 7% RO replaced all (C+RO group) or a proportion of (HF/LF+RO group) the lard. Effects of diet and RO and their interaction on bacterial activity and metabolite formations in the distal intestine, liver fat and glutathione concentration, plasma lipid profile, transaminase activities, and plasma concentrations of C-reactive protein (CRP) and tumor necrosis factor α (TNF-α) were tested. RESULTS Dietary RO decreased plasma alanine and aspartate transaminase activities (43.4 and 157 vs. 25.6 and 115 U/L, respectively; P < 0.05 and P < 0.005) and plasma TNF-α and triglyceride concentrations (132 pg/mL and 2.07 mmol/L vs. 86.5 pg/mL and 0.99 mmol/L, respectively; P < 0.05). In livers of the C+RO group, the fat concentration was decreased, whereas the glutathione to glutathione disulfide ratio was increased compared with the C group (30.1% and 6.20 μmol/g vs. 23.3% and 7.25 μmol/g, respectively; P ≤ 0.05); however, those differences were not observed between the HF/LF groups (P-interaction < 0.05). In the HF/LF+RO group, the plasma CRP concentration was lower than in the HF/LF group (88.1 vs. 765 pg/mL; P ≤ 0.05) and similar to that in the C and C+RO groups (158 and 128 pg/mL, respectively). CONCLUSION Dietary RO improves plasma lipid profile and liver functions and reduces low-grade systemic inflammation in rats; however, the extent of these beneficial effects is partly dependent on the diet type.
Collapse
Affiliation(s)
- Bartosz Fotschki
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Adam Jurgoński
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Jerzy Juśkiewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Zenon Zduńczyk
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| |
Collapse
|
140
|
Frank DN, Bales ES, Monks J, Jackman MJ, MacLean PS, Ir D, Robertson CE, Orlicky DJ, McManaman JL. Perilipin-2 Modulates Lipid Absorption and Microbiome Responses in the Mouse Intestine. PLoS One 2015; 10:e0131944. [PMID: 26147095 PMCID: PMC4493139 DOI: 10.1371/journal.pone.0131944] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/08/2015] [Indexed: 02/06/2023] Open
Abstract
Obesity and its co-morbidities, such as fatty liver disease, are increasingly prevalent worldwide health problems. Intestinal microorganisms have emerged as critical factors linking diet to host physiology and metabolic function, particularly in the context of lipid homeostasis. We previously demonstrated that deletion of the cytoplasmic lipid drop (CLD) protein Perilipin-2 (Plin2) in mice largely abrogates long-term deleterious effects of a high fat (HF) diet. Here we test the hypotheses that Plin2 function impacts the earliest steps of HF diet-mediated pathogenesis as well as the dynamics of diet-associated changes in gut microbiome diversity and function. WT and perilipin-2 null mice raised on a standard chow diet were randomized to either low fat (LF) or HF diets. After four days, animals were assessed for changes in physiological (body weight, energy balance, and fecal triglyceride levels), histochemical (enterocyte CLD content), and fecal microbiome parameters. Plin2-null mice had significantly lower respiratory exchange ratios, diminished frequencies of enterocyte CLDs, and increased fecal triglyceride levels compared with WT mice. Microbiome analyses, employing both 16S rRNA profiling and metagenomic deep sequencing, indicated that dietary fat content and Plin2 genotype were significantly and independently associated with gut microbiome composition, diversity, and functional differences. These data demonstrate that Plin2 modulates rapid effects of diet on fecal lipid levels, enterocyte CLD contents, and fuel utilization properties of mice that correlate with structural and functional differences in their gut microbial communities. Collectively, the data provide evidence of Plin2 regulated intestinal lipid uptake, which contributes to rapid changes in the gut microbial communities implicated in diet-induced obesity.
Collapse
Affiliation(s)
- Daniel N. Frank
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Microbiome Research Consortium, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Elise S. Bales
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jenifer Monks
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Matthew J. Jackman
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Paul S. MacLean
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Diana Ir
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Microbiome Research Consortium, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - James L. McManaman
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
141
|
|
142
|
The Influence of the Gut Microbiome on Obesity, Metabolic Syndrome and Gastrointestinal Disease. Clin Transl Gastroenterol 2015; 6:e91. [PMID: 26087059 PMCID: PMC4816244 DOI: 10.1038/ctg.2015.16] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/13/2015] [Indexed: 12/11/2022] Open
Abstract
There is a fine balance in the mutual relationship between the intestinal microbiota and its mammalian host. It is thought that disruptions in this fine balance contribute/account for the pathogenesis of many diseases. Recently, the significance of the relationship between gut microbiota and its mammalian host in the pathogenesis of obesity and the metabolic syndrome has been demonstrated. Emerging data has linked intestinal dysbiosis to several gastrointestinal diseases including inflammatory bowel disease, irritable bowel syndrome, nonalcoholic fatty liver disease, and gastrointestinal malignancy. This article is intended to review the role of gut microbiota maintenance/alterations of gut microbiota as a significant factor as a significant factor discriminating between health and common diseases. Based on current available data, the role of microbial manipulation in disease management remains to be further defined and a focus for further clinical investigation.
Collapse
|
143
|
Aron-Wisnewsky J, Pepin JL. New insights in the pathophysiology of chronic intermittent hypoxia-induced NASH: the role of gut–liver axis impairment. Thorax 2015; 70:713-5. [DOI: 10.1136/thoraxjnl-2015-207212] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
144
|
Spacek LA, Mudalel ML, Lewicki R, Tittel FK, Risby TH, Stoltzfus J, Munier JJ, Solga SF. Breath ammonia and ethanol increase in response to a high protein challenge. Biomarkers 2015; 20:149-56. [PMID: 26043432 DOI: 10.3109/1354750x.2015.1040840] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Quantifying changes in ammonia and ethanol in blood and body fluid assays in response to food is cumbersome. We used breath analysis of ammonia, ethanol, hydrogen (an accepted standard of gut transit) and acetone to investigate gastrointestinal physiology. In 30 healthy participants, we measured each metabolite serially over 6 h in control and high protein trials. Two-way repeated measures ANOVA compared treatment (control versus intervention), change from baseline to maximum and interaction of treatment and time change. Interaction was significant for ammonia (p < 0.0001) and hydrogen (p < 0.0001). We describe the dynamic measurement of multiple metabolites in response to an oral challenge.
Collapse
Affiliation(s)
- Lisa A Spacek
- Department of Medicine, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| | | | | | | | | | | | | | | |
Collapse
|
145
|
The effects of gastrointestinal surgery on gut microbiota: potential contribution to improved insulin sensitivity. Curr Atheroscler Rep 2015; 16:454. [PMID: 25214424 DOI: 10.1007/s11883-014-0454-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bariatric surgery induces weight loss and major improvement in insulin-resistance through many mechanisms some of which are weight independent. It is now well acknowledged that gut microbiota is involved in the development of obesity and its related metabolic diseases, at least in mice. However, its causal role in human obesity progression remains to be demonstrated. Few studies now pointed at changes in microbiota composition after bariatric surgery, suggesting links between gut microbiota switch and metabolic improvement observed after surgery. As such new potential mechanisms of actions have been proposed. The aim of this review is to describe microbiota modifications observed after bariatric surgery and its potential relationships with improved insulin resistance. We here list some hypotheses, which will need further demonstration.
Collapse
|
146
|
PATHOGENESIS OF HEPATOCELLULAR CARCINOMA DEVELOPMENT IN NON ALCOHOLIC FATTY LIVER DISEASE. ACTA ACUST UNITED AC 2015; 14:119-127. [PMID: 26114083 DOI: 10.1007/s11901-015-0260-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is being recognized as an increasingly important contributor to the burden of hepatocellular carcinoma (HCC) worldwide. It is often accompanied by obesity and diabetes mellitus and is believed to be the hepatic representation of the metabolic syndrome. HCC development in NAFLD is multifactorial and complex. It is dependent on not only the well-described mechanisms noted in chronic liver injury, but also on the molecular derangements associated with obesity and dysmetabolism. These include adipocyte remodeling, adipokine secretion, lipotoxicity and insulin resistance. Recent advances focus on the importance of the gut-liver axis in accelerating the process of oncogenesis in NAFLD. The farnesoid X nuclear receptor (FXR) has been demonstrated to have important metabolic effects and its pharmacological activation by obeticholic acid has been recently reported to produce histological improvement in NASH. It is hoped that delineating the mechanisms of hepatic fibrosis and oncogenesis in NASH will lead to enhanced strategies for cancer prevention, surveillance and therapy in this population.
Collapse
|
147
|
Lonardo A, Ballestri S, Marchesini G, Angulo P, Loria P. Nonalcoholic fatty liver disease: a precursor of the metabolic syndrome. Dig Liver Dis 2015; 47:181-90. [PMID: 25739820 DOI: 10.1016/j.dld.2014.09.020] [Citation(s) in RCA: 496] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/07/2023]
Abstract
The conventional paradigm of nonalcoholic fatty liver disease representing the "hepatic manifestation of the metabolic syndrome" is outdated. We identified and summarized longitudinal studies that, supporting the association of nonalcoholic fatty liver disease with either type 2 diabetes mellitus or metabolic syndrome, suggest that nonalcoholic fatty liver disease precedes the development of both conditions. Online Medical databases were searched, relevant articles were identified, their references were further assessed and tabulated data were checked. Although several cross-sectional studies linked nonalcoholic fatty liver disease to either diabetes and other components of the metabolic syndrome, we focused on 28 longitudinal studies which provided evidence for nonalcoholic fatty liver disease as a risk factor for the future development of diabetes. Moreover, additional 19 longitudinal reported that nonalcoholic fatty liver disease precedes and is a risk factor for the future development of the metabolic syndrome. Finally, molecular and genetic studies are discussed supporting the view that aetiology of steatosis and lipid intra-hepatocytic compartmentation are a major determinant of whether fatty liver is/is not associated with insulin resistance and metabolic syndrome. Data support the novel paradigm of nonalcoholic fatty liver disease as a strong determinant for the development of the metabolic syndrome, which has potentially relevant clinical implications for diagnosing, preventing and treating metabolic syndrome.
Collapse
Affiliation(s)
- Amedeo Lonardo
- AUSL Modena and University of Modena and Reggio Emilia, Department of Biomedical, Metabolic and Neural Sciences, Division of Internal Medicine, NOCSAE - Baggiovara, Modena, Italy.
| | - Stefano Ballestri
- AUSL Modena, Department of Internal Medicine, Division of Internal Medicine, Hospital of Pavullo, Pavullo nel Frignano, Italy
| | - Giulio Marchesini
- "Alma Mater Studiorum" University, Unit of Metabolic Diseases and Clinical Dietetics, Bologna, Italy
| | - Paul Angulo
- University of Kentucky, Division of Digestive Diseases & Nutrition, Section of Hepatology, Medical Center, Lexington, KY, USA
| | - Paola Loria
- AUSL Modena and University of Modena and Reggio Emilia, Department of Biomedical, Metabolic and Neural Sciences, Division of Internal Medicine, NOCSAE - Baggiovara, Modena, Italy
| |
Collapse
|
148
|
Abstract
PURPOSE OF REVIEW This review focuses on the latest understanding of the molecular mechanisms underlying the complex interactions between intestine and liver bile acid signaling, gut microbiota, and their impact on whole-body lipid, glucose and energy metabolism. RECENT FINDINGS Hepatic bile acid synthesis is tightly regulated by the bile acid negative feedback mechanisms. Modulating the enterohepatic bile acid signaling greatly impacts the whole-body metabolic homeostasis. Recently, a positive feedback mechanism through intestine farnesoid X receptor (FXR) antagonism has been proposed to link gut microbiota to the regulation of bile acid composition and pool size. Two studies identified intestine Diet1 and hepatic SHP-2 as novel regulators of CYP7A1 and bile acid synthesis through the gut-liver FXR-fibroblast growth factor 15/19-FGF receptor four signaling axis. New evidence suggests that enhancing bile acid signaling in the distal ileum and colon contributes to the metabolic benefits of bile acid sequestrants and bariatric surgery. SUMMARY Small-molecule ligands that target TGR5 and FXR have shown promise in treating various metabolic and inflammation-related human diseases. New insights into the mechanisms underlying the bariatric surgery and bile acid sequestrant treatment suggest that targeting the enterohepatic circulation to modulate gut-liver bile acid signaling, incretin production and microbiota represents a new strategy to treat obesity and type 2 diabetes.
Collapse
|
149
|
Flass T, Tong S, Frank DN, Wagner BD, Robertson CE, Kotter CV, Sokol RJ, Zemanick E, Accurso F, Hoffenberg EJ, Narkewicz MR. Intestinal lesions are associated with altered intestinal microbiome and are more frequent in children and young adults with cystic fibrosis and cirrhosis. PLoS One 2015; 10:e0116967. [PMID: 25658710 PMCID: PMC4319904 DOI: 10.1371/journal.pone.0116967] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/17/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Cirrhosis (CIR) occurs in 5-7% of cystic fibrosis (CF) patients. We hypothesized that alterations in intestinal function in CF contribute to the development of CIR. AIMS Determine the frequency of macroscopic intestinal lesions, intestinal inflammation, intestinal permeability and characterize fecal microbiome in CF CIR subjects and CF subjects with no liver disease (CFnoLIV). METHODS 11 subjects with CFCIR (6 M, 12.8 yrs ± 3.8) and 19 matched with CFnoLIV (10 M, 12.6 yrs ± 3.4) underwent small bowel capsule endoscopy, intestinal permeability testing by urinary lactulose: mannitol excretion ratio, fecal calprotectin determination and fecal microbiome characterization. RESULTS CFCIR and CFnoLIV did not differ in key demographics or CF complications. CFCIR had higher GGT (59±51 U/L vs 17±4 p = 0.02) and lower platelet count (187±126 vs 283±60 p = 0.04) and weight (-0.86 ± 1.0 vs 0.30 ± 0.9 p = 0.002) z scores. CFCIR had more severe intestinal mucosal lesions on capsule endoscopy (score ≥4, 4/11 vs 0/19 p = 0.01). Fecal calprotectin was similar between CFCIR and CFnoLIV (166 μg/g ±175 vs 136 ± 193 p = 0.58, nl <120). Lactulose:mannitol ratio was elevated in 27/28 subjects and was slightly lower in CFCIR vs CFnoLIV (0.08±0.02 vs 0.11±0.05, p = 0.04, nl ≤0.03). Small bowel transit time was longer in CFCIR vs CFnoLIV (195±42 min vs 167±68 p<0.001, nl 274 ± 41). Bacteroides were decreased in relative abundance in CFCIR and were associated with lower capsule endoscopy score whereas Clostridium were more abundant in CFCIR and associated with higher capsule endoscopy score. CONCLUSIONS CFCIR is associated with increased intestinal mucosal lesions, slower small bowel transit time and alterations in fecal microbiome. Abnormal intestinal permeability and elevated fecal calprotectin are common in all CF subjects. Disturbances in intestinal function in CF combined with changes in the microbiome may contribute to the development of hepatic fibrosis and intestinal lesions.
Collapse
Affiliation(s)
- Thomas Flass
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Suhong Tong
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital, Aurora, Colorado, United States of America
| | - Daniel N. Frank
- Department of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Brandie D. Wagner
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital, Aurora, Colorado, United States of America
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, United States of America
| | - Charles E. Robertson
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Cassandra Vogel Kotter
- Department of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Ronald J. Sokol
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
- Colorado Clinical and Translational Sciences Institute, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Edith Zemanick
- Section of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Frank Accurso
- Section of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Edward J. Hoffenberg
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Michael R. Narkewicz
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| |
Collapse
|
150
|
Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. MICROBIAL ECOLOGY IN HEALTH AND DISEASE 2015; 26:26191. [PMID: 25651997 PMCID: PMC4315779 DOI: 10.3402/mehd.v26.26191] [Citation(s) in RCA: 770] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is growing evidence that dysbiosis of the gut microbiota is associated with the pathogenesis of both intestinal and extra-intestinal disorders. Intestinal disorders include inflammatory bowel disease, irritable bowel syndrome (IBS), and coeliac disease, while extra-intestinal disorders include allergy, asthma, metabolic syndrome, cardiovascular disease, and obesity. In many of these conditions, the mechanisms leading to disease development involves the pivotal mutualistic relationship between the colonic microbiota, their metabolic products, and the host immune system. The establishment of a ‘healthy’ relationship early in life appears to be critical to maintaining intestinal homeostasis. Whilst we do not yet have a clear understanding of what constitutes a ‘healthy’ colonic microbiota, a picture is emerging from many recent studies identifying particular bacterial species associated with a healthy microbiota. In particular, the bacterial species residing within the mucus layer of the colon, either through direct contact with host cells, or through indirect communication via bacterial metabolites, may influence whether host cellular homeostasis is maintained or whether inflammatory mechanisms are triggered. In addition to inflammation, there is some evidence that perturbations in the gut microbiota is involved with the development of colorectal cancer. In this case, dysbiosis may not be the most important factor, rather the products of interaction between diet and the microbiome. High-protein diets are thought to result in the production of carcinogenic metabolites from the colonic microbiota that may result in the induction of neoplasia in the colonic epithelium. Ever more sensitive metabolomics methodologies reveal a suite of small molecules produced in the microbiome which mimic or act as neurosignallers or neurotransmitters. Coupled with evidence that probiotic interventions may alter psychological endpoints in both humans and in rodent models, these data suggest that CNS-related co-morbidities frequently associated with GI disease may originate in the intestine as a result of microbial dysbiosis. This review outlines the current evidence showing the extent to which the gut microbiota contributes to the development of disease. Based on evidence to date, we can assess the potential to positively modulate the composition of the colonic microbiota and ameliorate disease activity through bacterial intervention.
Collapse
Affiliation(s)
- Simon Carding
- Institute of Food Research, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | - Kristin Verbeke
- Translational Research in GastroIntestinal Disorders, KU Leuven, Leuven, Belgium
| | - Daniel T Vipond
- Institute of Food Research, Norwich, UK.,Norwich Medical School, University of East Anglia, Norwich, UK
| | - Bernard M Corfe
- Molecular Gastroenterology Research Group, Department of Oncology, University of Sheffield, Sheffield, UK.,Insigneo Institute for in silico Medicine, University of Sheffield, Sheffield, UK;
| | - Lauren J Owen
- Human Nutrition Unit, Department of Oncology, University of Sheffield, Sheffield, UK
| |
Collapse
|