351
|
Bluemel S, Williams B, Knight R, Schnabl B. Precision medicine in alcoholic and nonalcoholic fatty liver disease via modulating the gut microbiota. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1018-G1036. [PMID: 27686615 PMCID: PMC5206291 DOI: 10.1152/ajpgi.00245.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/25/2016] [Indexed: 02/08/2023]
Abstract
Alcoholic liver disease (ALD) and nonalcoholic fatty liver disease (NAFLD) represent a major health burden in industrialized countries. Although alcohol abuse and nutrition play a central role in disease pathogenesis, preclinical models support a contribution of the gut microbiota to ALD and NAFLD. This review describes changes in the intestinal microbiota compositions related to ALD and NAFLD. Findings from in vitro, animal, and human studies are used to explain how intestinal pathology contributes to disease progression. This review summarizes the effects of untargeted microbiome modifications using antibiotics and probiotics on liver disease in animals and humans. While both affect humoral inflammation, regression of advanced liver disease or mortality has not been demonstrated. This review further describes products secreted by Lactobacillus- and microbiota-derived metabolites, such as fatty acids and antioxidants, that could be used for precision medicine in the treatment of liver disease. A better understanding of host-microbial interactions is allowing discovery of novel therapeutic targets in the gut microbiota, enabling new treatment options that restore the intestinal ecosystem precisely and influence liver disease. The modulation options of the gut microbiota and precision medicine employing the gut microbiota presented in this review have excellent prospects to improve treatment of liver disease.
Collapse
Affiliation(s)
- Sena Bluemel
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Brandon Williams
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Rob Knight
- Departments of Pediatrics and Computer Science and Engineering, University of California San Diego, La Jolla, California; and
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California;
- Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
352
|
Wang L, Fouts DE, Stärkel P, Hartmann P, Chen P, Llorente C, DePew J, Moncera K, Ho SB, Brenner DA, Hooper LV, Schnabl B. Intestinal REG3 Lectins Protect against Alcoholic Steatohepatitis by Reducing Mucosa-Associated Microbiota and Preventing Bacterial Translocation. Cell Host Microbe 2016; 19:227-39. [PMID: 26867181 DOI: 10.1016/j.chom.2016.01.003] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/14/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Approximately half of all deaths from liver cirrhosis, the tenth leading cause of mortality in the United States, are related to alcohol use. Chronic alcohol consumption is accompanied by intestinal dysbiosis and bacterial overgrowth, yet little is known about the factors that alter the microbial composition or their contribution to liver disease. We previously associated chronic alcohol consumption with lower intestinal levels of the antimicrobial-regenerating islet-derived (REG)-3 lectins. Here, we demonstrate that intestinal deficiency in REG3B or REG3G increases numbers of mucosa-associated bacteria and enhances bacterial translocation to the mesenteric lymph nodes and liver, promoting the progression of ethanol-induced fatty liver disease toward steatohepatitis. Overexpression of Reg3g in intestinal epithelial cells restricts bacterial colonization of mucosal surfaces, reduces bacterial translocation, and protects mice from alcohol-induced steatohepatitis. Thus, alcohol appears to impair control of the mucosa-associated microbiota, and subsequent breach of the mucosal barrier facilitates progression of alcoholic liver disease.
Collapse
Affiliation(s)
- Lirui Wang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Peter Stärkel
- St. Luc University Hospital, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Phillipp Hartmann
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Cristina Llorente
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | | | - Samuel B Ho
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | - David A Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Lora V Hooper
- Howard Hughes Medical Institute; Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
353
|
Voigt RM, Summa KC, Forsyth CB, Green SJ, Engen P, Naqib A, Vitaterna MH, Turek FW, Keshavarzian A. The Circadian Clock Mutation Promotes Intestinal Dysbiosis. Alcohol Clin Exp Res 2016; 40:335-47. [PMID: 26842252 DOI: 10.1111/acer.12943] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/24/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Circadian rhythm disruption is a prevalent feature of modern day society that is associated with an increase in pro-inflammatory diseases, and there is a clear need for a better understanding of the mechanism(s) underlying this phenomenon. We have previously demonstrated that both environmental and genetic circadian rhythm disruption causes intestinal hyperpermeability and exacerbates alcohol-induced intestinal hyperpermeability and liver pathology. The intestinal microbiota can influence intestinal barrier integrity and impact immune system function; thus, in this study, we sought to determine whether genetic alteration of the core circadian clock gene, Clock, altered the intestinal microbiota community. METHODS Male Clock(Δ19) -mutant mice (mice homozygous for a dominant-negative-mutant allele) or littermate wild-type mice were fed 1 of 3 experimental diets: (i) a standard chow diet, (ii) an alcohol-containing diet, or (iii) an alcohol-control diet in which the alcohol calories were replaced with dextrose. Stool microbiota was assessed with 16S ribosomal RNA gene amplicon sequencing. RESULTS The fecal microbial community of Clock-mutant mice had lower taxonomic diversity, relative to wild-type mice, and the Clock(Δ19) mutation was associated with intestinal dysbiosis when mice were fed either the alcohol-containing or the control diet. We found that alcohol consumption significantly altered the intestinal microbiota in both wild-type and Clock-mutant mice. CONCLUSIONS Our data support a model by which circadian rhythm disruption by the Clock(Δ19) mutation perturbs normal intestinal microbial communities, and this trend was exacerbated in the context of a secondary dietary intestinal stressor.
Collapse
Affiliation(s)
- Robin M Voigt
- Department of Internal Medicine , Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Keith C Summa
- Center for Sleep and Circadian Biology , Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Christopher B Forsyth
- Department of Internal Medicine , Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois.,Department of Biochemistry , Rush University Medical Center, Chicago, Illinois
| | - Stefan J Green
- DNA Services Facility , Research Resources Center, University of Illinois at Chicago, Chicago, Illinois.,Department of Biological Sciences , University of Illinois at Chicago, Chicago, Illinois
| | - Phillip Engen
- Department of Internal Medicine , Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois
| | - Ankur Naqib
- DNA Services Facility , Research Resources Center, University of Illinois at Chicago, Chicago, Illinois
| | - Martha H Vitaterna
- Center for Sleep and Circadian Biology , Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Fred W Turek
- Center for Sleep and Circadian Biology , Department of Neurobiology, Northwestern University, Evanston, Illinois
| | - Ali Keshavarzian
- Department of Internal Medicine , Division of Gastroenterology, Rush University Medical Center, Chicago, Illinois.,Department of Pharmacology , Rush University Medical Center, Chicago, Illinois.,Division of Pharmacology , Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
354
|
Saturated and Unsaturated Dietary Fats Differentially Modulate Ethanol-Induced Changes in Gut Microbiome and Metabolome in a Mouse Model of Alcoholic Liver Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:765-76. [PMID: 27012191 DOI: 10.1016/j.ajpath.2015.11.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/21/2015] [Accepted: 11/17/2015] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) ranks among major causes of morbidity and mortality. Diet and crosstalk between the gut and liver are important determinants of ALD. We evaluated the effects of different types of dietary fat and ethanol on the gut microbiota composition and metabolic activity and the effect of these changes on liver injury in ALD. Compared with ethanol and a saturated fat diet (medium chain triglycerides enriched), an unsaturated fat diet (corn oil enriched) exacerbated ethanol-induced endotoxemia, liver steatosis, and injury. Major alterations in gut microbiota, including a reduction in Bacteroidetes and an increase in Proteobacteria and Actinobacteria, were seen in animals fed an unsaturated fat diet and ethanol but not a saturated fat diet and ethanol. Compared with a saturated fat diet and ethanol, an unsaturated fat diet and ethanol caused major fecal metabolomic changes. Moreover, a decrease in certain fecal amino acids was noted in both alcohol-fed groups. These data support an important role of dietary lipids in ALD pathogenesis and provide insight into mechanisms of ALD development. A diet enriched in unsaturated fats enhanced alcohol-induced liver injury and caused major fecal metagenomic and metabolomic changes that may play an etiologic role in observed liver injury. Dietary lipids can potentially serve as inexpensive interventions for the prevention and treatment of ALD.
Collapse
|
355
|
Zhang ZL, Duan ZJ. Gut-liver axis: An important target for prevention and treatment of liver diseases. Shijie Huaren Xiaohua Zazhi 2016; 24:3454-3460. [DOI: 10.11569/wcjd.v24.i23.3454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The human intestine harbors a complex and diverse community of microbes that promote metabolism and digestion in their symbiotic relationship with the host. Liver diseases have long been associated with qualitative (dysbiotic) and quantitative (overgrowth) changes in the intestinal microbiota. Extrinsic factors, such as diet and alcohol, contribute to intestinal microbiota dysbiosis. Dysbiosis results in intestinal inflammation, intestinal barrier breakdown, and translocation of microbial products in animal models, further aggravating hepatic injury and inflammation. Microbial metabolites produced in a dysbiotic intestinal environment and host factors are equally important in the pathogenesis of liver diseases. In the current review, we discuss the progress in understanding the role of gut-liver axis dysfunction in the progression of non-alcoholic fatty liver disease, alcoholic liver disease and cirrhosis, and the potential application value of the restoration of intestinal homeostasis in the prevention and treatment of liver diseases.
Collapse
|
356
|
Sung H, Kim SW, Hong M, Suk KT. Microbiota-based treatments in alcoholic liver disease. World J Gastroenterol 2016; 22:6673-6682. [PMID: 27547010 PMCID: PMC4970471 DOI: 10.3748/wjg.v22.i29.6673] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/21/2016] [Accepted: 06/13/2016] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota plays a key role in the pathogenesis of alcoholic liver disease (ALD). Consumption of alcohol leads to increased gut permeability, small intestinal bacterial overgrowth, and enteric dysbiosis. These factors contribute to the increased translocation of microbial products to the liver via the portal tract. Subsequently, bacterial endotoxins such as lipopolysaccharide, in association with the Toll-like receptor 4 signaling pathway, induce a gamut of damaging immune responses in the hepatic milieu. Because of the close association between deleterious inflammation and ALD-induced microbiota imbalance, therapeutic approaches that seek to reestablish gut homeostasis should be considered in the treatment of alcoholic patients. To this end, a number of preliminary studies on probiotics have confirmed their effectiveness in suppressing proinflammatory cytokines and improving liver function in the context of ALD. In addition, there have been few studies linking the administration of prebiotics and antibiotics with reduction of alcohol-induced liver damage. Because these preliminary results are promising, large-scale randomized studies are warranted to elucidate the impact of these microbiota-based treatments on the gut flora and associated immune responses, in addition to exploring questions about optimal delivery. Finally, fecal microbiota transplant has been shown to be an effective method of modulating gut microbiota and deserve further investigation as a potential therapeutic option for ALD.
Collapse
|
357
|
Fukui H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm Intest Dis 2016. [PMID: 29922669 DOI: 10.1159/000447252.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2022] Open
Abstract
Background Increased intestinal permeability due to barrier dysfunction is supposed to cause microbial translocation which may induce low-grade inflammation in various diseases. However, this series of events has not been comprehensively evaluated yet. Summary Intestinal epithelial barrier dysfunction and increased permeability have been described in patients with inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), alcoholic liver disease, nonalcoholic steatohepatitis (NASH), liver cirrhosis, acute pancreatitis, primary biliary cholangitis (PBC), type 1 and type 2 diabetes, chronic kidney disease, chronic heart failure (CHF), depression, and other diseases. Most clinical reports used either permeability assays of challenge tests or measurement of circulating bacterial markers like endotoxin for assessment of 'the leaky gut'. The intestinal permeability assessed by the challenge tests has often been related to the changes of tight junction proteins in the epithelium or circulating endotoxin levels. In patients with IBD, alcoholic liver disease, NASH, liver cirrhosis, PBC, obstructive jaundice, severe acute pancreatitis, and CHF, endotoxemia and proinflammatory cytokinemia have been found in addition to increased permeability. In the serum of patients with IBS and depression, antiflagellin antibodies and antilipid A antibodies were detected, respectively, together with increased permeability and proinflammatory cytokinemia. The site of infection, which is localized to the intestine in IBD and IBS, includes various extraintestinal organs in other diseases. The relation of gut dysbiosis to intestinal barrier dysfunction has gradually been clarified. Key Messages Although no direct cause-and-effect relationship has been confirmed, all clinical and experimental data suggest the importance of intestinal hyperpermeability in the inflammatory changes of various diseases. Increased intestinal permeability is a new target for disease prevention and therapy. Considering the close relationship of 'the leaky gut' and gut dysbiosis to the major diseases, we can conclude that meticulous dietetic and probiotic approaches to recover healthy microbiota have the potential to make a breakthrough in the management of these diseases tomorrow.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Kashihara, Japan
| |
Collapse
|
358
|
Fukui H. Increased Intestinal Permeability and Decreased Barrier Function: Does It Really Influence the Risk of Inflammation? Inflamm Intest Dis 2016; 1:135-145. [PMID: 29922669 DOI: 10.1159/000447252] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/30/2016] [Indexed: 12/13/2022] Open
Abstract
Background Increased intestinal permeability due to barrier dysfunction is supposed to cause microbial translocation which may induce low-grade inflammation in various diseases. However, this series of events has not been comprehensively evaluated yet. Summary Intestinal epithelial barrier dysfunction and increased permeability have been described in patients with inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), alcoholic liver disease, nonalcoholic steatohepatitis (NASH), liver cirrhosis, acute pancreatitis, primary biliary cholangitis (PBC), type 1 and type 2 diabetes, chronic kidney disease, chronic heart failure (CHF), depression, and other diseases. Most clinical reports used either permeability assays of challenge tests or measurement of circulating bacterial markers like endotoxin for assessment of 'the leaky gut'. The intestinal permeability assessed by the challenge tests has often been related to the changes of tight junction proteins in the epithelium or circulating endotoxin levels. In patients with IBD, alcoholic liver disease, NASH, liver cirrhosis, PBC, obstructive jaundice, severe acute pancreatitis, and CHF, endotoxemia and proinflammatory cytokinemia have been found in addition to increased permeability. In the serum of patients with IBS and depression, antiflagellin antibodies and antilipid A antibodies were detected, respectively, together with increased permeability and proinflammatory cytokinemia. The site of infection, which is localized to the intestine in IBD and IBS, includes various extraintestinal organs in other diseases. The relation of gut dysbiosis to intestinal barrier dysfunction has gradually been clarified. Key Messages Although no direct cause-and-effect relationship has been confirmed, all clinical and experimental data suggest the importance of intestinal hyperpermeability in the inflammatory changes of various diseases. Increased intestinal permeability is a new target for disease prevention and therapy. Considering the close relationship of 'the leaky gut' and gut dysbiosis to the major diseases, we can conclude that meticulous dietetic and probiotic approaches to recover healthy microbiota have the potential to make a breakthrough in the management of these diseases tomorrow.
Collapse
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, Kashihara, Japan
| |
Collapse
|
359
|
Tsuruya A, Kuwahara A, Saito Y, Yamaguchi H, Tsubo T, Suga S, Inai M, Aoki Y, Takahashi S, Tsutsumi E, Suwa Y, Morita H, Kinoshita K, Totsuka Y, Suda W, Oshima K, Hattori M, Mizukami T, Yokoyama A, Shimoyama T, Nakayama T. Ecophysiological consequences of alcoholism on human gut microbiota: implications for ethanol-related pathogenesis of colon cancer. Sci Rep 2016; 6:27923. [PMID: 27295340 PMCID: PMC4904738 DOI: 10.1038/srep27923] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 05/26/2016] [Indexed: 01/09/2023] Open
Abstract
Chronic consumption of excess ethanol increases the risk of colorectal cancer. The pathogenesis of ethanol-related colorectal cancer (ER-CRC) is thought to be partly mediated by gut microbes. Specifically, bacteria in the colon and rectum convert ethanol to acetaldehyde (AcH), which is carcinogenic. However, the effects of chronic ethanol consumption on the human gut microbiome are poorly understood, and the role of gut microbes in the proposed AcH-mediated pathogenesis of ER-CRC remains to be elaborated. Here we analyse and compare the gut microbiota structures of non-alcoholics and alcoholics. The gut microbiotas of alcoholics were diminished in dominant obligate anaerobes (e.g., Bacteroides and Ruminococcus) and enriched in Streptococcus and other minor species. This alteration might be exacerbated by habitual smoking. These observations could at least partly be explained by the susceptibility of obligate anaerobes to reactive oxygen species, which are increased by chronic exposure of the gut mucosa to ethanol. The AcH productivity from ethanol was much lower in the faeces of alcoholic patients than in faeces of non-alcoholic subjects. The faecal phenotype of the alcoholics could be rationalised based on their gut microbiota structures and the ability of gut bacteria to accumulate AcH from ethanol.
Collapse
Affiliation(s)
- Atsuki Tsuruya
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Akika Kuwahara
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Yuta Saito
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Haruhiko Yamaguchi
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Takahisa Tsubo
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Shogo Suga
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Makoto Inai
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Yuichi Aoki
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Seiji Takahashi
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Eri Tsutsumi
- Suntory World Research Center, Suntory Holdings Ltd., Soraku-gun, Kyoto 619-0284, Japan
| | - Yoshihide Suwa
- Suntory World Research Center, Suntory Holdings Ltd., Soraku-gun, Kyoto 619-0284, Japan
| | - Hidetoshi Morita
- Graduate School of Environmental and Life Science, Okayama University, 1-1-1 Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Kenji Kinoshita
- School of Pharmaceutical Sciences, Mukogawa Women's University, Nishinomiya, Hyogo 663-8179, Japan
| | - Yukari Totsuka
- Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Wataru Suda
- Center for Omics and Bioinformatics, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8561, Japan
| | - Kenshiro Oshima
- Center for Omics and Bioinformatics, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8561, Japan
| | - Masahira Hattori
- Center for Omics and Bioinformatics, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba 277-8561, Japan
| | - Takeshi Mizukami
- National Hospital Organization Kurihama Medical and Addiction Center, Yokosuka, Kanagawa 239-0841, Japan
| | - Akira Yokoyama
- National Hospital Organization Kurihama Medical and Addiction Center, Yokosuka, Kanagawa 239-0841, Japan
| | - Takefumi Shimoyama
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| | - Toru Nakayama
- Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, Sendai, Miyagi 980-8579 Japan
| |
Collapse
|
360
|
Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, Wesselingh S. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol Psychiatry 2016; 21:738-48. [PMID: 27090305 PMCID: PMC4879184 DOI: 10.1038/mp.2016.50] [Citation(s) in RCA: 657] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 02/06/2023]
Abstract
The human body hosts an enormous abundance and diversity of microbes, which perform a range of essential and beneficial functions. Our appreciation of the importance of these microbial communities to many aspects of human physiology has grown dramatically in recent years. We know, for example, that animals raised in a germ-free environment exhibit substantially altered immune and metabolic function, while the disruption of commensal microbiota in humans is associated with the development of a growing number of diseases. Evidence is now emerging that, through interactions with the gut-brain axis, the bidirectional communication system between the central nervous system and the gastrointestinal tract, the gut microbiome can also influence neural development, cognition and behaviour, with recent evidence that changes in behaviour alter gut microbiota composition, while modifications of the microbiome can induce depressive-like behaviours. Although an association between enteropathy and certain psychiatric conditions has long been recognized, it now appears that gut microbes represent direct mediators of psychopathology. Here, we examine roles of gut microbiome in shaping brain development and neurological function, and the mechanisms by which it can contribute to mental illness. Further, we discuss how the insight provided by this new and exciting field of research can inform care and provide a basis for the design of novel, microbiota-targeted, therapies.
Collapse
Affiliation(s)
- G B Rogers
- South Australian Health and Medical Research Institute, Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - D J Keating
- South Australian Health and Medical Research Institute, Centre for Neuroscience and Department of Human Physiology, Flinders University, Adelaide, SA, Australia
| | - R L Young
- South Australian Health and Medical Research Institute, Department of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - M-L Wong
- South Australian Health and Medical Research Institute, Mind and Brain Theme, and Flinders University, Adelaide, SA, Australia
| | - J Licinio
- South Australian Health and Medical Research Institute, Mind and Brain Theme, and Flinders University, Adelaide, SA, Australia
| | - S Wesselingh
- South Australian Health and Medical Research Institute, Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
361
|
Betrapally NS, Gillevet PM, Bajaj JS. Changes in the Intestinal Microbiome and Alcoholic and Nonalcoholic Liver Diseases: Causes or Effects? Gastroenterology 2016; 150:1745-1755.e3. [PMID: 26948887 PMCID: PMC5026236 DOI: 10.1053/j.gastro.2016.02.073] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/24/2016] [Accepted: 02/25/2016] [Indexed: 02/07/2023]
Abstract
The prevalence of fatty liver diseases is increasing rapidly worldwide; after treatment of hepatitis C virus infection becomes more widespread, fatty liver diseases are likely to become the most prevalent liver disorders. Although fatty liver diseases are associated with alcohol, obesity, and the metabolic syndrome, their mechanisms of pathogenesis are not clear. The development and progression of fatty liver, alcoholic, and nonalcoholic liver disease (NAFLD) all appear to be influenced by the composition of the microbiota. The intestinal microbiota have been shown to affect precirrhotic and cirrhotic stages of liver diseases, which could lead to new strategies for their diagnosis, treatment, and study. We review differences and similarities in the cirrhotic and precirrhotic stages of NAFLD and alcoholic liver disease. Differences have been observed in these stages of alcohol-associated disease in patients who continue to drink compared with those who stop, with respect to the composition and function of the intestinal microbiota and intestinal integrity. NAFLD and the intestinal microbiota also differ between patients with and without diabetes. We also discuss the potential of microbial therapy for patients with NAFLD and ALD.
Collapse
Affiliation(s)
- Naga S Betrapally
- Microbiome Analysis Center, George Mason University, Manassas, Virginia
| | | | - Jasmohan S Bajaj
- Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, Richmond, Virginia.
| |
Collapse
|
362
|
Yu J, Marsh S, Hu J, Feng W, Wu C. Gut Microbiota and Metagenomic Advancement in Digestive Disease. Gastroenterol Res Pract 2016; 2016:4703406. [PMID: 27247566 PMCID: PMC4877475 DOI: 10.1155/2016/4703406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 04/21/2016] [Indexed: 01/18/2023] Open
Affiliation(s)
- Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sharon Marsh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada T6G 2H7
| | - Junbo Hu
- Department of General Surgery, Tongji Hospital, Huazhong Science & Technology University, Wuhan, Hubei 430030, China
| | - Wenke Feng
- Department of Medicine, University of Louisville, Louisville, KY 40208, USA
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, Houston, TX 77843, USA
| |
Collapse
|
363
|
Hartmann P, Seebauer CT, Schnabl B. Alcoholic liver disease: the gut microbiome and liver cross talk. Alcohol Clin Exp Res 2016; 39:763-75. [PMID: 25872593 DOI: 10.1111/acer.12704] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 02/18/2015] [Indexed: 01/18/2023]
Abstract
Alcoholic liver disease (ALD) is a leading cause of morbidity and mortality worldwide. Alcoholic fatty liver disease can progress to steatohepatitis, alcoholic hepatitis, fibrosis, and cirrhosis. Patients with alcohol abuse show quantitative and qualitative changes in the composition of the intestinal microbiome. Furthermore, patients with ALD have increased intestinal permeability and elevated systemic levels of gut-derived microbial products. Maintaining eubiosis, stabilizing the mucosal gut barrier, or preventing cellular responses to microbial products protect from experimental ALD. Therefore, intestinal dysbiosis and pathological bacterial translocation appear fundamental for the pathogenesis of ALD. This review highlights causes for intestinal dysbiosis and pathological bacterial translocation, their relationship, and consequences for ALD. We also discuss how the liver affects the intestinal microbiota.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Medicine, University of California, San Diego, La Jolla, California
| | | | | |
Collapse
|
364
|
Shalikiani NV, Bakulin IG, Dubinkina VB, Ishchenko DS, Alexeev DG, Tyakht AV, Pavlenko AV, Ilyina EN, Kostryukova ES, Taraskina AE, Skorodumova LO, Maev IV, Govorun VM. [Specific features of the enteric microbiota composition in patients with alcoholic liver cirrhosis]. TERAPEVT ARKH 2016; 87:59-65. [PMID: 26978420 DOI: 10.17116/terarkh2015871259-65] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AIM To establish the specific features of the taxonomic and functional composition of the enteric microbiota in patients with alcoholic liver cirrhosis (LC). SUBJECTS AND METHODS Metagenomic analysis was used to study the taxonomic composition and functional potential of the enteric microbiota in 20 patients with alcoholic LC. Total DNA was isolated from the patients' fecal samples; thereafter full genome sequencing was carried out. The metagenomic analysis yielded the results of the relative taxonomic and functional abundance of microbial species in the test samples. These were comparatively analyzed with the previously published metagenomic datasets of healthy population cohorts in the Russian Federation, as well as in Denmark, China, and the USA. RESULTS In the majority of patients, the dominant part of the intestinal community represented bacterial species constituting the normal human intestinal flora. At the same time, abnormal gut microbiota composition, which was suggestive of marked dysbacteriosis, was identified in a number of patients. In addition, pooled analysis of the data could identify a number of species with a statistically significantly increase and decrease in the relative abundance as compared to the control groups. Thus, the enteric microbiota of the patients with alcoholic LC showed a high proportion of bacteria characteristic of the oral cavity. Analysis of the pooled metabolic potential of the microbiota in these patients demonstrated the higher abundance of enzyme genes involved in alcohol metabolism. CONCLUSION In the patients with alcoholic LC, the microbiota composition changes identified in individual bacterial species may be associated with gastrointestinal comorbidities, such as chronic erosive gastritis, chronic pancreatitis, and gastric ulcer. The alterations occurring in alcoholic cirrhosis promote the penetration and generation of oral cavity-specific microorganisms in the human intestine. This may a potential biomarker for the diagnosis of liver diseases. The bacterial enzyme genes involved in alcohol metabolism have an increased abundance in patients with alcoholic LC and healthy volunteers from the Russian Federation.
Collapse
Affiliation(s)
- N V Shalikiani
- Moscow Clinical Research and Practical Center, Moscow Healthcare Department, Moscow, Russia
| | - I G Bakulin
- Moscow Clinical Research and Practical Center, Moscow Healthcare Department, Moscow, Russia
| | - V B Dubinkina
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia
| | - D S Ishchenko
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - D G Alexeev
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - A V Tyakht
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - A V Pavlenko
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - E N Ilyina
- Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - E S Kostryukova
- Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia; Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia; Kazan Federal University, Kazan, Russia
| | - A E Taraskina
- Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - L O Skorodumova
- Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia
| | - I V Maev
- A.I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russia, Moscow, Russia
| | - V M Govorun
- Research Institute of Physicochemical Medicine, Federal Biomedical Agency of Russia, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudnyi, Russia
| |
Collapse
|
365
|
Dubinkina VB, Tyakht AV, Ilina EN, Ischenko DS, Kovarsky BA, Yarygin KS, Pavlenko AV, Popenko AS, Alexeev DG, Taraskina AE, Nasyrova RF, Krupitski EM, Skorodumova LO, Larin AK, Kostryukova ES, Govorun VM. [Metagenomic analysis of taxonomic and functional changes in gut microbiota of patients with alcoholic dependence syndrome]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2016; 61:742-9. [PMID: 26716747 DOI: 10.18097/pbmc20156106742] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Here we present the first metagenomic study of gut microbiota in patients with alcohol dependence syndrome (ADS) performed in the whole-genome ("shotgun") format. Taxonomic analysis highlighted changes in community "drivers" abundance previously associated with inflammatory processes (including increase in Ruminococcus gnavus and torques, as well as decrease in Faecalibacterium and Akkermansia). Microbiota of alcoholics manifested presence of specific opportunistic pathogens rarely detected in healthy control subjects of the world. Differential analysis of metabolic potential basing on changes in KEGG Orthology groups abundance revealed increase in pathways associated with response to oxidative stress. Analysis of two specific gene groups--alcohol metabolism and virulence factors--also showed increase in comparison with the control groups. We suggest that gut microbiota distinct in alcoholics by both taxonomic and functional composition plays role in modulating the effect of alcohol on host organism.
Collapse
Affiliation(s)
- V B Dubinkina
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - A V Tyakht
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - E N Ilina
- Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - D S Ischenko
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - B A Kovarsky
- Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - K S Yarygin
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia
| | - A V Pavlenko
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - A S Popenko
- Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - D G Alexeev
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - A E Taraskina
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Saint-Petersburg, Russia
| | - R F Nasyrova
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Saint-Petersburg, Russia
| | - E M Krupitski
- Saint-Petersburg Bekhterev Psychoneurological Research Institute, Saint-Petersburg, Russia
| | - L O Skorodumova
- Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - A K Larin
- Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - E S Kostryukova
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| | - V M Govorun
- Moscow Institute of Physics and Technology (State University), Dolgoprudny, Russia; Scientific Research Institute of Physico-Chemical Medicine, Department of Molecular Biology and Genetics, Moscow, Russia
| |
Collapse
|
366
|
Lactobacillus plantarum TWK10 Supplementation Improves Exercise Performance and Increases Muscle Mass in Mice. Nutrients 2016; 8:205. [PMID: 27070637 PMCID: PMC4848674 DOI: 10.3390/nu8040205] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 01/16/2023] Open
Abstract
Lactobacillus plantarum (L. plantarum) is a well-known probiotic among the ingested-microorganism probiotics (i.e., ingested microorganisms associated with beneficial effects for the host). However, few studies have examined the effects of L. plantarum TWK10 (LP10) supplementation on exercise performance, physical fatigue, and gut microbial profile. Male Institute of Cancer Research (ICR) strain mice were divided into three groups (n = 8 per group) for oral administration of LP10 for six weeks at 0, 2.05 × 10⁸, or 1.03 × 10⁹ colony-forming units/kg/day, designated the vehicle, LP10-1X and LP10-5X groups, respectively. LP10 significantly decreased final body weight and increased relative muscle weight (%). LP10 supplementation dose-dependently increased grip strength (p < 0.0001) and endurance swimming time (p < 0.001) and decreased levels of serum lactate (p < 0.0001), ammonia (p < 0.0001), creatine kinase (p = 0.0118), and glucose (p = 0.0151) after acute exercise challenge. The number of type I fibers (slow muscle) in gastrocnemius muscle significantly increased with LP10 treatment. In addition, serum levels of albumin, blood urea nitrogen, creatinine, and triacylglycerol significantly decreased with LP10 treatment. Long-term supplementation with LP10 may increase muscle mass, enhance energy harvesting, and have health-promotion, performance-improvement, and anti-fatigue effects.
Collapse
|
367
|
Wang J, Tang L, Glenn TC, Wang JS. Aflatoxin B1 Induced Compositional Changes in Gut Microbial Communities of Male F344 Rats. Toxicol Sci 2016; 150:54-63. [PMID: 26612839 PMCID: PMC5009611 DOI: 10.1093/toxsci/kfv259] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aflatoxins are a group of potent foodborne toxicants naturally occurring in maize and groundnuts. Differential species-specific sensitivity to aflatoxins has been documented but cannot be fully explained by the differences in metabolism of these toxicants among animal species. Commensal microbial communities (microbiota) are critical to human and animal health, but few studies have assessed interactions between xenobiotic toxins and those microbiota, and its potential effects to humans and animals. Here, an exploratory dosing experiment was conducted to explore effects of Aflatoxin B1 (AFB1) on the gut microbiota in a commonly used rat model. Male F344 rats were randomly divided into groups and treated with different concentrations of AFB1. Microbial communities in fecal samples were assessed using 16S rRNA sequence analysis. We found that samples from the control group had a phylogenetically diverse community, and that increasing AFB1 doses decreased this diversity but increased evenness of community composition. In addition, the gut microbiota from different samples was clustered according to their dosing regimens. There is no community shift at the phylum level but some lactic acid bacteria were significantly depleted by AFB1. These findings suggested that AFB1 could modify the gut microbiota in a dose-dependent manner.
Collapse
Affiliation(s)
- Jincheng Wang
- *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602 *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602
| | - Lili Tang
- *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602
| | - Travis C Glenn
- *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602 *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602
| | - Jia-Sheng Wang
- *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602 *Interdisciplinary Toxicology Program and Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, Georgia 30602
| |
Collapse
|
368
|
Gorky J, Schwaber J. The role of the gut-brain axis in alcohol use disorders. Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:234-41. [PMID: 26188287 PMCID: PMC4679635 DOI: 10.1016/j.pnpbp.2015.06.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 02/08/2023]
Abstract
Neuroimmune and inflammatory processes have been locally associated with the amygdala in alcohol exposure and withdrawal. We and others have suggested that this inflammation in the amygdala may cause disturbance of neural function observed as anxiety and autonomic distress in withdrawal. Despite the potential importance of the robust neuroinflammatory response, the mechanisms contributing to this response are not well understood. We review literature that suggests the effects of alcohol, and other substances of abuse, cause dysbiosis of the gut microbiome. This peripheral response may modulate neuroprotective vagal afferent signaling that permits and exacerbates a neuroinflammatory response in the amygdala. We will examine the mounting evidence that suggests that (1) gut dysbiosis contributes to neuroinflammation, especially in the context of alcohol exposure and withdrawal, (2) the neuroinflammation in the amygdala involves the microglia and astrocytes and their effect on neural cells, and (3) amygdala neuroinflammation itself contributes directly to withdrawal behavior and symptoms. The contribution of the gut to an anxiogenic response is a promising therapeutic target for patients suffering with withdrawal symptoms given the safe and well-established methods of modulating the gut microbiome.
Collapse
Affiliation(s)
- Jonathan Gorky
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - James Schwaber
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
369
|
Abstract
Despite extensive research, alcohol remains one of the most common causes of liver disease in the United States. Alcoholic liver disease (ALD) encompasses a broad spectrum of disorders, including steatosis, steatohepatitis, and cirrhosis. Although many agents and approaches have been tested in patients with ALD and in animals with experimental ALD in the past, there is still no FDA (Food and Drug Administration) approved therapy for any stage of ALD. With the increasing recognition of the importance of gut microbiota in the onset and development of a variety of diseases, the potential use of probiotics in ALD is receiving increasing investigative and clinical attention. In this review, we summarize recent studies on probiotic intervention in the prevention and treatment of ALD in experimental animal models and patients. Potential mechanisms underlying the probiotic function are also discussed.
Collapse
|
370
|
Abstract
The gut microbiome is composed of a vast number of microbes in the gastrointestinal tract, which benefit host metabolism, aid in digestion, and contribute to normal immune function. Alterations in microbial composition can result in intestinal dysbiosis, which has been implicated in several diseases including obesity, inflammatory bowel disease, and liver diseases. Over the past several years, significant interactions between the intestinal microbiota and liver have been discovered, with possible mechanisms for the development as well as progression of liver disease and promising therapeutic targets to either prevent or halt the progression of liver disease. In this review the authors examine mechanisms of dysbiosis-induced liver disease; highlight current knowledge regarding the role of dysbiosis in nonalcoholic liver disease, alcoholic liver disease, and cirrhosis; and discuss potential therapeutic targets.
Collapse
Affiliation(s)
- Gobind Anand
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, California
| | - Amir Zarrinpar
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, California
| | - Rohit Loomba
- Division of Gastroenterology, Department of Medicine, University of California at San Diego, La Jolla, California,NAFLD Translational Research Unit, Department of Medicine, University of California at San Diego, La Jolla, California
| |
Collapse
|
371
|
Alcoholic Liver Disease: A Mouse Model Reveals Protection by Lactobacillus fermentum. Clin Transl Gastroenterol 2016; 7:e138. [PMID: 26795070 PMCID: PMC4737872 DOI: 10.1038/ctg.2015.66] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/04/2015] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES Alcoholism is one of the most devastating diseases with high incidence, but knowledge of its pathology and treatment is still plagued with gaps mostly because of the inherent limitations of research with patients. We developed an animal model for studying liver histopathology, Hsp (heat-shock protein)-chaperones involvement, and response to treatment. METHODS The system was standardized using mice to which ethanol was orally administered alone or in combination with Lactobacillus fermentum following a precise schedule over time and applying, at predetermined intervals, a battery of techniques (histology, immunohistochemistry, western blotting, real-time PCR, immunoprecipitation, 3-nitrotyrosine labeling) to assess liver pathology (e.g., steatosis, fibrosis), and Hsp60 and iNOS (inducible form of nitric oxide synthase) gene expression and protein levels, and post-translational modifications. RESULTS Typical ethanol-induced liver pathology occurred and the effect of the probiotic could be reliably monitored. Steatosis score, iNOS levels, and nitrated proteins (e.g., Hsp60) decreased after probiotic intake. CONCLUSIONS We describe a mouse model useful for studying liver disease induced by chronic ethanol intake and for testing pertinent therapeutic agents, e.g., probiotics. We tested L. fermentum, which reduced considerably ethanol-induced tissue damage and deleterious post-translational modifications of the chaperone Hsp60. The model is available to test other agents and probiotics with therapeutic potential in alcoholic liver disease.
Collapse
|
372
|
Alcoholic Liver Disease: Update on the Role of Dietary Fat. Biomolecules 2016; 6:1. [PMID: 26751488 PMCID: PMC4808795 DOI: 10.3390/biom6010001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/23/2015] [Accepted: 12/07/2015] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease (ALD) spans a spectrum of liver pathology, including fatty liver, alcoholic steatohepatitis, and cirrhosis. Accumulating evidence suggests that dietary factors, including dietary fat, as well as alcohol, play critical roles in the pathogenesis of ALD. The protective effects of dietary saturated fat (SF) and deleterious effects of dietary unsaturated fat (USF) on alcohol-induced liver pathology are well recognized and documented in experimental animal models of ALD. Moreover, it has been demonstrated in an epidemiological study of alcoholic cirrhosis that dietary intake of SF was associated with a lower mortality rates, whereas dietary intake of USF was associated with a higher mortality. In addition, oxidized lipids (dietary and in vivo generated) may play a role in liver pathology. The understanding of how dietary fat contributes to the ALD pathogenesis will enhance our knowledge regarding the molecular mechanisms of ALD development and progression, and may result in the development of novel diet-based therapeutic strategies for ALD management. This review explores the relevant scientific literature and provides a current understanding of recent advances regarding the role of dietary lipids in ALD pathogenesis.
Collapse
|
373
|
Schneider ACR, Rico EP, de Oliveira DL, Rosemberg DB, Guizzo R, Meurer F, da Silveira TR. Lactobacillus rhamnosus GG Effect on Behavior of Zebrafish During Chronic Ethanol Exposure. Biores Open Access 2016; 5:1-5. [PMID: 26862467 PMCID: PMC4744878 DOI: 10.1089/biores.2015.0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ethanol is a widely consumed drug, which acts on the central nervous system to induce behavioral alterations ranging from disinhibition to sedation. Recent studies have produced accumulating evidence for the therapeutic role of probiotic bacteria in behavior. We aimed to investigate the effect of Lactobacillus rhamnosus GG (LGG) on the behavior of adult zebrafish chronically exposed to ethanol. Adult wild-type zebrafish were randomly divided into four groups, each containing 15 fish. The following groups were formed: Control (C), received unsupplemented feed during the trial period; Probiotic (P), fed with feed supplemented with LGG; Ethanol (E), received unsupplemented feed and 0.5% of ethanol directly added to the tank water; and Probiotic+Ethanol (P+E), group under ethanol exposure (0.5%) and fed with LGG supplemented feed. After 2 weeks of exposure, the novel tank test was used to evaluate fish behavior, which was analyzed using computer-aided video tracking. LGG alone did not alter swimming behavior of the fish. Ethanol exposure led to robust behavioral effects in the form of reduced anxiety levels, as indicated by increased vertical exploration and more time spent in the upper region of the novel tank. The group exposed to ethanol and treated with LGG behaved similarly to animals exposed to ethanol alone. Taken together, these results show that zebrafish behavior was not altered by LGG per se, as seen in murine models. This was the first study to investigate the effects of a probiotic diet on behavior after a chronic ethanol exposure.
Collapse
Affiliation(s)
- Ana Claudia Reis Schneider
- Programa de Pós-Graduação: Ciências em Gastroenterologia e Hepatologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Brazil
| | - Eduardo Pacheco Rico
- Programa de Pós-Graduação em Ciências da Saúde, Universidade do Extremo Sul Catarinense-UNESC , Criciúma, Brazil
| | - Diogo Losch de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, ICBS, UFRGS, Porto Alegre, Brazil
| | - Denis Broock Rosemberg
- Programa de Pós Graduação em Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Departamento de Bioquímica e Biologia Molecular, Universidade Federal de Santa Maria , Santa Maria, Brazil
| | - Ranieli Guizzo
- Universidade Federal de Ciências da Saúde de Porto Alegre , Porto Alegre, Brazil
| | - Fábio Meurer
- Engenharia de Alimentos, Universidade Federal do Paraná (UFPR) , Campus Jandaia do Sul, Jandaia do Sul, Brazil
| | - Themis Reverbel da Silveira
- Programa de Pós-Graduação: Ciências em Gastroenterologia e Hepatologia, Universidade Federal do Rio Grande do Sul , Porto Alegre, Brazil
| |
Collapse
|
374
|
Li F, Duan K, Wang C, McClain C, Feng W. Probiotics and Alcoholic Liver Disease: Treatment and Potential Mechanisms. Gastroenterol Res Pract 2015; 2016:5491465. [PMID: 26839540 PMCID: PMC4709639 DOI: 10.1155/2016/5491465] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/06/2015] [Accepted: 11/08/2015] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research, alcohol remains one of the most common causes of liver disease in the United States. Alcoholic liver disease (ALD) encompasses a broad spectrum of disorders, including steatosis, steatohepatitis, and cirrhosis. Although many agents and approaches have been tested in patients with ALD and in animals with experimental ALD in the past, there is still no FDA (Food and Drug Administration) approved therapy for any stage of ALD. With the increasing recognition of the importance of gut microbiota in the onset and development of a variety of diseases, the potential use of probiotics in ALD is receiving increasing investigative and clinical attention. In this review, we summarize recent studies on probiotic intervention in the prevention and treatment of ALD in experimental animal models and patients. Potential mechanisms underlying the probiotic function are also discussed.
Collapse
Affiliation(s)
- Fengyuan Li
- College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Departments of Medicine, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kangmin Duan
- College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
| | - Cuiling Wang
- College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Craig McClain
- Departments of Medicine, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Robley Rex Veterans Affairs Medical Center, Louisville, KY 40202, USA
| | - Wenke Feng
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
- Departments of Medicine, Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
375
|
Guan Y, Cao WJ, Zhang ZY. Relationship between gut microbiota and non-alcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2015; 23:5797-5802. [DOI: 10.11569/wcjd.v23.i36.5797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The incidence of non-alcoholic fatty liver disease (NAFLD) has been increasing during these years. As we understand more about gut microbiota, the relationship between gut microbiota and NAFLD has been revealed. Both animal experiments and clinical studies show that gut microbiota can not only act on NAFLD via the gut-liver axis and two-hit theory, but also play an important role in liver inflammation and hepatic fibrosis. Experiments also indicate that using probiotics, prebiotics, berberine and antibiotics to regulate gut microbiota can relieve inflammation, lower body mass index and improve insulin resistance, which can be a new treatment for NAFLD and other metabolic diseases.
Collapse
|
376
|
Tan J, Chen YX. Dietary and Lifestyle Factors Associated with Colorectal Cancer Risk and Interactions with Microbiota: Fiber, Red or Processed Meat and Alcoholic Drinks. Gastrointest Tumors 2015; 3:17-24. [PMID: 27722153 DOI: 10.1159/000442831] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Diets and lifestyles have been strongly associated with colorectal cancer (CRC). In the past several decades, emerging evidence has suggested that the gut microbiota may have a role in the development of CRC. Its interaction with diets and lifestyles could affect the carcinogenesis of CRC. SUMMARY This review presents the most recent epidemiologic and experimental evidence of three factors that may convincingly have a role in CRC, including fiber, red or processed meat, and alcohol, focusing on potential mechanisms and their interactions with the gut microbiota. KEY MESSAGE High consumption of fiber, low consumption of red or processed red meat as well as minimizing alcohol intake have been associated with a lower risk of CRC. Many microbial metabolites formed from those three substances may mediate the microbial diversity and the composition and abundance of the gut microbiota, which eventually affects the balance between health and disease, including CRC. PRACTICAL IMPLICATIONS Based on our synthetic review, clinicians may probably offer some recommendations and explanations to their patients who may want to modulate their diet and lifestyle to prevent CRC. As an easily modifiable environmental factor, it may be possible that applying dietary or lifestyle intervention could effectively protect against the development of CRC in the future.
Collapse
Affiliation(s)
- Juan Tan
- Division of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institution of Digestive Disease, Shanghai, China
| | - Ying-Xuan Chen
- Division of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Institution of Digestive Disease, Shanghai, China
| |
Collapse
|
377
|
Azimirad M, Rostami-Nejad M, Rostami K, Naji T, Zali MR. The Susceptibility of Celiac Disease Intestinal Microbiota to Clostridium difficile Infection. Am J Gastroenterol 2015; 110:1740-1741. [PMID: 26673511 DOI: 10.1038/ajg.2015.360] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Masoumeh Azimirad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Department of Coeliac Disease, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rostami
- Department of Gastroenterology, Alexandra Hospital, Worcestershire, UK
| | - Tahere Naji
- Department of Cell and Molecular Biology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Reza Zali
- Department of Coeliac Disease, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
378
|
Natarajan SK, Pachunka JM, Mott JL. Role of microRNAs in Alcohol-Induced Multi-Organ Injury. Biomolecules 2015; 5:3309-38. [PMID: 26610589 PMCID: PMC4693280 DOI: 10.3390/biom5043309] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 11/16/2015] [Indexed: 12/12/2022] Open
Abstract
Alcohol consumption and its abuse is a major health problem resulting in significant healthcare cost in the United States. Chronic alcoholism results in damage to most of the vital organs in the human body. Among the alcohol-induced injuries, alcoholic liver disease is one of the most prevalent in the United States. Remarkably, ethanol alters expression of a wide variety of microRNAs that can regulate alcohol-induced complications or dysfunctions. In this review, we will discuss the role of microRNAs in alcoholic pancreatitis, alcohol-induced liver damage, intestinal epithelial barrier dysfunction, and brain damage including altered hippocampus structure and function, and neuronal loss, alcoholic cardiomyopathy, and muscle damage. Further, we have reviewed the role of altered microRNAs in the circulation, teratogenic effects of alcohol, and during maternal or paternal alcohol consumption.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Joseph M Pachunka
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Justin L Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, 985870 Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
379
|
Chen P, Miyamoto Y, Mazagova M, Lee KC, Eckmann L, Schnabl B. Microbiota Protects Mice Against Acute Alcohol-Induced Liver Injury. Alcohol Clin Exp Res 2015; 39:2313-23. [PMID: 26556636 DOI: 10.1111/acer.12900] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Our aim is to investigate the physiological relevance of the intestinal microbiota in alcohol-induced liver injury. Chronic alcohol abuse is associated with intestinal bacterial overgrowth, increased intestinal permeability, and translocation of microbial products from the intestine to the portal circulation and liver. Translocated microbial products contribute to experimental alcoholic liver disease. METHODS We subjected germ-free and conventional C57BL/6 mice to a model of acute alcohol exposure that mimics binge drinking. RESULTS Germ-free mice showed significantly greater liver injury and inflammation after oral gavage of ethanol (EtOH) compared with conventional mice. In parallel, germ-free mice exhibited increased hepatic steatosis and up-regulated expression of genes involved in fatty acid and triglyceride synthesis compared with conventional mice after acute EtOH administration. The absence of microbiota was also associated with increased hepatic expression of EtOH-metabolizing enzymes, which led to faster EtOH elimination from the blood and lower plasma EtOH concentrations. Intestinal levels of EtOH-metabolizing genes showed regional expression differences and were overall higher in germ-free mice relative to conventional mice. CONCLUSIONS Our findings indicate that absence of the intestinal microbiota increases hepatic EtOH metabolism and the susceptibility to binge-like alcohol drinking.
Collapse
Affiliation(s)
- Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, California.,Department of Pathophysiology, Southern Medical University, Guangzhou, China
| | - Yukiko Miyamoto
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Magdalena Mazagova
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Kuei-Chuan Lee
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Lars Eckmann
- Department of Medicine, University of California San Diego, La Jolla, California
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California.,Department of Medicine, VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
380
|
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
|
381
|
Modulation of Intestinal Barrier and Bacterial Endotoxin Production Contributes to the Beneficial Effect of Nicotinic Acid on Alcohol-Induced Endotoxemia and Hepatic Inflammation in Rats. Biomolecules 2015; 5:2643-58. [PMID: 26501337 PMCID: PMC4693251 DOI: 10.3390/biom5042643] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/12/2022] Open
Abstract
Alcohol consumption causes nicotinic acid deficiency. The present study was undertaken to determine whether dietary nicotinic acid supplementation provides beneficial effects on alcohol-induced endotoxin signaling and the possible mechanisms at the gut-liver axis. Male Sprague-Dawley rats were pair-fed the Lieber-DeCarli liquid diets containing ethanol or isocaloric maltose dextrin for eight weeks, with or without dietary supplementation with 750 mg/liter nicotinic acid. Chronic alcohol feeding elevated the plasma endotoxin level and activated hepatic endotoxin signaling cascade, which were attenuated by nicotinic acid supplementation. Alcohol consumption remarkably decreased the mRNA levels of claudin-1, claudin-5, and ZO-1 in the distal intestine, whereas nicotinic acid significantly up-regulated these genes. The concentrations of endotoxin, ethanol, and acetaldehyde in the intestinal contents were increased by alcohol exposure, and niacin supplementation reduced the intestinal endotoxin and acetaldehyde levels. Nicotinic acid supplementation upregulated the intestinal genes involved in aldehyde detoxification via transcriptional regulation. These results demonstrate that modulation of the intestinal barrier function and bacterial endotoxin production accounts for the inhibitory effects of nicotinic acid on alcohol-induced endotoxemia and hepatic inflammation.
Collapse
|
382
|
Alcohol and the Intestine. Biomolecules 2015; 5:2573-88. [PMID: 26501334 PMCID: PMC4693248 DOI: 10.3390/biom5042573] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 09/24/2015] [Accepted: 10/05/2015] [Indexed: 02/07/2023] Open
Abstract
Alcohol abuse is a significant contributor to the global burden of disease and can lead to tissue damage and organ dysfunction in a subset of alcoholics. However, a subset of alcoholics without any of these predisposing factors can develop alcohol-mediated organ injury. The gastrointestinal tract (GI) could be an important source of inflammation in alcohol-mediated organ damage. The purpose of review was to evaluate mechanisms of alcohol-induced endotoxemia (including dysbiosis and gut leakiness), and highlight the predisposing factors for alcohol-induced dysbiosis and gut leakiness to endotoxins. Barriers, including immunologic, physical, and biochemical can regulate the passage of toxins into the portal and systemic circulation. In addition, a host of environmental interactions including those influenced by circadian rhythms can impact alcohol-induced organ pathology. There appears to be a role for therapeutic measures to mitigate alcohol-induced organ damage by normalizing intestinal dysbiosis and/or improving intestinal barrier integrity. Ultimately, the inflammatory process that drives progression into organ damage from alcohol appears to be multifactorial. Understanding the role of the intestine in the pathogenesis of alcoholic liver disease can pose further avenues for pathogenic and treatment approaches.
Collapse
|
383
|
Parsa Yeganeh L, Azarbaijani R, Mousavi H, Shahzadeh Fazeli SA, Amoozgar MA, Salekdeh GH. Genome-Wide Analysis of Oceanimonas sp. GK1 Isolated from Gavkhouni Wetland (Iran) Demonstrates Presence of Genes for Virulence and Pathogenicity. CELL JOURNAL 2015; 17:451-60. [PMID: 26464816 PMCID: PMC4601865 DOI: 10.22074/cellj.2015.6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 01/06/2015] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The bacterium Oceanimonas sp. (O. sp.) GK1 is a member of the Aeromonadaceae family and its genome represents several virulence genes involved in fish and human pathogenicity. In this original research study we aimed to identify and characterize the putative virulence factors and pathogenicity of this halotolerant marine bacterium using genome wide analysis. MATERIALS AND METHODS The genome data of O. sp. GK1 was obtained from NCBI. Comparative genomic study was done using MetaCyc database. RESULTS Whole genome data analysis of the O. sp. GK1 revealed that the bacterium possesses some important virulence genes (e.g. ZOT, RTX toxin, thermostable hemolysin, lateral flagella and type IV pili) which have been implicated in adhesion and biofilm formation and infection in some other pathogenic bacteria. CONCLUSION This is the first report of the putative pathogenicity of O. sp.GK1. The genome wide analysis of the bacterium demonstrates the presence of virulence genes causing infectious diseases in many warmand cold-blooded animals.
Collapse
Affiliation(s)
- Laleh Parsa Yeganeh
- Molecular Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Reza Azarbaijani
- Molecular Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Hossein Mousavi
- Molecular Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Seyed Abolhassan Shahzadeh Fazeli
- Molecular Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran ; Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | | | - Ghasem Hosseini Salekdeh
- Molecular Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran ; Agricultural Biotechnology Research Institute of Iran, Karaj, Iran ; Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
384
|
Affiliation(s)
- Irina A. Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of MedicineUniversity of Louisville School of MedicineLouisvilleKY,Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleKY
| | - Dipendra Parajuli
- Division of Gastroenterology, Hepatology, and Nutrition, Department of MedicineUniversity of Louisville School of MedicineLouisvilleKY,Robley Rex Veterans Medical CenterLouisvilleKY
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of MedicineUniversity of Louisville School of MedicineLouisvilleKY,Department of Pharmacology and ToxicologyUniversity of Louisville School of MedicineLouisvilleKY,Robley Rex Veterans Medical CenterLouisvilleKY
| |
Collapse
|
385
|
Bagyánszki M, Bódi N. Gut region-dependent alterations of nitrergic myenteric neurons after chronic alcohol consumption. World J Gastrointest Pathophysiol 2015; 6:51-57. [PMID: 26301118 PMCID: PMC4540706 DOI: 10.4291/wjgp.v6.i3.51] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/27/2015] [Accepted: 06/02/2015] [Indexed: 02/06/2023] Open
Abstract
Chronic alcohol abuse damages nearly every organ in the body. The harmful effects of ethanol on the brain, the liver and the pancreas are well documented. Although chronic alcohol consumption causes serious impairments also in the gastrointestinal tract like altered motility, mucosal damage, impaired absorption of nutrients and inflammation, the effects of chronically consumed ethanol on the enteric nervous system are less detailed. While the nitrergic myenteric neurons play an essential role in the regulation of gastrointestinal peristalsis, it was hypothesised, that these neurons are the first targets of consumed ethanol or its metabolites generated in the different gastrointestinal segments. To reinforce this hypothesis the effects of ethanol on the gastrointestinal tract was investigated in different rodent models with quantitative immunohistochemistry, in vivo and in vitro motility measurements, western blot analysis, evaluation of nitric oxide synthase enzyme activity and bio-imaging of nitric oxide synthesis. These results suggest that chronic alcohol consumption did not result significant neural loss, but primarily impaired the nitrergic pathways in gut region-dependent way leading to disturbed gastrointestinal motility. The gut segment-specific differences in the effects of chronic alcohol consumption highlight the significance the ethanol-induced neuronal microenvironment involving oxidative stress and intestinal microbiota.
Collapse
|
386
|
Joshi-Barve S, Kirpich I, Cave MC, Marsano LS, McClain CJ. Alcoholic, Nonalcoholic, and Toxicant-Associated Steatohepatitis: Mechanistic Similarities and Differences. Cell Mol Gastroenterol Hepatol 2015; 1:356-367. [PMID: 28210688 PMCID: PMC5301292 DOI: 10.1016/j.jcmgh.2015.05.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022]
Abstract
Hepatic steatosis and steatohepatitis are common histologic findings that can be caused by multiple etiologies. The three most frequent causes for steatosis/steatohepatitis are alcohol (alcoholic steatohepatitis, ASH), obesity/metabolic syndrome (nonalcoholic steatohepatitis, NASH), and environmental toxicants (toxicant-associated steatohepatitis, TASH). Hepatic steatosis is an early occurrence in all three forms of liver disease, and they often share common pathways to disease progression/severity. Disease progression is a result of both direct effects on the liver as well as indirect alterations in other organs/tissues such as intestine, adipose tissue, and the immune system. Although the three liver diseases (ASH, NASH, and TASH) share many common pathogenic mechanisms, they also exhibit distinct differences. Both shared and divergent mechanisms can be potential therapeutic targets. This review provides an overview of selected important mechanistic similarities and differences in ASH, NASH, and TASH.
Collapse
Key Words
- ALD, alcoholic liver disease
- ALT, alanine aminotransferase
- ASH, alcoholic steatohepatitis
- AST, aspartate transaminase
- Alcoholic Steatohepatitis
- BMI, body mass index
- CYP2E1, cytochrome P450 isoform 2E1
- ECM, extracellular matrix
- ER, endoplasmic reticulum
- HCC, hepatocellular carcinoma
- HDAC, histone deacetylase
- HSC, hepatic stellate cell
- IL, interleukin
- LA, linoleic acid
- LPS, lipopolysaccharide
- Mechanisms
- NAFLD, nonalcoholic fatty liver disease
- NASH, nonalcoholic steatohepatitis
- NK, natural killer
- NKT, natural killer T
- Nonalcoholic Steatohepatitis
- OXLAM, oxidized linoleic acid metabolite
- PAI-1, plasminogen activator inhibitor-1
- PCB153, 2,2′,4,4′,5,5′-hexachlorobiphenyl
- PPAR, peroxisome proliferator-activated receptor
- RNS, reactive nitrogen species
- SNP, single-nucleotide polymorphism
- TAFLD, toxicant-associated fatty liver disease
- TASH, toxicant-associated steatohepatitis
- TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
- TH, helper T cell
- TLR, Toll-like receptor
- TNF, tumor necrosis factor
- Toxicant-Associated Steatohepatitis
- VA, U.S. Department of Veterans Affairs/Veterans Administration
- miR, microRNA
Collapse
Affiliation(s)
- Swati Joshi-Barve
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Irina Kirpich
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Matthew C. Cave
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Luis S. Marsano
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky
| | - Craig J. McClain
- Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky,Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky,Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky,Correspondence Address correspondence to: Craig J. McClain, MD, University of Louisville, 505 South Hancock Street, Louisville, Kentucky 40292. fax: (502) 852-8927.
| |
Collapse
|
387
|
Vassallo G, Mirijello A, Ferrulli A, Antonelli M, Landolfi R, Gasbarrini A, Addolorato G. Review article: Alcohol and gut microbiota - the possible role of gut microbiota modulation in the treatment of alcoholic liver disease. Aliment Pharmacol Ther 2015; 41:917-27. [PMID: 25809237 DOI: 10.1111/apt.13164] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/25/2014] [Accepted: 02/27/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alcohol abuse represents the most common cause of liver disease in the Western countries. Pre-clinical and clinical studies showed that alcohol consumption affects amount and composition of gut microbiota. Moreover, gut flora plays an important role in the pathogenesis of alcoholic liver injury. AIM To review the relationship between alcohol administration and changes on gut microbiota, its involvement in the pathogenesis of alcoholic liver disease, and how gut microbiota modulation could be a target for the treatment of alcoholic liver disease. METHODS Articles were identified using the PubMed database with the search terms 'Alcohol', 'Gut Microbiota', 'Alcoholic liver disease', 'Probiotic', 'Prebiotic', 'Symbiotic' and 'Antibiotic'. English-language articles were screened for relevance. Full review of publications for the relevant studies was conducted, including additional publications that were identified from individual article reference lists. RESULTS Alcohol abuse induces changes in the composition of gut microbiota, although the exact mechanism for this alteration is not well known. The translocation of bacterial products into the portal blood appears to play a key role in alcohol-induced liver damage. Several studies show that the modulation of gut microbiota seem to be a promising strategy to reduce alcohol-induced liver injury. CONCLUSIONS Further studies are needed to better understand the relationship between alcohol administration and changes in gut microbiota, and its involvement in alcoholic liver disease. Moreover larger studies are needed to confirm the preliminary results on the therapeutic effects of gut microbiota modulation.
Collapse
Affiliation(s)
- G Vassallo
- Department of Internal Medicine, Catholic University of Rome, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
388
|
Abstract
The leaky gut hypothesis links translocating microbial products with the onset and progression of liver disease, and for a long time was considered one of its major contributors. However, a more detailed picture of the intestinal microbiota contributing to liver disease started to evolve. The gut is colonized by trillions of microbes that aid in digestion, modulate immune response, and generate a variety of products that result from microbial metabolic activities. These products together with host-bacteria interactions influence both normal physiology and disease susceptibility. A disruption of the symbiosis between microbiota and host is known as dysbiosis and can have profound effects on health. Qualitative changes such as increased proportions of harmful bacteria and reduced levels of beneficial bacteria, and also quantitative changes in the total amount of bacteria (overgrowth) have been associated with liver disease. Understanding the link between the pathophysiology of liver diseases and compositional and functional changes of the microbiota will help in the design of innovative therapies. In this review, we focus on factors resulting in dysbiosis, and discuss how dysbiosis can disrupt intestinal homeostasis and contribute to liver disease.
Collapse
Key Words
- dysbiosis
- leaky gut
- alcoholic liver disease
- nash
- nafld
- cirrhosis
- microbiome
- pamps
- ald, alcoholic liver disease
- amp, antimicrobial peptides and proteins
- fiaf, fasting-induced adipocyte factor
- hfd, high-fat diet
- ibd, inflammatory bowel disease
- il, interleukin
- lcfa, long-chain fatty acid
- lps, lipopolysaccharide
- nafld, nonalcoholic fatty liver disease
- nash, nonalcoholic steatohepatitis
- nlrp, nucleotide-binding domain and leucine rich repeat-containing protein
- nod2, nucleotide-binding oligomerization domain 2
- pamps, pathogen-associated molecular patterns
- reg3, regenerating islet-derived 3
- tlr, toll-like receptor
- tnf, tumor necrosis factor
- tnfr, tumor necrosis factor receptor
Collapse
Affiliation(s)
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, and Department of Medicine, VA San Diego Healthcare System, San Diego, California
| |
Collapse
|
389
|
Massey VL, Stocke KS, Schmidt RH, Tan M, Ajami N, Neal RE, Petrosino JF, Barve S, Arteel GE. Oligofructose protects against arsenic-induced liver injury in a model of environment/obesity interaction. Toxicol Appl Pharmacol 2015; 284:304-14. [PMID: 25759243 PMCID: PMC4515777 DOI: 10.1016/j.taap.2015.02.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 02/25/2015] [Accepted: 02/26/2015] [Indexed: 12/11/2022]
Abstract
Arsenic (As) tops the ATSDR list of hazardous environmental chemicals and is known to cause liver injury. Although the concentrations of As found in the US water supply are generally too low to directly damage the liver, subhepatotoxic doses of As sensitize the liver to experimental NAFLD. It is now suspected that GI microbiome dysbiosis plays an important role in development of NALFD. Importantly, arsenic has also been shown to alter the microbiome. The purpose of the current study was to test the hypothesis that the prebiotic oligofructose (OFC) protects against enhanced liver injury caused by As in experimental NAFLD. Male C57Bl6/J mice were fed low fat diet (LFD), high fat diet (HFD), or HFD containing oligofructose (OFC) during concomitant exposure to either tap water or As-containing water (4.9ppm as sodium arsenite) for 10weeks. HFD significantly increased body mass and caused fatty liver injury, as characterized by an increased liver weight-to-body weight ratio, histologic changes and transaminases. As observed previously, As enhanced HFD-induced liver damage, which was characterized by enhanced inflammation. OFC supplementation protected against the enhanced liver damage caused by As in the presence of HFD. Interestingly, arsenic, HFD and OFC all caused unique changes to the gut flora. These data support previous findings that low concentrations of As enhance liver damage caused by high fat diet. Furthermore, these results indicate that these effects of arsenic may be mediated, at least in part, by GI tract dysbiosis and that prebiotic supplementation may confer significant protective effects.
Collapse
Affiliation(s)
- Veronica L Massey
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Kendall S Stocke
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Robin H Schmidt
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Min Tan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Nadim Ajami
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Rachel E Neal
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Joseph F Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Shirish Barve
- Department of Medicine, University of Louisville Health Sciences Center, Louisville, KY 40292, USA
| | - Gavin E Arteel
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY 40292, USA.
| |
Collapse
|
390
|
de Almada CN, Nunes de Almada C, Martinez RCR, Sant’Ana ADS. Characterization of the intestinal microbiota and its interaction with probiotics and health impacts. Appl Microbiol Biotechnol 2015; 99:4175-99. [DOI: 10.1007/s00253-015-6582-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Revised: 04/01/2015] [Accepted: 04/03/2015] [Indexed: 02/08/2023]
|
391
|
Explaining diversity in metagenomic datasets by phylogenetic-based feature weighting. PLoS Comput Biol 2015; 11:e1004186. [PMID: 25815895 PMCID: PMC4376673 DOI: 10.1371/journal.pcbi.1004186] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 02/12/2015] [Indexed: 02/06/2023] Open
Abstract
Metagenomics is revolutionizing our understanding of microbial communities, showing that their structure and composition have profound effects on the ecosystem and in a variety of health and disease conditions. Despite the flourishing of new analysis methods, current approaches based on statistical comparisons between high-level taxonomic classes often fail to identify the microbial taxa that are differentially distributed between sets of samples, since in many cases the taxonomic schema do not allow an adequate description of the structure of the microbiota. This constitutes a severe limitation to the use of metagenomic data in therapeutic and diagnostic applications. To provide a more robust statistical framework, we introduce a class of feature-weighting algorithms that discriminate the taxa responsible for the classification of metagenomic samples. The method unambiguously groups the relevant taxa into clades without relying on pre-defined taxonomic categories, thus including in the analysis also those sequences for which a taxonomic classification is difficult. The phylogenetic clades are weighted and ranked according to their abundance measuring their contribution to the differentiation of the classes of samples, and a criterion is provided to define a reduced set of most relevant clades. Applying the method to public datasets, we show that the data-driven definition of relevant phylogenetic clades accomplished by our ranking strategy identifies features in the samples that are lost if phylogenetic relationships are not considered, improving our ability to mine metagenomic datasets. Comparison with supervised classification methods currently used in metagenomic data analysis highlights the advantages of using phylogenetic information. In metagenomics, the composition of complex microbial communities is characterized using Next Generation Sequencing technologies. Thanks to the decreasing cost of sequencing, large amounts of data have been generated for environmental samples and for a variety of health-associated conditions. In parallel there has been a flourishing of statistical methods to analyze metagenomic datasets, concentrating mainly on the problem of assessing the existence of significant differences between microbial communities in different conditions. However, for a large number of therapeutic and diagnostic applications it would be essential to identify and rank the microbial taxa that are most relevant in these comparisons. Here we present PhyloRelief, a novel feature-ranking algorithm that fills this gap by integrating the phylogenetic relationships amongst the taxa into a statistical feature weighting procedure. Without relying on a precompiled taxonomy, PhyloRelief determines the lineages most relevant to the diversification of the samples guided by the data. As such, PhyloRelief can be applied both to cases in which sequences can be classified according to a known taxonomy, and to cases in which this is not feasible, a common occurrence in metagenomic data analysis given the increasing number of new and uncultivable taxa that are discovered using these technologies.
Collapse
|
392
|
Couch RD, Dailey A, Zaidi F, Navarro K, Forsyth CB, Mutlu E, Engen PA, Keshavarzian A. Alcohol induced alterations to the human fecal VOC metabolome. PLoS One 2015; 10:e0119362. [PMID: 25751150 PMCID: PMC4353727 DOI: 10.1371/journal.pone.0119362] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022] Open
Abstract
Studies have shown that excessive alcohol consumption impacts the intestinal microbiota composition, causing disruption of homeostasis (dysbiosis). However, this observed change is not indicative of the dysbiotic intestinal microbiota function that could result in the production of injurious and toxic products. Thus, knowledge of the effects of alcohol on the intestinal microbiota function and their metabolites is warranted, in order to better understand the role of the intestinal microbiota in alcohol associated organ failure. Here, we report the results of a differential metabolomic analysis comparing volatile organic compounds (VOC) detected in the stool of alcoholics and non-alcoholic healthy controls. We performed the analysis with fecal samples collected after passage as well as with samples collected directly from the sigmoid lumen. Regardless of the approach to fecal collection, we found a stool VOC metabolomic signature in alcoholics that is different from healthy controls. The most notable metabolite alterations in the alcoholic samples include: (1) an elevation in the oxidative stress biomarker tetradecane; (2) a decrease in five fatty alcohols with anti-oxidant property; (3) a decrease in the short chain fatty acids propionate and isobutyrate, important in maintaining intestinal epithelial cell health and barrier integrity; (4) a decrease in alcohol consumption natural suppressant caryophyllene; (5) a decrease in natural product and hepatic steatosis attenuator camphene; and (6) decreased dimethyl disulfide and dimethyl trisulfide, microbial products of decomposition. Our results showed that intestinal microbiota function is altered in alcoholics which might promote alcohol associated pathologies.
Collapse
Affiliation(s)
- Robin D. Couch
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
- * E-mail:
| | - Allyson Dailey
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Fatima Zaidi
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Karl Navarro
- Department of Chemistry and Biochemistry, George Mason University, Manassas, Virginia, United States of America
| | - Christopher B. Forsyth
- Department of Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Biochemistry, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Ece Mutlu
- Department of Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Phillip A. Engen
- Department of Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Ali Keshavarzian
- Department of Medicine, Division of Digestive Diseases and Nutrition, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Pharmacology, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
393
|
Olivares M, Neef A, Castillejo G, Palma GD, Varea V, Capilla A, Palau F, Nova E, Marcos A, Polanco I, Ribes-Koninckx C, Ortigosa L, Izquierdo L, Sanz Y. The HLA-DQ2 genotype selects for early intestinal microbiota composition in infants at high risk of developing coeliac disease. Gut 2015; 64:406-17. [PMID: 24939571 DOI: 10.1136/gutjnl-2014-306931] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Intestinal dysbiosis has been associated with coeliac disease (CD), but whether the alterations are cause or consequence of the disease is unknown. This study investigated whether the human leukocyte antigen (HLA)-DQ2 genotype is an independent factor influencing the early gut microbiota composition of healthy infants at family risk of CD. DESIGN As part of a larger prospective study, a subset (n=22) of exclusively breastfed and vaginally delivered infants with either high genetic risk (HLA-DQ2 carriers) or low genetic risk (non-HLA-DQ2/8 carriers) of developing CD were selected from a cohort of healthy infants with at least one first-degree relative with CD. Infant faecal microbiota was analysed by 16S rRNA gene pyrosequencing and real time quantitative PCR. RESULTS Infants with a high genetic risk had significantly higher proportions of Firmicutes and Proteobacteria and lower proportions of Actinobacteria compared with low-risk infants. At genus level, high-risk infants had significantly less Bifidobacterium and unclassified Bifidobacteriaceae proportions and more Corynebacterium, Gemella, Clostridium sensu stricto, unclassified Clostridiaceae, unclassified Enterobacteriaceae and Raoultella proportions. Quantitative real time PCR also revealed lower numbers of Bifidobacterium species in infants with high genetic risk than in those with low genetic risk. In high-risk infants negative correlations were identified between Bifidobacterium species and several genera of Proteobacteria (Escherichia/Shigella) and Firmicutes (Clostridium). CONCLUSIONS The genotype of infants at family risk of developing CD, carrying the HLA-DQ2 haplotypes, influences the early gut microbiota composition. This finding suggests that a specific disease-biased host genotype may also select for the first gut colonisers and could contribute to determining disease risk.
Collapse
Affiliation(s)
- M Olivares
- Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Valencia, Spain
| | - A Neef
- Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Valencia, Spain
| | - G Castillejo
- Hospital Universitario Sant Joan de Reus, Tarragona, Spain
| | - G De Palma
- Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Valencia, Spain
| | - V Varea
- Gastroenterología, Nutrición y Hepatología Pediátrica, Hospital Universitario Sant Joan de Deu and Unidad de Gastroenterología Pediátrica del Institut Dexeus, Barcelona, Spain
| | - A Capilla
- Centro de Investigación Príncipe Felipe (CIPF) and IBV-CSIC Associated Unit, CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - F Palau
- Centro de Investigación Príncipe Felipe (CIPF) and IBV-CSIC Associated Unit, CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - E Nova
- Department Metabolismo y Nutrición, ICTAN-CSIC, Madrid, Spain
| | - A Marcos
- Department Metabolismo y Nutrición, ICTAN-CSIC, Madrid, Spain
| | - I Polanco
- Servicio de Gastroenterología y Nutrición Pediátrica, Hospital Universitario La Paz, Madrid, Spain
| | - C Ribes-Koninckx
- Unidad de Gastroenterología Pediátrica, Hospital Universitario La Fe, Valencia, Spain
| | - L Ortigosa
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Canarias, Spain
| | - L Izquierdo
- Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Valencia, Spain
| | - Y Sanz
- Instituto de Agroquímica y Tecnología de Alimentos, Consejo Superior de Investigaciones Científicas (IATA-CSIC), Valencia, Spain
| |
Collapse
|
394
|
Barve S, Kirpich IA, McClain CJ. Tumor necrosis factor alpha-induced receptor 1 signaling in alcoholic liver disease: A gut reaction? Hepatology 2015; 61:754-6. [PMID: 25482079 PMCID: PMC6445372 DOI: 10.1002/hep.27640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 12/02/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Shirish Barve
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY
| | | | | |
Collapse
|
395
|
Manley S, Ding W. Role of farnesoid X receptor and bile acids in alcoholic liver disease. Acta Pharm Sin B 2015; 5:158-67. [PMID: 26579442 PMCID: PMC4629219 DOI: 10.1016/j.apsb.2014.12.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/20/2014] [Accepted: 12/29/2014] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease (ALD) is one of the major causes of liver morbidity and mortality worldwide. Chronic alcohol consumption leads to development of liver pathogenesis encompassing steatosis, inflammation, fibrosis, cirrhosis, and in extreme cases, hepatocellular carcinoma. Moreover, ALD may also associate with cholestasis. Emerging evidence now suggests that farnesoid X receptor (FXR) and bile acids also play important roles in ALD. In this review, we discuss the effects of alcohol consumption on FXR, bile acids and gut microbiome as well as their impacts on ALD. Moreover, we summarize the findings on FXR, FoxO3a (forkhead box-containing protein class O3a) and PPARα (peroxisome proliferator-activated receptor alpha) in regulation of autophagy-related gene transcription program and liver injury in response to alcohol exposure.
Collapse
Key Words
- 6ECDCA, 6α-ethyl-chenodeoxycholic acid
- ADH, alcohol dehydrogenase
- AF, activation function
- AKT, protein kinase B
- ALD, alcoholic liver disease
- ALT, alanine aminotransferase
- ASBT, apical sodium dependent bile acid transporter
- Alcoholic liver disease
- Atg, autophagy-related
- Autophagy
- BAAT, bile acid CoA:amino acid N-acyltransferase
- BACS, bile acid CoA synthetase
- BSEP, bile salt export pump
- Bile acids
- CA, cholic acid
- CB1R, cannabinoid receptor type 1
- CDCA, chenodeoxycholic acid
- CREB, cAMP response element-binding protein
- CREBH, cAMP response element-binding protein, hepatocyte specific
- CRTC2, CREB regulated transcription coactivator 2
- CYP, cytochrome P450
- DCA, deoxycholic acid
- DR1, direct repeat 1
- FGF15/19, fibroblast growth factor 15/19
- FGFR4, fibroblast growth factor receptor 4
- FXR, farnesoid X receptor
- Farnesoid X receptor
- FoxO3
- FoxO3a, forkhead box-containing protein class O3a
- GGT, gamma-glutamyltranspeptidase
- HCC, hepatocellular carcinoma
- IR-1, inverted repeat-1
- KO, knockout
- LC3, light chain 3
- LRH-1, liver receptor homolog 1
- LXR, liver X receptor
- MRP4, multidrug resistance protein 4
- NAD+, nicotinamide adenine dinucleotide
- NTCP, sodium taurocholate cotransporting polypeptide
- OSTα/β, organic solute transporter α/β
- PE, phosphatidylethanolamine
- PPARα, peroxisome proliferator-activated receptor alpha
- ROS, reactive oxygen species
- RXRα, retinoid X receptor-alpha
- SHP, small heterodimer partner
- SQSTM, sequestome-1
- SREBP1, sterol regulatory element-binding protein 1
- Sirt1, sirtuin 1
- TCA, taurocholic acid
- TFEB, transcription factor EB
- TLR4, toll-like receptor 4
- TUDCA, tauro-ursodeoxycholic acid
- UDCA, ursodeoxycholic acid
- WAY, WAY-362450
- WT, wild type
Collapse
Affiliation(s)
| | - Wenxing Ding
- Corresponding author. Tel.: +1 913 5889813; fax: +1 913 5887501.
| |
Collapse
|
396
|
Chen P, Stärkel P, Turner JR, Ho SB, Schnabl B. Dysbiosis-induced intestinal inflammation activates tumor necrosis factor receptor I and mediates alcoholic liver disease in mice. Hepatology 2015; 61:883-94. [PMID: 25251280 PMCID: PMC4340725 DOI: 10.1002/hep.27489] [Citation(s) in RCA: 251] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 09/22/2014] [Indexed: 12/15/2022]
Abstract
UNLABELLED Intestinal barrier dysfunction is an important contributor to alcoholic liver disease (ALD). Translocated microbial products trigger an inflammatory response in the liver and contribute to steatohepatitis. Our aim was to investigate mechanisms of barrier disruption after chronic alcohol feeding. A Lieber-DeCarli model was used to induce intestinal dysbiosis, increased intestinal permeability, and liver disease in mice. Alcohol feeding for 8 weeks induced intestinal inflammation in the jejunum, which is characterized by an increased number of tumor necrosis factor alpha (TNF-α)-producing monocytes and macrophages. These findings were confirmed in duodenal biopsies from patients with chronic alcohol abuse. Intestinal decontamination with nonabsorbable antibiotics restored eubiosis, decreased intestinal inflammation and permeability, and reduced ALD in mice. TNF-receptor I (TNFRI) mutant mice were protected from intestinal barrier dysfunction and ALD. To investigate whether TNFRI on intestinal epithelial cells mediates intestinal barrier dysfunction and ALD, we used TNFRI mutant mice carrying a conditional gain-of-function allele for this receptor. Reactivation of TNFRI on intestinal epithelial cells resulted in increased intestinal permeability and liver disease that is similar to wild-type mice after alcohol feeding, suggesting that enteric TNFRI promotes intestinal barrier dysfunction. Myosin light-chain kinase (MLCK) is a downstream target of TNF-α and was phosphorylated in intestinal epithelial cells after alcohol administration. Using MLCK-deficient mice, we further demonstrate a partial contribution of MLCK to intestinal barrier dysfunction and liver disease after chronic alcohol feeding. CONCLUSION Dysbiosis-induced intestinal inflammation and TNFRI signaling in intestinal epithelial cells mediate a disruption of the intestinal barrier. Therefore, intestinal TNFRI is a crucial mediator of ALD.
Collapse
Affiliation(s)
- Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, CA
| | - Peter Stärkel
- St. Luc University Hospital, Université Catholique de Louvain, Brussels, Belgium
| | | | - Samuel B. Ho
- Department of Medicine, University of California San Diego, La Jolla, CA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
397
|
Hong M, Kim SW, Han SH, Kim DJ, Suk KT, Kim YS, Kim MJ, Kim MY, Baik SK, Ham YL. Probiotics (Lactobacillus rhamnosus R0011 and acidophilus R0052) reduce the expression of toll-like receptor 4 in mice with alcoholic liver disease. PLoS One 2015; 10:e0117451. [PMID: 25692549 PMCID: PMC4333821 DOI: 10.1371/journal.pone.0117451] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/25/2014] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE The role of lipopolysaccharide (LPS) and toll-like receptor 4 (TLR 4) in the pathogenesis of alcoholic liver disease (ALD) has been widely established. We evaluated the biological effects of probiotics (Lactobacillus rhamnosus R0011 and acidophilus R0052), KRG (Korea red ginseng), and urushiol (Rhus verniciflua Stokes) on ALD, including their effects on normal and high-fat diet in mice. METHODS One hundred C57BL/6 mice were classified into normal (N) and high-fat diet (H) groups. Each group was divided into 5 sub-groups: control, alcohol, alcohol+probiotics, alcohol+KRG, and alcohol+urushiol. A liver function test, histology, electron-microscopy, interleukin (IL)-1β, tumor necrosis factor (TNF)-α, IL-6, and IL-10, and TLR 4 were evaluated and compared. RESULTS In the N group, probiotics, KRG, and urushiol significantly reduced levels of TNF-α (12.3±5.1, 13.4±3.9, and 12.1±4.3 vs. 27.9±15.2 pg/mL) and IL-1β (108.4±39.4, 75.0±51.0, and 101.1±26.8 vs. 162.4±37.5 pg/mL), which were increased by alcohol. Alcohol-induced TLR 4 expression was reduced by probiotics and urushiol (0.7±0.2, and 0.8±0.1 vs. 1.0±0.3, p<0.001). In the H group, IL-10 was significantly increased by probiotics and KRG, compared with alcohol (25.3±15.6 and 20.4±6.2 vs. 7.6±5.6 pg/mL) and TLR 4 expression was reduced by probiotics (0.8±0.2 vs. 1.0±0.3, p = 0.007). CONCLUSIONS Alcohol-induced TLR 4 expression was down-regulated by probiotics in the normal and high-fat diet groups. Probiotics, KRG, and urushiol might be effective in the treatment of ALD by regulating the gut-liver axis.
Collapse
Affiliation(s)
- Meegun Hong
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
| | - Seung Woo Kim
- Department of Biomedical science, Hallym University College of Medicine, Chuncheon, South Korea
| | - Sang Hak Han
- Department of Pathology, Hallym University College of Medicine, Chuncheon, South Korea
| | - Dong Joon Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
| | - Ki Tae Suk
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
- Department of Medicine, Columbia University, New York, New York, United States of America
| | - Yeon Soo Kim
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, South Korea
| | - Myong Jo Kim
- College of Agriculture and Life Science, Kangwon National University, Chuncheon, South Korea
| | - Moon Young Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Soon Koo Baik
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Young Lim Ham
- Department of Emergency medical technology, Daewon University College, Jecheon, South Korea
| |
Collapse
|
398
|
Shi X, Wei X, Yin X, Wang Y, Zhang M, Zhao C, Zhao H, McClain CJ, Feng W, Zhang X. Hepatic and fecal metabolomic analysis of the effects of Lactobacillus rhamnosus GG on alcoholic fatty liver disease in mice. J Proteome Res 2015; 14:1174-82. [PMID: 25592873 DOI: 10.1021/pr501121c] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The interactions among the gut, liver, and immune system play an important role in liver disease. Probiotics have been used for the treatment and prevention of many pathological conditions, including liver diseases. Comprehensive two-dimensional gas chromatography time-of-flight mass spectrometry (GC×GC-TOF MS) was used herein, in conjunction with chemometric data analysis, to identify metabolites significantly affected by probiotics in mice fed with or without alcohol. The metabolomics analysis indicates that the levels of fatty acids increased in mouse liver and decreased in mouse feces when mice were chronically exposed to alcohol. Supplementing the alcohol-fed mice with culture supernatant from Lactobacillus rhamnosus GG (LGGs) normalized these alcohol-induced abnormalities and prevented alcoholic liver disease (ALD). These results agree well with previous studies. In addition to diet-derived long chain fatty acids (LCFAs), LGGs may positively modify the gut's bacterial population to stimulate LCFA synthesis, which has been shown to enhance intestinal barrier function, reduce endotoxemia, and prevent ALD. We also found that several amino acids, including l-isoleucine, a branched chain amino acid, were downregulated in the liver and fecal samples from animals exposed to alcohol and that the levels of these amino acids were corrected by LGGs. These results demonstrate that LGGs alleviates alcohol-induced fatty liver by mechanisms involving increasing intestinal and decreasing hepatic fatty acids and increasing amino acid concentration.
Collapse
Affiliation(s)
- Xue Shi
- Departments of Chemistry, ‡Pharmacology & Toxicology, §Medicine, and ⊥Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville , Louisville, Kentucky 40292, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
399
|
Abstract
Alcoholic liver disease (ALD) has been among the leading causes of cirrhosis and liver-related death worldwide for decades. Early discoveries in alcoholic liver disease identified increased levels of bacterial endotoxin in the portal circulation, suggesting a role for gut-derived toxins in ALD. Indeed, alcohol consumption can disrupt the intestinal epithelial barrier and result in increased gut permeability that increasingly is recognized as a major factor in ALD. Bacterial endotoxin, lipopolysaccharide, is a prototypic microbe-derived inflammatory signal that contributes to inflammation in ALD through activation of the Toll-like receptor 4. Recent studies also have shown that alcohol consumption is associated with alterations in the gut microbiome, and the dysbalance of pathogenic and commensal organisms in the intestinal microbiome may contribute to the abnormal gut-liver axis in ALD. Indeed, bacterial decontamination improves ALD both in human and animal models. This short review summarizes recent findings and highlights emerging trends in the gut-liver axis relevant to ALD.
Collapse
Affiliation(s)
- Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts.
| |
Collapse
|
400
|
Engen PA, Green SJ, Voigt RM, Forsyth CB, Keshavarzian A. The Gastrointestinal Microbiome: Alcohol Effects on the Composition of Intestinal Microbiota. Alcohol Res 2015; 37:223-36. [PMID: 26695747 PMCID: PMC4590619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The excessive use of alcohol is a global problem causing many adverse pathological health effects and a significant financial health care burden. This review addresses the effect of alcohol consumption on the microbiota in the gastrointestinal tract (GIT). Although data are limited in humans, studies highlight the importance of changes in the intestinal microbiota in alcohol-related disorders. Alcohol-induced changes in the GIT microbiota composition and metabolic function may contribute to the well-established link between alcohol-induced oxidative stress, intestinal hyperpermeability to luminal bacterial products, and the subsequent development of alcoholic liver disease (ALD), as well as other diseases. In addition, clinical and preclinical data suggest that alcohol-related disorders are associated with quantitative and qualitative dysbiotic changes in the intestinal microbiota and may be associated with increased GIT inflammation, intestinal hyperpermeability resulting in endotoxemia, systemic inflammation, and tissue damage/organ pathologies including ALD. Thus, gut-directed interventions, such as probiotic and synbiotic modulation of the intestinal microbiota, should be considered and evaluated for prevention and treatment of alcohol-associated pathologies.
Collapse
|