101
|
Cresci GA, Glueck B, McMullen MR, Xin W, Allende D, Nagy LE. Prophylactic tributyrin treatment mitigates chronic-binge ethanol-induced intestinal barrier and liver injury. J Gastroenterol Hepatol 2017; 32:1587-1597. [PMID: 28087985 PMCID: PMC5511097 DOI: 10.1111/jgh.13731] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 12/09/2016] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Impaired gut-liver axis is a potential factor contributing to alcoholic liver disease. Ethanol depletes intestinal integrity and causes gut dysbiosis. Butyrate, a fermentation byproduct of gut microbiota, is altered negatively following chronic ethanol exposure. This study aimed to determine whether prophylactic tributyrin could protect the intestinal barrier and liver in mice during combined chronic-binge ethanol exposure. METHODS C57BL/6J mice exposed to 5% v/v ethanol-containing diet for 10 days received a single ethanol gavage (5 g/kg) 9 h before euthanasia. Control mice were isocalorically pair-fed maltose dextrin for ethanol. Diets were supplemented (5 mM) with tributyrin or glycerol. Intestine and liver disease activity was assessed histologically. Protein and mRNA expression of tight junction (TJ) proteins, toll-like receptors, and tumor necrosis factor-alpha were assessed. Caco-2 monolayers with or without ethanol exposure and/or sodium butyrate were used to test butyrate's direct effects on intestinal integrity. RESULTS Chronic-binge ethanol feeding impaired intestinal TJ protein co-localization staining; however, tributyrin co-treatment mitigated these effects. Ethanol depleted TJ and transepithelial electrical resistance in Caco-2 monolayers, but butyrate co-treatment reduced these effects. Hepatic toll-like receptor mRNA expression and tumor necrosis factor-alpha protein expression was induced by ethanol; however, the response was significantly dampened in mice co-treated with tributyrin. Tributyrin altered localization of both neutrophils and single hepatocyte death: Leukocytes and apoptotic hepatocytes localized predominantly around the portal tract in ethanol-only treated mice, whereas localization predominated around the central vein in ethanol-tributyrin mice. CONCLUSIONS Prophylactic tributyrin supplementation mitigated effects of combined chronic-binge ethanol exposure on disruption of intestinal TJ localization and intestinal permeability and liver injury.
Collapse
Affiliation(s)
- Gail A Cresci
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Gastroenterology/Hepatology, Cleveland Clinic, Cleveland, Ohio, USA.,Pediatric Research Center, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Bryan Glueck
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Megan R McMullen
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wei Xin
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | | | - Laura E Nagy
- Department of Pathobiology, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
102
|
Methamphetamine-Induced Brain Injury and Alcohol Drinking. J Neuroimmune Pharmacol 2017; 13:53-63. [PMID: 28856500 DOI: 10.1007/s11481-017-9764-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022]
Abstract
A majority of methamphetamine (Meth) abusers also abuse alcohol but the neurochemical consequences of this co-abuse are unknown. Individually, alcohol and Meth cause inflammation and long-term alterations in dopamine and serotonin signaling within the brain. Experiments were conducted to identify if serial exposure to alcohol and Meth has neurochemical consequences that are greater than after either drug alone. Male Sprague Dawley rats voluntarily drank 10% ethanol (EtOH) every other day for 4 weeks and were then exposed to a binge injection regimen of Meth (10 mg/kg injected every 2 h, for a total of 4 injections). EtOH drinking and preference increased over the 4 weeks and caused inflammation evidenced by increases in serum and brain lipopolysaccharide (LPS) and brain cyclooxygenase-2 (COX-2) 24 h after the last day of drinking. Meth alone depleted dopamine and serotonin in the striatum, as well as serotonin in the prefrontal cortex when measured 1 week later. In contrast, EtOH drinking alone did not affect dopamine and serotonin content in the striatum and prefrontal cortex, but prior EtOH drinking followed by injections of Meth enhanced Meth-induced depletions of dopamine, serotonin, as well as dopamine and serotonin transporter immunoreactivities in a manner that was correlated with the degree of EtOH consumption. Cyclooxygenase inhibition by ketoprofen during EtOH drinking blocked the increases in LPS and COX-2 and the enhanced decreases in dopamine and serotonin produced by Meth. Therefore, prior EtOH drinking causes an increase in inflammatory mediators that mediate a synergistic interaction with Meth to cause an enhanced neurotoxicity.
Collapse
|
103
|
Cassard AM, Gérard P, Perlemuter G. Microbiota, Liver Diseases, and Alcohol. Microbiol Spectr 2017; 5:10.1128/microbiolspec.bad-0007-2016. [PMID: 28840806 PMCID: PMC11687517 DOI: 10.1128/microbiolspec.bad-0007-2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Indexed: 02/08/2023] Open
Abstract
Being overweight and obesity are the leading causes of liver disease in Western countries. Liver damage induced by being overweight can range from steatosis, harmless in its simple form, to steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma. Alcohol consumption is an additional major cause of liver disease. Not all individuals who are overweight or excessively consume alcohol develop nonalcoholic fatty liver diseases (NAFLD) or alcoholic liver disease (ALD) and advanced liver disease. The role of the intestinal microbiota (IM) in the susceptibility to liver disease in this context has been the subject of recent studies. ALD and NAFLD appear to be influenced by the composition of the IM, and dysbiosis is associated with ALD and NAFLD in rodent models and human patient cohorts. Several microbial metabolites, such as short-chain fatty acids and bile acids, are specifically associated with dysbiosis. Recent studies have highlighted the causal role of the IM in the development of liver diseases, and the use of probiotics or prebiotics improves some parameters associated with liver disease. Several studies have made progress in deciphering the mechanisms associated with the modulation of the IM. These data have demonstrated the intimate relationship between the IM and metabolic liver disease, suggesting that targeting the gut microbiota could be a new preventive or therapeutic strategy for these diseases.
Collapse
Affiliation(s)
- Anne-Marie Cassard
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud, Université Paris-Saclay, 92140 Clamart, France
| | - Philippe Gérard
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Gabriel Perlemuter
- INSERM U996 Inflammation, Chemokines and Immunopathology, DHU Hepatinov, Univ Paris-Sud, Université Paris-Saclay, 92140 Clamart, France
- AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| |
Collapse
|
104
|
Jarido V, Kennedy L, Hargrove L, Demieville J, Thomson J, Stephenson K, Francis H. The emerging role of mast cells in liver disease. Am J Physiol Gastrointest Liver Physiol 2017; 313:G89-G101. [PMID: 28473331 PMCID: PMC5582878 DOI: 10.1152/ajpgi.00333.2016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/12/2017] [Accepted: 04/24/2017] [Indexed: 01/31/2023]
Abstract
The depth of our knowledge regarding mast cells has widened exponentially in the last 20 years. Once thought to be only important for allergy-mediated events, mast cells are now recognized to be important regulators of a number of pathological processes. The revelation that mast cells can influence organs, tissues, and cells has increased interest in mast cell research during liver disease. The purpose of this review is to refresh the reader's knowledge of the development, type, and location of mast cells and to review recent work that demonstrates the role of hepatic mast cells during diseased states. This review focuses primarily on liver diseases and mast cells during autoimmune disease, hepatitis, fatty liver disease, liver cancer, and aging in the liver. Overall, these studies demonstrate the potential role of mast cells in disease progression.
Collapse
Affiliation(s)
- Veronica Jarido
- Baylor Scott & White Health and Medicine, Temple, Texas; and
| | - Lindsey Kennedy
- Research, Central Texas Veterans Health Care System, Temple, Texas
- Texas A & M Health Science Center, Temple, Texas
| | | | | | - Joanne Thomson
- Research, Central Texas Veterans Health Care System, Temple, Texas
| | | | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, Texas;
- Baylor Scott & White Health and Medicine, Temple, Texas; and
- Texas A & M Health Science Center, Temple, Texas
| |
Collapse
|
105
|
A role for the peripheral immune system in the development of alcohol use disorders? Neuropharmacology 2017; 122:148-160. [DOI: 10.1016/j.neuropharm.2017.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023]
|
106
|
Crescenzo R, Mazzoli A, Di Luccia B, Bianco F, Cancelliere R, Cigliano L, Liverini G, Baccigalupi L, Iossa S. Dietary fructose causes defective insulin signalling and ceramide accumulation in the liver that can be reversed by gut microbiota modulation. Food Nutr Res 2017; 61:1331657. [PMID: 28659742 PMCID: PMC5475320 DOI: 10.1080/16546628.2017.1331657] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 05/09/2017] [Indexed: 12/21/2022] Open
Abstract
Objective: The link between metabolic derangement of the gut-2013liver-visceral white adipose tissue (v-WAT) axis and gut microbiota was investigated. Methods: Rats were fed a fructose-rich diet and treated with an antibiotic mix. Inflammation was measured in portal plasma, ileum, liver, and v-WAT, while insulin signalling was analysed by measuring levels of phosphorylated kinase Akt. The function and oxidative status of hepatic mitochondria and caecal microbiota composition were also evaluated. Results: Ileal inflammation, increase in plasma transaminases, plasma peroxidised lipids, portal concentrations of tumour necrosis factor alpha, lipopolysaccharide, and non-esterified fatty acids, were induced by fructose and were reversed by antibiotic. The increased hepatic ceramide content, inflammation and decreased insulin signaling in liver and v-WAT induced by fructose was reversed by antibiotic. Antibiotic also blunted the increase in hepatic mitochondrial efficiency and oxidative damage of rats fed fructose-rich diet. Three genera, Coprococcus, Ruminococcus, and Clostridium, significantly increased, while the Clostridiaceae family significantly decreased in rats fed a fructose-rich diet, and antibiotic abolished these variations Conclusions: When gut microbiota modulation by fructose is prevented by antibiotic, inflammatory flow from the gut to the liver and v-WAT are reversed.
Collapse
Affiliation(s)
| | - Arianna Mazzoli
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Blanda Di Luccia
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Francesca Bianco
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Rosa Cancelliere
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Luisa Cigliano
- Department of Biology, Federico II University of Naples, Naples, Italy
| | - Giovanna Liverini
- Department of Biology, Federico II University of Naples, Naples, Italy
| | | | - Susanna Iossa
- Department of Biology, Federico II University of Naples, Naples, Italy
| |
Collapse
|
107
|
Crews FT, Walter TJ, Coleman LG, Vetreno RP. Toll-like receptor signaling and stages of addiction. Psychopharmacology (Berl) 2017; 234:1483-1498. [PMID: 28210782 PMCID: PMC5420377 DOI: 10.1007/s00213-017-4560-6] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/03/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Athina Markou and her colleagues discovered persistent changes in adult behavior following adolescent exposure to ethanol or nicotine consistent with increased risk for developing addiction. Building on Dr. Markou's important work and that of others in the field, researchers at the Bowles Center for Alcohol Studies have found that persistent changes in behavior following adolescent stress or alcohol exposure may be linked to induction of immune signaling in brain. AIM This study aims to illuminate the critical interrelationship of the innate immune system (e.g., toll-like receptors [TLRs], high-mobility group box 1 [HMGB1]) in the neurobiology of addiction. METHOD This study reviews the relevant research regarding the relationship between the innate immune system and addiction. CONCLUSION Emerging evidence indicates that TLRs in brain, particularly those on microglia, respond to endogenous innate immune agonists such as HMGB1 and microRNAs (miRNAs). Multiple TLRs, HMGB1, and miRNAs are induced in the brain by stress, alcohol, and other drugs of abuse and are increased in the postmortem human alcoholic brain. Enhanced TLR-innate immune signaling in brain leads to epigenetic modifications, alterations in synaptic plasticity, and loss of neuronal cell populations, which contribute to cognitive and emotive dysfunctions. Addiction involves progressive stages of drug binges and intoxication, withdrawal-negative affect, and ultimately compulsive drug use and abuse. Toll-like receptor signaling within cortical-limbic circuits is modified by alcohol and stress in a manner consistent with promoting progression through the stages of addiction.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - T Jordan Walter
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| |
Collapse
|
108
|
Israel Y, Karahanian E, Ezquer F, Morales P, Ezquer M, Rivera-Meza M, Herrera-Marschitz M, Quintanilla ME. Acquisition, Maintenance and Relapse-Like Alcohol Drinking: Lessons from the UChB Rat Line. Front Behav Neurosci 2017; 11:57. [PMID: 28420969 PMCID: PMC5378819 DOI: 10.3389/fnbeh.2017.00057] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/20/2017] [Indexed: 12/31/2022] Open
Abstract
This review article addresses the biological factors that influence: (i) the acquisition of alcohol intake; (ii) the maintenance of chronic alcohol intake; and (iii) alcohol relapse-like drinking behavior in animals bred for their high-ethanol intake. Data from several rat strains/lines strongly suggest that catalase-mediated brain oxidation of ethanol into acetaldehyde is an absolute requirement (up 80%–95%) for rats to display ethanol’s reinforcing effects and to initiate chronic ethanol intake. Acetaldehyde binds non-enzymatically to dopamine forming salsolinol, a compound that is self-administered. In UChB rats, salsolinol: (a) generates marked sensitization to the motivational effects of ethanol; and (b) strongly promotes binge-like drinking. The specificity of salsolinol actions is shown by the finding that only the R-salsolinol enantiomer but not S-salsolinol accounted for the latter effects. Inhibition of brain acetaldehyde synthesis does not influence the maintenance of chronic ethanol intake. However, a prolonged ethanol withdrawal partly returns the requirement for acetaldehyde synthesis/levels both on chronic ethanol intake and on alcohol relapse-like drinking. Chronic ethanol intake, involving the action of lipopolysaccharide diffusing from the gut, and likely oxygen radical generated upon catechol/salsolinol oxidation, leads to oxidative stress and neuro-inflammation, known to potentiate each other. Data show that the administration of N-acetyl cysteine (NAC) a strong antioxidant inhibits chronic ethanol maintenance by 60%–70%, without inhibiting its initial intake. Intra-cerebroventricular administration of mesenchymal stem cells (MSCs), known to release anti-inflammatory cytokines, to elevate superoxide dismutase levels and to reverse ethanol-induced hippocampal injury and cognitive deficits, also inhibited chronic ethanol maintenance; further, relapse-like ethanol drinking was inhibited up to 85% for 40 days following intracerebral stem cell administration. Thus: (i) ethanol must be metabolized intracerebrally into acetaldehyde, and further into salsolinol, which appear responsible for promoting the acquisition of the early reinforcing effects of ethanol; (ii) acetaldehyde is not responsible for the maintenance of chronic ethanol intake, while other mechanisms are indicated; (iii) the systemic administration of NAC, a strong antioxidant markedly inhibits the maintenance of chronic ethanol intake; and (iv) the intra-cerebroventricular administration of anti-inflammatory and antioxidant MSCs inhibit both the maintenance of chronic ethanol intake and relapse-like drinking.
Collapse
Affiliation(s)
- Yedy Israel
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| | - Eduardo Karahanian
- Center for Biomedical Research, Universidad Autónoma de ChileSantiago, Chile
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del DesarrolloSantiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| | - Marcelo Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del DesarrolloSantiago, Chile
| | - Mario Rivera-Meza
- Department of Pharmacological and Toxicological Chemistry, Faculty of Chemical and Pharmaceutical Sciences, University of ChileSantiago, Chile
| | - Mario Herrera-Marschitz
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| | - María E Quintanilla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of ChileSantiago, Chile
| |
Collapse
|
109
|
Lowe PP, Gyongyosi B, Satishchandran A, Iracheta-Vellve A, Ambade A, Kodys K, Catalano D, Ward DV, Szabo G. Alcohol-related changes in the intestinal microbiome influence neutrophil infiltration, inflammation and steatosis in early alcoholic hepatitis in mice. PLoS One 2017; 12:e0174544. [PMID: 28350851 PMCID: PMC5370121 DOI: 10.1371/journal.pone.0174544] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/10/2017] [Indexed: 12/20/2022] Open
Abstract
Background Alcohol-induced intestinal dysbiosis disrupts homeostatic gut-liver axis function and is essential in the development of alcoholic liver disease. Here, we investigate changes in enteric microbiome composition in a model of early alcoholic steatohepatitis and dissect the pathogenic role of intestinal microbes in alcohol-induced liver pathology. Materials and methods Wild type mice received a 10-day diet that was either 5% alcohol-containing or an isocaloric control diet plus a single binge. 16S rDNA sequencing defined the bacterial communities in the cecum of alcohol- and pair-fed animals. Some mice were treated with an antibiotic cocktail prior to and throughout alcohol feeding. Liver neutrophils, cytokines and steatosis were evaluated. Results Acute-on-chronic alcohol administration induced shifts in various bacterial phyla in the cecum, including increased Actinobacteria and a reduction in Verrucomicrobia driven entirely by a reduction in the genus Akkermansia. Antibiotic treatment reduced the gut bacterial load and circulating bacterial wall component lipopolysaccharide (LPS). We found that bacterial load suppression prevented alcohol-related increases in the number of myeloperoxidase- (MPO) positive infiltrating neutrophils in the liver. Expression of liver mRNA tumor necrosis factor alpha (Tnfα), C-X-C motif chemokine ligand 1 (Cxcl1) and circulating protein monocyte chemoattractant protein-1 (MCP-1) were also reduced in antibiotic-treated alcohol-fed mice. Alcohol-induced hepatic steatosis measured by Oil-Red O staining was significantly reduced in antibiotic treated mice. Genes regulating lipid production and storage were also altered by alcohol and antibiotic treatment. Interestingly, antibiotic treatment did not protect from alcohol-induced increases in serum aminotransferases (ALT/AST). Conclusions Our data indicate that acute-on-chronic alcohol feeding alters the microflora at multiple taxonomic levels and identifies loss of Akkermansia as an early marker of alcohol-induced gut dysbiosis. We conclude that gut microbes influence liver inflammation, neutrophil infiltration and liver steatosis following alcohol consumption and these data further emphasize the role of the gut-liver axis in early alcoholic liver disease.
Collapse
Affiliation(s)
- Patrick P. Lowe
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Benedek Gyongyosi
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Abhishek Satishchandran
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Arvin Iracheta-Vellve
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Aditya Ambade
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Karen Kodys
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Donna Catalano
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Doyle V. Ward
- Center for Microbiome Research, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
110
|
Lu R, Voigt RM, Zhang Y, Kato I, Xia Y, Forsyth CB, Keshavarzian A, Sun J. Alcohol Injury Damages Intestinal Stem Cells. Alcohol Clin Exp Res 2017; 41:727-734. [PMID: 28195397 DOI: 10.1111/acer.13351] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/07/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alcohol consumption is associated with intestinal injury including intestinal leakiness and the risk of developing progressive gastrointestinal cancer. Alcoholics have disruption of intestinal barrier dysfunction that persists weeks after stopping alcohol intake, and this occurs in spite of the fact that intestinal epithelial cells turn over every 3 to 5 days. The renewal and functional regulation of the intestinal epithelium largely relies on intestinal stem cells (ISCs). Chronic inflammation and tissue damage in the intestine can injure stem cells including accumulation of mutations that may result in ISC dysfunction and transformation. ISCs are a key element in intestinal function and pathology; however, very little is known about the effects of alcohol on ISCs. We hypothesize that dysregulation of ISCs is one mechanism by which alcohol induces long-lasting intestinal damage. METHODS In Vivo: Small intestinal samples from alcohol- and control-fed mice were assessed for ISC markers (Lgr5 and Bmi1) and the changes of the β-catenin signaling using immunofluorescent microscopy, Western blotting, and RT-PCR. Ex Vivo: Organoids were generated from small intestine tissue and subsequently exposed to alcohol and analyzed for ISC markers, β-catenin signaling. RESULTS Chronic alcohol consumption significantly decreased the expression of stem cell markers, Bmi1 in the small intestine of the alcohol-fed mice and also resulted in dysregulation of the β-catenin signaling-an essential regulator of its target gene Lgr5 and ISC function. Exposure of small intestine-derived organoids to 0.2% alcohol significantly reduced the growth of the organoids, including budding, and total surface area of the organoid cultures. Alcohol also significantly decreased the expression of Lgr5, p-β-catenin (ser552), and Bmi1 in the organoid model. CONCLUSIONS Both chronic alcohol feeding and acute exposure of alcohol resulted in ISC dysregulation which might be one mechanism for alcohol-induced long-lasting intestinal damage.
Collapse
Affiliation(s)
- Rong Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Robin M Voigt
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Ikuko Kato
- Departments of Oncology and Pathology, Wayne State University School of Medicine, Detroit, Michigan
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Christopher B Forsyth
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Ali Keshavarzian
- Division of Digestive Diseases and Nutrition, Department of Medicine, Rush University Medical Center, Chicago, Illinois
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
111
|
Wells JM, Brummer RJ, Derrien M, MacDonald TT, Troost F, Cani PD, Theodorou V, Dekker J, Méheust A, de Vos WM, Mercenier A, Nauta A, Garcia-Rodenas CL. Homeostasis of the gut barrier and potential biomarkers. Am J Physiol Gastrointest Liver Physiol 2017; 312:G171-G193. [PMID: 27908847 PMCID: PMC5440615 DOI: 10.1152/ajpgi.00048.2015] [Citation(s) in RCA: 410] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 11/09/2016] [Accepted: 11/19/2016] [Indexed: 02/07/2023]
Abstract
The gut barrier plays a crucial role by spatially compartmentalizing bacteria to the lumen through the production of secreted mucus and is fortified by the production of secretory IgA (sIgA) and antimicrobial peptides and proteins. With the exception of sIgA, expression of these protective barrier factors is largely controlled by innate immune recognition of microbial molecular ligands. Several specialized adaptations and checkpoints are operating in the mucosa to scale the immune response according to the threat and prevent overreaction to the trillions of symbionts inhabiting the human intestine. A healthy microbiota plays a key role influencing epithelial barrier functions through the production of short-chain fatty acids (SCFAs) and interactions with innate pattern recognition receptors in the mucosa, driving the steady-state expression of mucus and antimicrobial factors. However, perturbation of gut barrier homeostasis can lead to increased inflammatory signaling, increased epithelial permeability, and dysbiosis of the microbiota, which are recognized to play a role in the pathophysiology of a variety of gastrointestinal disorders. Additionally, gut-brain signaling may be affected by prolonged mucosal immune activation, leading to increased afferent sensory signaling and abdominal symptoms. In turn, neuronal mechanisms can affect the intestinal barrier partly by activation of the hypothalamus-pituitary-adrenal axis and both mast cell-dependent and mast cell-independent mechanisms. The modulation of gut barrier function through nutritional interventions, including strategies to manipulate the microbiota, is considered a relevant target for novel therapeutic and preventive treatments against a range of diseases. Several biomarkers have been used to measure gut permeability and loss of barrier integrity in intestinal diseases, but there remains a need to explore their use in assessing the effect of nutritional factors on gut barrier function. Future studies should aim to establish normal ranges of available biomarkers and their predictive value for gut health in human cohorts.
Collapse
Affiliation(s)
- Jerry M. Wells
- 1Host-Microbe Interactomics, Animal Sciences, Wageningen University, Wageningen, The Netherlands;
| | - Robert J. Brummer
- 2Nutrition-Gut-Brain Interactions Research Centre, School of Medicine and Health, Örebro University, Örebro, Sweden;
| | - Muriel Derrien
- 3Centre Daniel Carasso, Danone Research, Palaiseau, France;
| | - Thomas T. MacDonald
- 4Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Whitechapel, London, United Kingdom;
| | - Freddy Troost
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, University Hospital Maastricht, Maastricht University Medical Centre, Maastricht, The Netherlands;
| | - Patrice D. Cani
- 6Louvain Drug Research Institute, WELBIO (Walloon Excellence in Life Sciences and BIOtechnology), Metabolism and Nutrition Research Group, Université Catholique de Louvain, Brussels, Belgium;
| | - Vassilia Theodorou
- 7Neuro-Gastroenterology and Nutrition Group, Institut National de la Recherche Agronomique, Toulouse, France;
| | - Jan Dekker
- 1Host-Microbe Interactomics, Animal Sciences, Wageningen University, Wageningen, The Netherlands;
| | | | - Willem M. de Vos
- 9Laboratory of Microbiology, Wageningen UR, Wageningen, The Netherlands;
| | - Annick Mercenier
- 10Institute of Nutritional Science, Nestlé Research Center, Lausanne, Switzerland; and
| | - Arjen Nauta
- 11FrieslandCampina, Amersfoort, The Netherlands
| | | |
Collapse
|
112
|
The link between inflammation, bugs, the intestine and the brain in alcohol dependence. Transl Psychiatry 2017; 7:e1048. [PMID: 28244981 PMCID: PMC5545644 DOI: 10.1038/tp.2017.15] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/02/2017] [Accepted: 01/09/2017] [Indexed: 02/08/2023] Open
Abstract
In recent years, some new processes have been proposed to explain how alcohol may influence behavior, psychological symptoms and alcohol seeking in alcohol-dependent subjects. In addition to its important effect on brain and neurotransmitters equilibrium, alcohol abuse also affects peripheral organs including the gut. By yet incompletely understood mechanisms, chronic alcohol abuse increases intestinal permeability and alters the composition of the gut microbiota, allowing bacterial components from the gut lumen to reach the systemic circulation. These gut-derived bacterial products are recognized by immune cells circulating in the blood or residing in target organs, which consequently synthesize and release pro-inflammatory cytokines. Circulating cytokines are considered important mediators of the gut-brain communication, as they can reach the central nervous system and induce neuroinflammation that is associated with change in mood, cognition and drinking behavior. These observations support the possibility that targeting the gut microbiota, by the use of probiotics or prebiotics, could restore the gut barrier function, reduce systemic inflammation and may have beneficial effect in treating alcohol dependence and in reducing alcohol relapse.
Collapse
|
113
|
Monnig MA. Immune activation and neuroinflammation in alcohol use and HIV infection: evidence for shared mechanisms. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2017; 43:7-23. [PMID: 27532935 PMCID: PMC5250549 DOI: 10.1080/00952990.2016.1211667] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/07/2016] [Accepted: 07/07/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Emerging research points to innate immune mechanisms in the neuropathological and behavioral consequences of heavy alcohol use. Alcohol use is common among people living with HIV infection (PLWH), a chronic condition that carries its own set of long-term effects on brain and behavior. Notably, neurobiological and cognitive profiles associated with heavy alcohol use and HIV infection share several prominent features. This observation raises questions about interacting biological mechanisms as well as compounded impairment when HIV infection and heavy drinking co-occur. OBJECTIVE AND METHOD This narrative overview discusses peer-reviewed research on specific immune mechanisms of alcohol that exhibit apparent potential to compound the neurobiological and psychiatric sequelae of HIV infection. These include microbial translocation, systemic immune activation, blood-brain barrier compromise, microglial activation, and neuroinflammation. RESULTS Clinical and preclinical evidence supports overlapping mechanistic actions of HIV and alcohol use on peripheral and neural immune systems. In preclinical studies, innate immune signaling mediates many of the detrimental neurocognitive and behavioral effects of alcohol use. Neuropsychopharmacological research suggests potential for a feed-forward cycle in which heavy drinking induces innate immune signaling, which in turn stimulates subsequent alcohol use behavior. CONCLUSION Alcohol-induced immune activation and neuroinflammation are a serious health concern for PLWH. Future research to investigate specific immune effects of alcohol in the context of HIV infection has potential to identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mollie A. Monnig
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI
| |
Collapse
|
114
|
Samak G, Gangwar R, Meena AS, Rao RG, Shukla PK, Manda B, Narayanan D, Jaggar JH, Rao R. Calcium Channels and Oxidative Stress Mediate a Synergistic Disruption of Tight Junctions by Ethanol and Acetaldehyde in Caco-2 Cell Monolayers. Sci Rep 2016; 6:38899. [PMID: 27958326 PMCID: PMC5153649 DOI: 10.1038/srep38899] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
Ethanol is metabolized into acetaldehyde in most tissues. In this study, we investigated the synergistic effect of ethanol and acetaldehyde on the tight junction integrity in Caco-2 cell monolayers. Expression of alcohol dehydrogenase sensitized Caco-2 cells to ethanol-induced tight junction disruption and barrier dysfunction, whereas aldehyde dehydrogenase attenuated acetaldehyde-induced tight junction disruption. Ethanol up to 150 mM did not affect tight junction integrity or barrier function, but it dose-dependently increased acetaldehyde-mediated tight junction disruption and barrier dysfunction. Src kinase and MLCK inhibitors blocked this synergistic effect of ethanol and acetaldehyde on tight junction. Ethanol and acetaldehyde caused a rapid and synergistic elevation of intracellular calcium. Calcium depletion by BAPTA or Ca2+-free medium blocked ethanol and acetaldehyde-induced barrier dysfunction and tight junction disruption. Diltiazem and selective knockdown of TRPV6 or CaV1.3 channels, by shRNA blocked ethanol and acetaldehyde-induced tight junction disruption and barrier dysfunction. Ethanol and acetaldehyde induced a rapid and synergistic increase in reactive oxygen species by a calcium-dependent mechanism. N-acetyl-L-cysteine and cyclosporine A, blocked ethanol and acetaldehyde-induced barrier dysfunction and tight junction disruption. These results demonstrate that ethanol and acetaldehyde synergistically disrupt tight junctions by a mechanism involving calcium, oxidative stress, Src kinase and MLCK.
Collapse
Affiliation(s)
- Geetha Samak
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Ruchika Gangwar
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Avtar S Meena
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Roshan G Rao
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Pradeep K Shukla
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Bhargavi Manda
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Damodaran Narayanan
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| | - RadhaKrishna Rao
- Department of Physiology, University of Tennessee Health Science Center, 894 Union Avenue, Memphis TN 38163, USA
| |
Collapse
|
115
|
Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, Howles PN, Hokari R, Miura S, Tso P. Antibiotics Suppress Activation of Intestinal Mucosal Mast Cells and Reduce Dietary Lipid Absorption in Sprague-Dawley Rats. Gastroenterology 2016; 151:923-932. [PMID: 27436071 PMCID: PMC5391873 DOI: 10.1053/j.gastro.2016.07.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/28/2016] [Accepted: 07/08/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.
Collapse
Affiliation(s)
- Hirokazu Sato
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Linda S Zhang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristina Martinez
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Qing Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Fei Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip N Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Soichiro Miura
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
116
|
Shu XL, Yu TT, Kang K, Zhao J. Effects of glutamine on markers of intestinal inflammatory response and mucosal permeability in abdominal surgery patients: A meta-analysis. Exp Ther Med 2016; 12:3499-3506. [PMID: 28105083 PMCID: PMC5228558 DOI: 10.3892/etm.2016.3799] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/14/2016] [Indexed: 01/20/2023] Open
Abstract
The present meta-analysis was carried out to determine whether supplementation with glutamine (Gln) would reduce the intestinal inflammatory response and mucosal permeability in patients undergoing abdominal surgery. The PubMed, EMBASE, Web of Science, and The Cochrane Library databases were searched for randomized controlled trials on the effects of supplementation with Gln, and published from August, 1966 to June 2014. Inclusion criteria for the meta-analysis were: i) Study design was a randomized controlled trial, ii) study included patients undergoing abdominal surgery, iii) study patients received a supplementation with Gln peptide (Ala-Gln or Gly-Gln) whereas control patients did not use any supplements, and iv) study outcomes included inflammatory markers [C-reactive protein (CRP), tumor necrosis factor-α (TNF-α), and interleukin (IL)-6, and IL-2 receptor] and markers of intestinal permeability [lactulose/mannitol, diamine oxidase, D(−)lactic acid, and endotoxin]. Qualities of controlled trials were assessed using the Jadad score. Meta-analyses were performed with fixed- or random-effect models depending on the heterogeneity of studies. There were 21 trials meeting the inclusion criteria. The meta-analysis revealed that the levels of CRP, TNF-α, and IL-6 in patients supplemented with Gln were significantly lower than those in control patients, whereas the levels of IL-2 receptor were increased by Gln supplementation. Gln also significantly decreased the lactulose/mannitol ratio, the levels of diamine oxidase and endotoxin, and tended to decrease the levels of cyclic D-lactic acid. In conclusion, Gln appears to effectively reduce the inflammatory response and intestinal mucosal permeability in patients after abdominal surgery.
Collapse
Affiliation(s)
- Xiao-Liang Shu
- Department of Nutrition, Jinshan Hospital, Fudan University School of Medicine, Shanghai 201508, P.R. China
| | - Ting-Ting Yu
- Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Kai Kang
- Tongji University School of Medicine, Shanghai 200092, P.R. China
| | - Jian Zhao
- Tongji University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
117
|
Jones H, Hargrove L, Kennedy L, Meng F, Graf-Eaton A, Owens J, Alpini G, Johnson C, Bernuzzi F, Demieville J, DeMorrow S, Invernizzi P, Francis H. Inhibition of mast cell-secreted histamine decreases biliary proliferation and fibrosis in primary sclerosing cholangitis Mdr2(-/-) mice. Hepatology 2016; 64:1202-1216. [PMID: 27351144 PMCID: PMC5033697 DOI: 10.1002/hep.28704] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 06/11/2016] [Accepted: 06/23/2016] [Indexed: 01/03/2023]
Abstract
UNLABELLED Hepatic fibrosis is marked by activation of hepatic stellate cells (HSCs). Cholestatic injury precedes liver fibrosis, and cholangiocytes interact with HSCs promoting fibrosis. Mast cells (MCs) infiltrate following liver injury and release histamine, increasing biliary proliferation. We evaluated if inhibition of MC-derived histamine decreases biliary proliferation and fibrosis. Wild-type and multidrug resistance 2 knockout mice (9-11 weeks) were treated with cromolyn sodium for 1 week to block MC-derived histamine. Biliary mass and proliferation were evaluated by immunohistochemistry for cytokeratin 19 and Ki-67. Bile flow, bicarbonate excretion, and total bile acids were measured in all mice. Fibrosis was evaluated by sirius red/fast green staining and by quantitative polymerase chain reaction for alpha-smooth muscle actin, fibronectin, collagen type 1a, and transforming growth factor-beta 1. HSC activation was evaluated by quantitative polymerase chain reaction in total liver and immunofluorescent staining in tissues for synaptophysin 9. Histamine serum secretion was measured by enzymatic immunoassay. Mouse liver and human liver samples from control or primary sclerosing cholangitis patients were evaluated for MC markers by quantitative polymerase chain reaction and immunohistochemistry. In vitro, cultured MCs were transfected with histidine decarboxylase short hairpin RNA to decrease histamine secretion and subsequently cocultured with cholangiocytes or HSCs prior to measuring fibrosis markers, proliferation, and transforming growth factor-beta 1 secretion. Treatment with cromolyn sodium decreased biliary proliferation, fibrosis, histamine secretion, and bile flow in multidrug resistance 2 knockout mice. Primary sclerosing cholangitis mice and patients have increased MCs. Knockdown of MC histidine decarboxylase decreased cholangiocyte and HSC proliferation/activation. CONCLUSION MCs are recruited to proliferating cholangiocytes and promote fibrosis. Inhibition of MC-derived histamine decreases fibrosis, and regulation of MC mediators may be therapeutic for primary sclerosing cholangitis. (Hepatology 2016;64:1202-1216).
Collapse
Affiliation(s)
- Hannah Jones
- Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Rozzano, Milan, Italy
| | - Laura Hargrove
- Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Rozzano, Milan, Italy
| | - Lindsey Kennedy
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
- Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Rozzano, Milan, Italy
- Medicine, Texas A&M Health Science Center, Rozzano, Milan, Italy
| | - Allyson Graf-Eaton
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
| | - Jennifer Owens
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
- Medicine, Texas A&M Health Science Center, Rozzano, Milan, Italy
| | | | - Francesca Bernuzzi
- Temple, Texas, USA and Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | | | - Sharon DeMorrow
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
- Medicine, Texas A&M Health Science Center, Rozzano, Milan, Italy
| | - Pietro Invernizzi
- Temple, Texas, USA and Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Rozzano, Milan, Italy
- Scott & White Digestive Disease Research Center, Baylor Scott & White Health, Rozzano, Milan, Italy
- Medicine, Texas A&M Health Science Center, Rozzano, Milan, Italy
| |
Collapse
|
118
|
Zhang ZH, Liu XQ, Zhang C, He W, Wang H, Chen YH, Liu XJ, Chen X, Xu DX. Tlr4-mutant mice are resistant to acute alcohol-induced sterol-regulatory element binding protein activation and hepatic lipid accumulation. Sci Rep 2016; 6:33513. [PMID: 27627966 PMCID: PMC5024165 DOI: 10.1038/srep33513] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Accepted: 08/30/2016] [Indexed: 12/20/2022] Open
Abstract
Previous studies demonstrated that acute alcohol intoxication caused hepatic lipid accumulation. The present study showed that acute alcohol intoxication caused hepatic lipid accumulation in Tlr4-wild-type mice but not in Tlr4-mutant mice. Hepatic sterol-regulatory element binding protein (SREBP)-1, a transcription factor regulating fatty acid and triglyceride (TG) synthesis, was activated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Hepatic Fas, Acc, Scd-1 and Dgat-2, the key genes for fatty acid and TG synthesis, were up-regulated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Additional experiment showed that hepatic MyD88 was elevated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Hepatic NF-κB was activated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Moreover, hepatic GSH content was reduced and hepatic MDA level was elevated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Hepatic CYP2E1 was elevated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Hepatic p67phox and gp91phox, two NADPH oxidase subunits, were up-regulated in alcohol-treated Tlr4-wild-type mice but not in Tlr4-mutant mice. Alpha-phenyl-N-t-butylnitrone (PBN), a free radical spin-trapping agent, protected against alcohol-induced hepatic SREBP-1 activation and hepatic lipid accumulation. In conclusion, Tlr4-mutant mice are resistant to acute alcohol-induced hepatic SREBP-1 activation and hepatic lipid accumulation.
Collapse
Affiliation(s)
- Zhi-Hui Zhang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Qian Liu
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Wei He
- First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Yuan-Hua Chen
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Xiao-Jing Liu
- First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - Xi Chen
- First Affiliated Hospital, Anhui Medical University, Hefei, 230032, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
119
|
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
|
120
|
Park B, Lee HR, Lee YJ. Alcoholic liver disease: focus on prodromal gut health. J Dig Dis 2016; 17:493-500. [PMID: 27356233 DOI: 10.1111/1751-2980.12375] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/02/2016] [Accepted: 06/19/2016] [Indexed: 12/11/2022]
Abstract
Alcoholic liver disease (ALD) is implicated in gut disturbances, both functionally and structurally. It has been noticed that the gut-liver interaction is an important feature in the prevention of systemic inflammation as well as liver health. The optimal functioning of the gut-liver axis depends on gut health. Therefore, gut problems may be important for estimating liver inflammation, while our knowledge of ALD could also provide an insight into gut health. Gut problems accompanied by ALD include gut motility and absorption problems, mucosal damage and the dysbiosis of gut microbiota and gastrointestinal carcinogenesis. Moreover, there is emerging evidence that besides direct inflammatory injury caused by alcohol, gut problems related to ALD play a crucial role in the pathogenesis of cardiovascular and immunological disorders. In this regard, we should consider ALD in relation to both gut health and chronic systemic low-grade inflammation. Accordingly, integrative therapeutic strategies are warranted for treating and preventing ALD and systemic inflammation as well as alcohol-related gut problems.
Collapse
Affiliation(s)
- Byoungjin Park
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Ree Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Jae Lee
- Department of Family Medicine, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
121
|
Chen P, Miyamoto Y, Mazagova M, Lee KC, Eckmann L, Schnabl B. Microbiota and Alcoholic Liver Disease. Alcohol Clin Exp Res 2016; 40:1791-2. [PMID: 27364225 DOI: 10.1111/acer.13129] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 05/16/2016] [Indexed: 01/13/2023]
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
|
122
|
Neyrinck AM, Etxeberria U, Taminiau B, Daube G, Van Hul M, Everard A, Cani PD, Bindels LB, Delzenne NM. Rhubarb extract prevents hepatic inflammation induced by acute alcohol intake, an effect related to the modulation of the gut microbiota. Mol Nutr Food Res 2016; 61. [PMID: 26990039 DOI: 10.1002/mnfr.201500899] [Citation(s) in RCA: 129] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/26/2016] [Accepted: 03/14/2016] [Indexed: 12/17/2022]
Abstract
SCOPE Binge consumption of alcohol is an alarming global health problem. Acute ethanol intoxication is characterized by hepatic inflammation and oxidative stress, which could be promoted by gut barrier function alterations. In this study, we have tested the hypothesis of the hepatoprotective effect of rhubarb extract in a mouse model of binge drinking and we explored the contribution of the gut microbiota in the related metabolic effects. METHODS AND RESULTS Mice were fed a control diet supplemented with or without 0.3% rhubarb extract for 17 days and were necropsied 6 h after an alcohol challenge. Supplementation with rhubarb extract changed the microbial ecosystem (assessed by 16S rDNA pyrosequencing) in favor of Akkermansia muciniphila and Parabacteroides goldsteinii. Furthermore, it improved alcohol-induced hepatic injury, downregulated key markers of both inflammatory and oxidative stresses in the liver tissue, without affecting significantly steatosis. In the gut, rhubarb supplementation increased crypt depth, tissue weight, and the expression of antimicrobial peptides. CONCLUSIONS These findings suggest that some bacterial genders involved in gut barrier function, are promoted by phytochemicals present in rhubarb extract, and could therefore be involved in the modulation of the susceptibility to hepatic diseases linked to acute alcohol consumption.
Collapse
Affiliation(s)
- Audrey M Neyrinck
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium
| | - Usune Etxeberria
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium.,Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain
| | - Bernard Taminiau
- Fundamental and Applied Research for Animal and Health-Department of Food Sciences, Université de Liège, Liège, Belgium
| | - Georges Daube
- Fundamental and Applied Research for Animal and Health-Department of Food Sciences, Université de Liège, Liège, Belgium
| | - Matthias Van Hul
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Brussels, Belgium
| | - Amandine Everard
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Brussels, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium.,Walloon Excellence in Life sciences and BIOtechnology (WELBIO), Louvain Drug Research Institute, Brussels, Belgium
| | - Laure B Bindels
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Louvain Drug Research Institute, Metabolism and Nutrition Research Group, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
123
|
Llopis M, Cassard AM, Wrzosek L, Boschat L, Bruneau A, Ferrere G, Puchois V, Martin JC, Lepage P, Le Roy T, Lefèvre L, Langelier B, Cailleux F, González-Castro AM, Rabot S, Gaudin F, Agostini H, Prévot S, Berrebi D, Ciocan D, Jousse C, Naveau S, Gérard P, Perlemuter G. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut 2016; 65:830-9. [PMID: 26642859 DOI: 10.1136/gutjnl-2015-310585] [Citation(s) in RCA: 410] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/01/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE There is substantial inter-individual diversity in the susceptibility of alcoholics to liver injury. Alterations of intestinal microbiota (IM) have been reported in alcoholic liver disease (ALD), but the extent to which they are merely a consequence or a cause is unknown. We aimed to demonstrate that a specific dysbiosis contributes to the development of alcoholic hepatitis (AH). DESIGN We humanised germ-free and conventional mice using human IM transplant from alcoholic patients with or without AH. The consequences on alcohol-fed recipient mice were studied. RESULTS A specific dysbiosis was associated with ALD severity in patients. Mice harbouring the IM from a patient with severe AH (sAH) developed more severe liver inflammation with an increased number of liver T lymphocyte subsets and Natural Killer T (NKT) lymphocytes, higher liver necrosis, greater intestinal permeability and higher translocation of bacteria than mice harbouring the IM from an alcoholic patient without AH (noAH). Similarly, CD45+ lymphocyte subsets were increased in visceral adipose tissue, and CD4(+)T and NKT lymphocytes in mesenteric lymph nodes. The IM associated with sAH and noAH could be distinguished by differences in bacterial abundance and composition. Key deleterious species were associated with sAH while the Faecalibacterium genus was associated with noAH. Ursodeoxycholic acid was more abundant in faeces from noAH mice. Additionally, in conventional mice humanised with the IM from an sAH patient, a second subsequent transfer of IM from an noAH patient improved alcohol-induced liver lesions. CONCLUSIONS Individual susceptibility to ALD is substantially driven by IM. It may, therefore, be possible to prevent and manage ALD by IM manipulation.
Collapse
Affiliation(s)
- M Llopis
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - A M Cassard
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - L Wrzosek
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - L Boschat
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - A Bruneau
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - G Ferrere
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - V Puchois
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - J C Martin
- Faculté de Médecine, Aix-Marseille University, Marseille, France INSERM, UMR1062 NORT, Marseille, France
| | - P Lepage
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - T Le Roy
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - L Lefèvre
- INRA, UMR 1313, GABI-LGS Plateforme ICE, Jouy-en-Josas, France
| | - B Langelier
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - F Cailleux
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France
| | - A M González-Castro
- Department of Gastroenterology, Digestive System Research Unit, Hospital University Vall d'Hebron and VHIR, UAB, Spain
| | - S Rabot
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - F Gaudin
- IPSIT, IFR141, Faculté de Pharmacie, Univ Paris-Sud, Châtenay-Malabry, France
| | - H Agostini
- AP-HP, Hôpital Bicêtre, Unité de recherche clinique Paris-Sud, Kremlin-Bicêtre, France
| | - S Prévot
- Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France AP-HP, Anatomie-pathologique, Hôpital Antoine-Béclère, Clamart, France
| | - D Berrebi
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France AP-HP, Anatomie et de Cytologie Pathologiques, Hôpital Robert Debré, Paris, France
| | - D Ciocan
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - C Jousse
- Université Clermont Auvergne, Institut de Chimie de Clermont-Ferrand UMR CNRS 6296, Clermont-Ferrand, France
| | - S Naveau
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| | - P Gérard
- INRA, UMR1319 Micalis, Jouy-en-Josas, France AgroParisTech, UMR Micalis, Jouy-en-Josas, France
| | - G Perlemuter
- INSERM UMR996-Inflammation, Chemokines and Immunopathology, Clamart, France Univ Paris-Sud, Univ Paris-Saclay, DHU Hepatinov, Labex Lermit, CHU Bicêtre, Kremlin-Bicêtre, France AP-HP, Hepatogastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
| |
Collapse
|
124
|
Abstract
RATIONALE Alcoholism is a primary, chronic relapsing disease of brain reward, motivation, memory, and related circuitry. It is characterized by an individual's continued drinking despite negative consequences related to alcohol use, which is exemplified by alcohol use leading to clinically significant impairment or distress. Chronic alcohol consumption increases the expression of innate immune signaling molecules (ISMs) in the brain that alter cognitive processes and promote alcohol drinking. OBJECTIVES Unraveling the mechanisms of alcohol-induced neuroimmune gene induction is complicated by positive loops of multiple cytokines and other signaling molecules that converge on nuclear factor kappa-light-chain-enhancer of activated B cells and activator protein-1 leading to induction of additional neuroimmune signaling molecules that amplify and expand the expression of ISMs. RESULTS Studies from our laboratory employing reverse transcription polymerase chain reaction (RT-PCR) to assess mRNA, immunohistochemistry and Western blot analysis to assess protein expression, and others suggest that ethanol increases brain neuroimmune gene and protein expression through two distinct mechanisms involving (1) systemic induction of innate immune molecules that are transported from blood to the brain and (2) the direct release of high-mobility group box 1 (HMGB1) from neurons in the brain. Released HMGB1 signals through multiple receptors, particularly Toll-like receptor (TLR) 4, that potentiate cytokine receptor responses leading to a hyperexcitable state that disrupts neuronal networks and increases excitotoxic neuronal death. Innate immune gene activation in brain is persistent, consistent with the chronic relapsing disease that is alcoholism. Expression of HMGB1, TLRs, and other ISMs is increased several-fold in the human orbital frontal cortex, and expression of these molecules is highly correlated with each other as well as lifetime alcohol consumption and age of drinking onset. CONCLUSIONS The persistent and cumulative nature of alcohol on HMGB1 and TLR gene induction support their involvement in alcohol-induced long-term changes in brain function and neurodegeneration.
Collapse
Affiliation(s)
- Fulton T Crews
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA.
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill, School of Medicine, CB# 7178, 1021 Thurston-Bowles Building, Chapel Hill, NC, 27599-7178, USA
| |
Collapse
|
125
|
Mast cells in gastrointestinal disorders. Eur J Pharmacol 2016; 778:139-45. [DOI: 10.1016/j.ejphar.2016.02.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/25/2016] [Accepted: 02/03/2016] [Indexed: 01/01/2023]
|
126
|
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
|
127
|
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
|
128
|
Bomhof MR, Paul HA, Geuking MB, Eller LK, Reimer RA. Improvement in adiposity with oligofructose is modified by antibiotics in obese rats. FASEB J 2016; 30:2720-32. [PMID: 27059718 DOI: 10.1096/fj.201600151r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
Given the intimate link between gut microbiota and host physiology, there is growing interest in understanding the mechanisms by which diet influences gut microbiota and affects human metabolic health. Using antibiotics and the prebiotic oligofructose, which has been shown to counteract excess fat mass, we explored the gut microbiota-dependent effects of oligofructose on body composition and host metabolism. Diet-induced obese male Sprague Dawley rats, fed a background high-fat/sucrose diet, were randomized to one of the following diets for 6 wk: 1) high-energy control; 2) 10% oligofructose; 3) ampicillin; 4) ampicillin + 10% oligofructose; 5) ampicillin/neomycin; or 6) ampicillin/neomycin + 10% oligofructose. Combining oligofructose with ampicillin treatment blunted the decrease in adiposity seen with oligofructose. Although ampicillin did not affect total bacteria, ampicillin impeded oligofructose-induced increases in Bifidobacterium and Lactobacillus In contrast, the combination of ampicillin and neomycin reduced total bacteria but did not abrogate the oligofructose-induced decrease in adiposity. Oligofructose-mediated effects on host adiposity and metabolic health appear to be in part dependent on the presence of specific microbial species within the gut.-Bomhof, M. R., Paul, H. A., Geuking, M. B., Eller, L. K., Reimer, R. A. Improvement in adiposity with oligofructose is modified by antibiotics in obese rats.
Collapse
Affiliation(s)
- Marc R Bomhof
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta Canada
| | - Heather A Paul
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta Canada; and
| | - Markus B Geuking
- Division of Gastroenterology, Department of Clinical Research, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - Lindsay K Eller
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta Canada
| | - Raylene A Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta Canada; and
| |
Collapse
|
129
|
Singh V, Yeoh BS, Chassaing B, Zhang B, Saha P, Xiao X, Awasthi D, Shashidharamurthy R, Dikshit M, Gewirtz A, Vijay-Kumar M. Microbiota-inducible Innate Immune, Siderophore Binding Protein Lipocalin 2 is Critical for Intestinal Homeostasis. Cell Mol Gastroenterol Hepatol 2016; 2:482-498.e6. [PMID: 27458605 PMCID: PMC4957954 DOI: 10.1016/j.jcmgh.2016.03.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Lipocalin 2 (Lcn2) is a multifunctional innate immune protein whose expression closely correlates with extent of intestinal inflammation. However, whether Lcn2 plays a role in the pathogenesis of gut inflammation is unknown. Herein, we investigated the extent to which Lcn2 regulates inflammation and gut bacterial dysbiosis in mouse models of IBD. METHODS Lcn2 expression was monitored in murine colitis models and upon microbiota ablation/restoration. WT and Lcn2 knockout (Lcn2KO) mice were analyzed for gut bacterial load, composition by 16S rRNA gene pyrosequencing and, their colitogenic potential by co-housing with Il-10KO mice. Acute (dextran sodium sulfate) and chronic (IL-10R neutralization and T-cell adoptive transfer) colitis was induced in WT and Lcn2KO mice with or without antibiotics. RESULTS Lcn2 expression was dramatically induced upon inflammation and was dependent upon presence of a gut microbiota and MyD88 signaling. Use of bone-marrow chimeric mice revealed non-immune cells are the major contributors of circulating Lcn2. Lcn2KO mice exhibited elevated levels of entA-expressing gut bacteria burden and, moreover, a broadly distinct bacterial community relative to WT littermates. Lcn2KO mice developed highly colitogenic T-cells and exhibited exacerbated colitis upon exposure to DSS or neutralization of IL-10. Such exacerbated colitis could be prevented by antibiotic treatment. Moreover, exposure to the microbiota of Lcn2KO mice, via cohousing, resulted in severe colitis in Il-10KO mice. CONCLUSION Lcn2 is a bacterially-induced, MyD88-dependent, protein that play an important role in gut homeostasis and a pivotal role upon challenge. Hence, therapeutic manipulation of Lcn2 levels may provide a strategy to help manage diseases driven by alteration of the gut microbiota.
Collapse
Affiliation(s)
- Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Beng San Yeoh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Benoit Chassaing
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Benyue Zhang
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Piu Saha
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Xia Xiao
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania
| | - Deepika Awasthi
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | | | - Madhu Dikshit
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Andrew Gewirtz
- Center for Inflammation, Immunity and Infection, Institute of Biomedical Sciences, Georgia State University, Atlanta, Georgia
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, Pennsylvania,Department of Medicine, The Pennsylvania State University Medical Center, Hershey, Pennsylvania,Correspondence Address correspondence to: Matam Vijay-Kumar, PhD, Department of Nutritional Sciences 222, Chandlee Laboratory, The Pennsylvania State University, University Park, Pennsylvania 16802. fax: (814) 863-6103.Department of Nutritional Sciences 222Chandlee LaboratoryThe Pennsylvania State UniversityUniversity ParkPennsylvania 16802
| |
Collapse
|
130
|
Beceiro C, Campos J, Valcarcel MA, Fenger RV, Lojo S, Linneberg A, Vidal C, Gonzalez-Quintela A. Serum concentrations of mast cell tryptase are reduced in heavy drinkers. Alcohol Clin Exp Res 2016; 39:672-8. [PMID: 25833028 DOI: 10.1111/acer.12682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/19/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Baseline serum tryptase concentrations are commonly used in clinical practice as a marker of the body's mast cell burden. This study aimed to investigate serum tryptase concentrations in heavy drinkers. METHODS Serum tryptase concentrations were determined in 126 heavy drinkers (75% males, median age 47 years) who were admitted to the hospital because of alcohol withdrawal syndrome (n = 60), general symptoms with abnormalities on biochemical tests that indicated acute liver disease (n = 19), complications of advanced liver disease (n = 33), and miscellaneous reasons (n = 14). Results were compared with those of 70 healthy controls (66% males, median age 40 years). RESULTS Serum tryptase concentrations were lower in heavy drinkers than in healthy controls (median 2.23 μg/l vs. median 3.25 μg/l, p < 0.001). Ten heavy drinkers (7.9%) had undetectable (<1 μg/l) serum tryptase levels versus none of the healthy controls (p = 0.01). The association of low tryptase levels with heavy drinking was independent of age, gender, and smoking status. Among heavy drinkers, the lowest tryptase concentrations were observed in patients with alcohol withdrawal syndrome and patients with general symptoms with abnormalities on biochemical tests that indicated acute liver disease. Furthermore, serum tryptase concentrations were negatively correlated with markers of acute liver damage or alcohol consumption (serum aspartate aminotransferase and gamma-glutamyl transferase). Atopy (skin prick test positivity) was not associated with serum tryptase concentrations in heavy drinkers. CONCLUSIONS Serum concentrations of mast cell tryptase are lower in heavy drinkers than in healthy controls.
Collapse
Affiliation(s)
- Carmen Beceiro
- Department of Internal Medicine, Hospital Clinico Universitario, Santiago de Compostela, Spain
| | | | | | | | | | | | | | | |
Collapse
|
131
|
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
|
132
|
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
|
133
|
De Santis S, Cavalcanti E, Mastronardi M, Jirillo E, Chieppa M. Nutritional Keys for Intestinal Barrier Modulation. Front Immunol 2015; 6:612. [PMID: 26697008 PMCID: PMC4670985 DOI: 10.3389/fimmu.2015.00612] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/22/2015] [Indexed: 12/14/2022] Open
Abstract
The intestinal tract represents the largest interface between the external environment and the human body. Nutrient uptake mostly happens in the intestinal tract, where the epithelial surface is constantly exposed to dietary antigens. Since inflammatory response toward these antigens may be deleterious for the host, a plethora of protective mechanisms take place to avoid or attenuate local damage. For instance, the intestinal barrier is able to elicit a dynamic response that either promotes or impairs luminal antigens adhesion and crossing. Regulation of intestinal barrier is crucial to control intestinal permeability whose increase is associated with chronic inflammatory conditions. The cross talk among bacteria, immune, and dietary factors is able to modulate the mucosal barrier function, as well as the intestinal permeability. Several nutritional products have recently been proposed as regulators of the epithelial barrier, even if their effects are in part contradictory. At the same time, the metabolic function of the microbiota generates new products with different effects based on the dietary content. Besides conventional treatments, novel therapies based on complementary nutrients are now growing. Fecal therapy has been recently used for the clinical treatment of refractory Clostridium difficile infection instead of the classical antibiotic therapy. In the present review, we will outline the epithelial response to nutritional components derived from dietary intake and microbial fermentation focusing on the consequent effects on the integrity of the epithelial barrier.
Collapse
Affiliation(s)
- Stefania De Santis
- Laboratory of Experimental Immunopathology, IRCCS "De Bellis" , Castellana Grotte , Italy
| | - Elisabetta Cavalcanti
- Laboratory of Experimental Immunopathology, IRCCS "De Bellis" , Castellana Grotte , Italy
| | - Mauro Mastronardi
- Department of Gastroenterology, IRCCS "De Bellis" , Castellana Grotte , Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari , Bari , Italy
| | - Marcello Chieppa
- Laboratory of Experimental Immunopathology, IRCCS "De Bellis" , Castellana Grotte , Italy ; Istituto Comprensivo Bregante-Volta , Monopoli , Italy
| |
Collapse
|
134
|
Chassaing B, Miles-Brown J, Pellizzon M, Ulman E, Ricci M, Zhang L, Patterson AD, Vijay-Kumar M, Gewirtz AT. Lack of soluble fiber drives diet-induced adiposity in mice. Am J Physiol Gastrointest Liver Physiol 2015; 309:G528-41. [PMID: 26185332 PMCID: PMC4593822 DOI: 10.1152/ajpgi.00172.2015] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 07/08/2015] [Indexed: 01/31/2023]
Abstract
Diet-induced obesity is often modeled by comparing mice fed high-fat diet (HFD), which is made from purified ingredients, vs. normal chow diet (NCD), which is a low-fat assemblage of relatively unrefined plant and animal products. The mechanism by which HFD promotes adiposity is complex but thought to involve low-grade inflammation and altered gut microbiota. The goal of this study was to investigate the extent to which HFD-induced adiposity is driven by fat content vs. other factors that differentiate HFD vs. NCD. Mice were fed NCD, HFD, or other compositionally defined diets (CDD), designed to mimic NCD and/or explore the role of HFD components. A range of metabolic parameters reflecting low-grade inflammation and adiposity were assayed. Relative to NCD, HFD, and to a lesser, but, nonetheless, significant extent, CDD induced increased adiposity, indicating both lipid content and other aspects of HFD are obesogenic. Moreover, HFD and CDD induced a rapid and marked loss of cecal and colonic mass. Such CDD-induced effects were not affected by adjusting dietary protein levels/types but could be largely eliminated by exchanging insoluble fiber (cellulose) for soluble fiber (inulin). Replacing cellulose with inulin in HFD also protected mice against decreased intestinal mass, hyperphagia, and increased adiposity. Such beneficial effects of inulin were microbiota dependent, correlated with elevated fecal short-chain fatty acid levels analyzed via (1)H-NMR-based metabolomics and were partially recapitulated by administration of short-chain fatty acid. HFD-induced obesity is strongly promoted by its lack of soluble fiber, which supports microbiota-mediated intestinal tissue homeostasis that prevents inflammation driving obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Benoit Chassaing
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| | - Jennifer Miles-Brown
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| | | | - Edward Ulman
- 2Research Diets, Inc., New Brunswick, New Jersey;
| | | | - Limin Zhang
- 3Departments of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania; and
| | - Andrew D. Patterson
- 3Departments of Veterinary and Biomedical Sciences, Pennsylvania State University, University Park, Pennsylvania; and
| | - Matam Vijay-Kumar
- 4Departments of Nutritional Sciences & Medicine, Pennsylvania State University, University Park, Pennsylvania
| | - Andrew T. Gewirtz
- 1Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia;
| |
Collapse
|
135
|
Murai A, Kitahara K, Okumura S, Kobayashi M, Horio F. Oral antibiotics enhance antibody responses to keyhole limpet hemocyanin in orally but not muscularly immunized chickens. Anim Sci J 2015; 87:257-65. [DOI: 10.1111/asj.12424] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 02/15/2015] [Accepted: 02/19/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Atsushi Murai
- Laboratory of Animal Nutrition, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural SciencesNagoya University Nagoya Japan
| | - Kazuki Kitahara
- Laboratory of Animal Nutrition, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural SciencesNagoya University Nagoya Japan
| | - Shouta Okumura
- Laboratory of Animal Nutrition, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural SciencesNagoya University Nagoya Japan
| | - Misato Kobayashi
- Laboratory of Animal Nutrition, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural SciencesNagoya University Nagoya Japan
| | - Fumihiko Horio
- Laboratory of Animal Nutrition, Department of Applied Molecular Biosciences, Graduate School of Bioagricultural SciencesNagoya University Nagoya Japan
| |
Collapse
|
136
|
Di Luccia B, Crescenzo R, Mazzoli A, Cigliano L, Venditti P, Walser JC, Widmer A, Baccigalupi L, Ricca E, Iossa S. Rescue of Fructose-Induced Metabolic Syndrome by Antibiotics or Faecal Transplantation in a Rat Model of Obesity. PLoS One 2015; 10:e0134893. [PMID: 26244577 PMCID: PMC4526532 DOI: 10.1371/journal.pone.0134893] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/15/2015] [Indexed: 02/01/2023] Open
Abstract
A fructose-rich diet can induce metabolic syndrome, a combination of health disorders that increases the risk of diabetes and cardiovascular diseases. Diet is also known to alter the microbial composition of the gut, although it is not clear whether such alteration contributes to the development of metabolic syndrome. The aim of this work was to assess the possible link between the gut microbiota and the development of diet-induced metabolic syndrome in a rat model of obesity. Rats were fed either a standard or high-fructose diet. Groups of fructose-fed rats were treated with either antibiotics or faecal samples from control rats by oral gavage. Body composition, plasma metabolic parameters and markers of tissue oxidative stress were measured in all groups. A 16S DNA-sequencing approach was used to evaluate the bacterial composition of the gut of animals under different diets. The fructose-rich diet induced markers of metabolic syndrome, inflammation and oxidative stress, that were all significantly reduced when the animals were treated with antibiotic or faecal samples. The number of members of two bacterial genera, Coprococcus and Ruminococcus, was increased by the fructose-rich diet and reduced by both antibiotic and faecal treatments, pointing to a correlation between their abundance and the development of the metabolic syndrome. Our data indicate that in rats fed a fructose-rich diet the development of metabolic syndrome is directly correlated with variations of the gut content of specific bacterial taxa.
Collapse
Affiliation(s)
- Blanda Di Luccia
- Department of Biology, University “Federico II” of Naples, Naples, Italy
| | | | - Arianna Mazzoli
- Department of Biology, University “Federico II” of Naples, Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University “Federico II” of Naples, Naples, Italy
| | - Paola Venditti
- Department of Biology, University “Federico II” of Naples, Naples, Italy
| | | | - Alex Widmer
- Institute of Integrative Biology (IBZ), ETH Zurich, Zurich, Switzerland
| | | | - Ezio Ricca
- Department of Biology, University “Federico II” of Naples, Naples, Italy
| | - Susanna Iossa
- Department of Biology, University “Federico II” of Naples, Naples, Italy
- * E-mail:
| |
Collapse
|
137
|
Wang XP, Lei F, Du F, Chai YS, Jiang JF, Wang YG, Yu X, Yan XJ, Xing DM, Du LJ. Protection of Gastrointestinal Mucosa from Acute Heavy Alcohol Consumption: The Effect of Berberine and Its Correlation with TLR2, 4/IL1β-TNFα Signaling. PLoS One 2015. [PMID: 26226164 PMCID: PMC4520689 DOI: 10.1371/journal.pone.0134044] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The purpose of the present study is to confirm the protective effect of berberine (BBR) on gastrointestinal injury caused by acute heavy alcohol exposure, an effect that has not been reported previously. Our research details how BBR protects against gastrointestinal injuries from acute alcohol exposure using both in vivo and in vitro experiments. Acute high alcohol concentrations lead to obvious damage to the gastrointestinal mucosa, resulting in necrosis of the intestinal mucosa. Oral administration of BBR was able to significantly reduce this alcohol-induced damage, inhibit increases of alcohol-induced TNFα and IL-1β expression in gastrointestinal mucosa as well as their upstream signals TLR2 and TLR4, and regulate cytokines that modulate tight junctions. Alcohol consumption is a popular human social behavior worldwide, and the present study reports a comprehensive mechanism by which BBR protects against gastrointestinal injuries from alcohol stress, providing people with a novel application of BBR.
Collapse
Affiliation(s)
- Xin-Pei Wang
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fan Lei
- School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Feng Du
- Department of Mathematics, Tulane University, New Orleans, LA, 70118, United States of America
| | - Yu-Shuang Chai
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jing-Fei Jiang
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yu-Gang Wang
- MD Anderson Cancer Center, University of Texas, Houston, Texas, 77030, United States of America
| | - Xuan Yu
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiao-Jin Yan
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Dong-Ming Xing
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Li-Jun Du
- MOE Key Laboratory of Protein Sciences, Laboratory of Molecular Pharmacology and Pharmaceutical Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
- * E-mail:
| |
Collapse
|
138
|
Chaudhry KK, Samak G, Shukla PK, Mir H, Gangwar R, Manda B, Isse T, Kawamoto T, Salaspuro M, Kaihovaara P, Dietrich P, Dragatsis I, Nagy LE, Rao RK. ALDH2 Deficiency Promotes Ethanol-Induced Gut Barrier Dysfunction and Fatty Liver in Mice. Alcohol Clin Exp Res 2015; 39:1465-75. [PMID: 26173414 DOI: 10.1111/acer.12777] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Acetaldehyde, the toxic ethanol (EtOH) metabolite, disrupts intestinal epithelial barrier function. Aldehyde dehydrogenase (ALDH) detoxifies acetaldehyde into acetate. Subpopulations of Asians and Native Americans show polymorphism with loss-of-function mutations in ALDH2. We evaluated the effect of ALDH2 deficiency on EtOH-induced disruption of intestinal epithelial tight junctions and adherens junctions, gut barrier dysfunction, and liver injury. METHODS Wild-type and ALDH2-deficient mice were fed EtOH (1 to 6%) in Lieber-DeCarli diet for 4 weeks. Gut permeability in vivo was measured by plasma-to-luminal flux of FITC-inulin, tight junction and adherens junction integrity was analyzed by confocal microscopy, and liver injury was assessed by the analysis of plasma transaminase activity, histopathology, and liver triglyceride. RESULTS EtOH feeding elevated colonic mucosal acetaldehyde, which was significantly greater in ALDH2-deficient mice. ALDH2(-/-) mice showed a drastic reduction in the EtOH diet intake. Therefore, this study was continued only in wild-type and ALDH2(+/-) mice. EtOH feeding elevated mucosal inulin permeability in distal colon, but not in proximal colon, ileum, or jejunum of wild-type mice. In ALDH2(+/-) mice, EtOH-induced inulin permeability in distal colon was not only higher than that in wild-type mice, but inulin permeability was also elevated in the proximal colon, ileum, and jejunum. Greater inulin permeability in distal colon of ALDH2(+/-) mice was associated with a more severe redistribution of tight junction and adherens junction proteins from the intercellular junctions. In ALDH2(+/-) mice, but not in wild-type mice, EtOH feeding caused a loss of junctional distribution of tight junction and adherens junction proteins in the ileum. Histopathology, plasma transaminases, and liver triglyceride analyses showed that EtOH-induced liver damage was significantly greater in ALDH2(+/-) mice compared to wild-type mice. CONCLUSIONS These data demonstrate that ALDH2 deficiency enhances EtOH-induced disruption of intestinal epithelial tight junctions, barrier dysfunction, and liver damage.
Collapse
Affiliation(s)
| | - Geetha Samak
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Pradeep K Shukla
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Hina Mir
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Ruchika Gangwar
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Bhargavi Manda
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Toyohi Isse
- University of Occupational and Environmental Health , Kitakyushu, Japan
| | | | - Mikko Salaspuro
- Res Unit Acetaldehyde and Cancer, University of Helsinki, Helsinki, Finland
| | - Pertti Kaihovaara
- Res Unit Acetaldehyde and Cancer, University of Helsinki, Helsinki, Finland
| | - Paula Dietrich
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | - Ioannis Dragatsis
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| | | | - Radha Krishna Rao
- Department of Physiology , University of Tennessee, Memphis, Tennessee
| |
Collapse
|
139
|
Afrin LB, Khoruts A. Mast Cell Activation Disease and Microbiotic Interactions. Clin Ther 2015; 37:941-53. [PMID: 25773459 DOI: 10.1016/j.clinthera.2015.02.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE This article reviews the diagnostically challenging presentation of mast cell activation disease (MCAD) and current thoughts regarding interactions between microbiota and MCs. METHODS A search for all studies on interactions between mast cells, mast cell activation disease, and microbiota published on pubmed.gov and scholar.google.com between 1960 and 2015 was conducted using the search terms mast cell, mastocyte, mastocytosis, mast cell activation, mast cell activation disease, mast cell activation syndrome, microbiome, microbiota. A manual review of the references from identified studies was also conducted. Studies were excluded if they were not accessible electronically or by interlibrary loan. FINDINGS Research increasingly is revealing essential involvement of MCs in normal human biology and in human disease. Via many methods, normal MCs-present sparsely in every tissue-sense their environment and reactively exert influences that, directly and indirectly, locally and remotely, improve health. The dysfunctional MCs of the "iceberg" of MCAD, on the other hand, sense abnormally, react abnormally, activate constitutively, and sometimes (in mastocytosis, the "tip" of the MCAD iceberg) even proliferate neoplastically. MCAD causes chronic multisystem illness generally, but not necessarily, of an inflammatory ± allergic theme and with great variability in behavior among patients and within any patient over time. Furthermore, the range of signals to which MCs respond and react include signals from the body's microbiota, and regardless of whether an MCAD patient has clonal mastocytosis or the bulk of the iceberg now known as MC activation syndrome (also suspected to be clonal but without significant MC proliferation), dysfunctional MCs interact as dysfunctionally with those microbiota as they interact with other human tissues, potentially leading to many adverse consequences. IMPLICATIONS Interactions between microbiota and MCs are complex at baseline. The potential for both pathology and benefit may be amplified when compositionally variant microbiota interact with aberrant MCs in various types of MCAD. More research is needed to better understand and leverage these interactions.
Collapse
Affiliation(s)
- Lawrence B Afrin
- Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, Minnesota.
| | - Alexander Khoruts
- Division of Gastroenterology and Center for Immunology, BioTechnology Institute, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
140
|
Voican CS, Njiké-Nakseu M, Boujedidi H, Barri-Ova N, Bouchet-Delbos L, Agostini H, Maitre S, Prévot S, Cassard-Doulcier AM, Naveau S, Perlemuter G. Alcohol withdrawal alleviates adipose tissue inflammation in patients with alcoholic liver disease. Liver Int 2015; 35:967-78. [PMID: 24766056 DOI: 10.1111/liv.12575] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 04/19/2014] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS Patients with alcoholic liver disease (ALD) display inflammation of the subcutaneous adipose tissue (SAT) which correlates with liver lesions. We examined macrophage markers and polarization in the SAT of alcoholic patients and adipokine expression according to liver inflammation; we studied the consequences of alcohol withdrawal. PATIENTS AND METHODS Forty-seven patients with ALD were prospectively included. SAT and blood samples were collected at inclusion and after 1 week of alcohol withdrawal. Pro-inflammatory cytokines/chemokines, inflammasome components and products, adipokine expression levels, macrophage markers and polarization in liver and SAT samples were assessed by RT-PCR arrays. RESULTS mRNA expression level of chemokines (IL8, semaphorin 7A) correlated with hepatic steatosis in both liver and SAT. Liver expression of inflammasome components (IL1β, IL18, caspase-1) and SAT IL6 and CCL2 correlated with liver damage. In patients with mild ALD, 1 week of alcohol withdrawal was sufficient to decrease expression level of total macrophage markers in the adipose tissue, to orient adipose tissue macrophages (ATM) towards an anti-inflammatory M2 phenotype and to decrease the mRNA expression of cytokines/chemokines (IL18, CCL2, osteopontin, semaphorin 7A). In patients with severe ALD, 1 week of abstinence was also associated with an increase in CCL18 expression. CONCLUSIONS In alcoholic patients, upregulation of chemotactic factors in the liver and SAT is an early event that begins as early as the steatosis stage. The inflammasome pathway is upregulated in the liver of patients with ALD. One week of alcohol withdrawal alleviates macrophage infiltration in SAT and orients ATM towards a M2 anti-inflammatory phenotype; this implicates alcohol in adipose tissue inflammation (ClinicalTrials.gov NCT00388323).
Collapse
Affiliation(s)
- Cosmin S Voican
- INSERM U996, IPSIT, Clamart, F-92140, France; Univ Paris-Sud, Faculté de médecine Paris-Sud, Kremlin-Bicêtre, F-94270, France; AP-HP, Hôpital Antoine Béclère, Service d'hépato-gastroentérologie et nutrition, Clamart, F-92140, France; DHU Hépatinov, Clamart, F-92140, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Hsieh CY, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep 2015; 3:e12327. [PMID: 25780093 PMCID: PMC4393161 DOI: 10.14814/phy2.12327] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 02/04/2015] [Accepted: 02/12/2015] [Indexed: 01/09/2023] Open
Abstract
Epithelial barrier dysfunction has been implicated as one of the major contributors to the pathogenesis of inflammatory bowel disease. The increase in intestinal permeability allows the translocation of luminal antigens across the intestinal epithelium, leading to the exacerbation of colitis. Thus, therapies targeted at specifically restoring tight junction barrier function are thought to have great potential as an alternative or supplement to immunology-based therapies. In this study, we screened Bifidobacterium, Enterococcus, and Lactobacillus species for beneficial microbes to strengthen the intestinal epithelial barrier, using the human intestinal epithelial cell line (Caco-2) in an in vitro assay. Some Bifidobacterium and Lactobacillus species prevented epithelial barrier disruption induced by TNF-α, as assessed by measuring the transepithelial electrical resistance (TER). Furthermore, live Bifidobacterium species promoted wound repair in Caco-2 cell monolayers treated with TNF-α for 48 h. Time course (1)H-NMR-based metabonomics of the culture supernatant revealed markedly enhanced production of acetate after 12 hours of coincubation of B. bifidum and Caco-2. An increase in TER was observed by the administration of acetate to TNF-α-treated Caco-2 monolayers. Interestingly, acetate-induced TER-enhancing effect in the coculture of B. bifidum and Caco-2 cells depends on the differentiation stage of the intestinal epithelial cells. These results suggest that Bifidobacterium species enhance intestinal epithelial barrier function via metabolites such as acetate.
Collapse
Affiliation(s)
- Chen-Yu Hsieh
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Toshifumi Osaka
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Eri Moriyama
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Yasuhiro Date
- RIKEN Center for Sustainable Resource Science, Yokohama Kanagawa, Japan Graduate School of Medical Life Science, Yokohama City University, Yokohama Kanagawa, Japan
| | - Jun Kikuchi
- RIKEN Center for Sustainable Resource Science, Yokohama Kanagawa, Japan Graduate School of Medical Life Science, Yokohama City University, Yokohama Kanagawa, Japan Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya Aichi, Japan
| | - Satoshi Tsuneda
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| |
Collapse
|
142
|
Crews FT, Sarkar DK, Qin L, Zou J, Boyadjieva N, Vetreno RP. Neuroimmune Function and the Consequences of Alcohol Exposure. Alcohol Res 2015; 37:331-41, 344-51. [PMID: 26695754 PMCID: PMC4590627] [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: 10/25/2022] Open
Abstract
Induction of neuroimmune genes by binge drinking increases neuronal excitability and oxidative stress, contributing to the neurobiology of alcohol dependence and causing neurodegeneration. Ethanol exposure activates signaling pathways featuring high-mobility group box 1 and Toll-like receptor 4 (TLR4), resulting in induction of the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells, which regulates expression of several cytokine genes involved in innate immunity, and its target genes. This leads to persistent neuroimmune responses to ethanol that stimulate TLRs and/or certain glutamate receptors (i.e., N-methyl-d-aspartate receptors). Alcohol also alters stress responses, causing elevation of peripheral cytokines, which further sensitize neuroimmune responses to ethanol. Neuroimmune signaling and glutamate excitotoxicity are linked to alcoholic neurodegeneration. Models of alcohol abuse have identified significant frontal cortical degeneration and loss of hippocampal neurogenesis, consistent with neuroimmune activation pathology contributing to these alcohol-induced, long-lasting changes in the brain. These alcohol-induced long-lasting increases in brain neuroimmune-gene expression also may contribute to the neurobiology of alcohol use disorder.
Collapse
|
143
|
Abstract
Alcoholic liver disease is a leading cause of morbidity and liver-related death worldwide. Intestinal bacterial overgrowth and dysbiosis induced by ethanol ingestion play an important role in the pathogenesis of alcoholic liver disease. After exposure to alcohol in the lumen, enteric bacteria alter their metabolism and thereby disturb intestinal homeostasis. Disruption of the mucosal barrier results in the translocation of microbial products that contribute to liver disease by inducing hepatic inflammation. In this review, we will discuss the effects of alcohol on the intestinal microbiome, and in particular, its effects on bacterial metabolism, bacterial translocation and ecological balance. A better understanding of the interactions among alcohol, the host and the microbiome will reveal new targets for therapy and lead to new treatments.
Collapse
Affiliation(s)
- Peng Chen
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
144
|
Intestinal barrier function and the brain-gut axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:73-113. [PMID: 24997030 DOI: 10.1007/978-1-4939-0897-4_4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The luminal-mucosal interface of the intestinal tract is the first relevant location where microorganism-derived antigens and all other potentially immunogenic particles face the scrutiny of the powerful mammalian immune system. Upon regular functioning conditions, the intestinal barrier is able to effectively prevent most environmental and external antigens to interact openly with the numerous and versatile elements that compose the mucosal-associated immune system. This evolutionary super system is capable of processing an astonishing amount of antigens and non-immunogenic particles, approximately 100 tons in one individual lifetime, only considering food-derived components. Most important, to develop oral tolerance and proper active immune responses needed to prevent disease and inflammation, this giant immunogenic load has to be managed in a way that physiological inflammatory balance is constantly preserved. Adequate functioning of the intestinal barrier involves local and distant regulatory networks integrating the so-called brain-gut axis. Along this complex axis both brain and gut structures participate in the processing and execution of response signals to external and internal changes coming from the digestive tract, using multidirectional pathways to communicate. Dysfunction of brain-gut axis facilitates malfunctioning of the intestinal barrier, and vice versa, increasing the risk of uncontrolled immunological reactions that may trigger mucosal and brain low-grade inflammation, a putative first step to the initiation of more permanent gut disorders. In this chapter, we describe the structure, function and interactions of intestinal barrier, microbiota and brain-gut axis in both healthy and pathological conditions.
Collapse
|
145
|
Intestinal CYP2E1: A mediator of alcohol-induced gut leakiness. Redox Biol 2014; 3:40-6. [PMID: 25462064 PMCID: PMC4297927 DOI: 10.1016/j.redox.2014.10.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 02/07/2023] Open
Abstract
Chronic alcohol use can result in many pathological effects including alcoholic liver disease (ALD). While alcohol is necessary for the development of ALD, only 20-30% of alcoholics develop alcoholic steatohepatitis (ASH) with progressive liver disease leading to cirrhosis and liver failure (ALD). This suggests that while chronic alcohol consumption is necessary it is not sufficient to induce clinically relevant liver damage in the absence of a secondary risk factor. Studies in rodent models and alcoholic patients show that increased intestinal permeability to microbial products like endotoxin play a critical role in promoting liver inflammation in ALD pathogenesis. Therefore identifying mechanisms of alcohol-induced intestinal permeability is important in identifying mechanisms of ALD and for designing new avenues for therapy. Cyp2e1 is a cytochrome P450 enzyme that metabolizes alcohol has been shown to be upregulated by chronic alcohol use and to be a major source of oxidative stress and liver injury in alcoholics and in animal and in vitro models of chronic alcohol use. Because Cyp2e1 is also expressed in the intestine and is upregulated by chronic alcohol use, we hypothesized it could play a role in alcohol-induced intestinal hyperpermeability. Our in vitro studies with intestinal Caco-2 cells and in mice fed alcohol showed that circadian clock proteins CLOCK and PER2 are required for alcohol-induced permeability. We also showed that alcohol increases Cyp2e1 protein and activity but not mRNA in Caco-2 cells and that an inhibitor of oxidative stress or siRNA knockdown of Cyp2e1 prevents the increase in CLOCK or PER2 proteins and prevents alcohol-induced hyperpermeability. With our collaborators we have also shown that Cyp2e1 knockout mice are resistant to alcohol-induced gut leakiness and liver inflammation. Taken together our data support a novel Cyp2e1-circadian clock protein mechanism for alcohol-induced gut leakiness that could provide new avenues for therapy of ALD.
Collapse
|
146
|
Intestinal permeability, gut-bacterial dysbiosis, and behavioral markers of alcohol-dependence severity. Proc Natl Acad Sci U S A 2014; 111:E4485-93. [PMID: 25288760 DOI: 10.1073/pnas.1415174111] [Citation(s) in RCA: 684] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Alcohol dependence has traditionally been considered a brain disorder. Alteration in the composition of the gut microbiota has recently been shown to be present in psychiatric disorders, which suggests the possibility of gut-to-brain interactions in the development of alcohol dependence. The aim of the present study was to explore whether changes in gut permeability are linked to gut-microbiota composition and activity in alcohol-dependent subjects. We also investigated whether gut dysfunction is associated with the psychological symptoms of alcohol dependence. Finally, we tested the reversibility of the biological and behavioral parameters after a short-term detoxification program. We found that some, but not all, alcohol-dependent subjects developed gut leakiness, which was associated with higher scores of depression, anxiety, and alcohol craving after 3 wk of abstinence, which may be important psychological factors of relapse. Moreover, subjects with increased gut permeability also had altered composition and activity of the gut microbiota. These results suggest the existence of a gut-brain axis in alcohol dependence, which implicates the gut microbiota as an actor in the gut barrier and in behavioral disorders. Thus, the gut microbiota seems to be a previously unidentified target in the management of alcohol dependence.
Collapse
|
147
|
Elamin E, Masclee A, Troost F, Pieters HJ, Keszthelyi D, Aleksa K, Dekker J, Jonkers D. Ethanol impairs intestinal barrier function in humans through mitogen activated protein kinase signaling: a combined in vivo and in vitro approach. PLoS One 2014; 9:e107421. [PMID: 25226407 PMCID: PMC4165763 DOI: 10.1371/journal.pone.0107421] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/08/2014] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Ethanol-induced gut barrier disruption is associated with several gastrointestinal and liver disorders. AIM Since human data on effects of moderate ethanol consumption on intestinal barrier integrity and involved mechanisms are limited, the objectives of this study were to investigate effects of a single moderate ethanol dose on small and large intestinal permeability and to explore the role of mitogen activated protein kinase (MAPK) pathway as a primary signaling mechanism. METHODS Intestinal permeability was assessed in 12 healthy volunteers after intraduodenal administration of either placebo or 20 g ethanol in a randomised cross-over trial. Localization of the tight junction (TJ) and gene expression, phosphorylation of the MAPK isoforms p38, ERK and JNK as indicative of activation were analyzed in duodenal biopsies. The role of MAPK was further examined in vitro using Caco-2 monolayers. RESULTS Ethanol increased small and large intestinal permeability, paralleled by redistribution of ZO-1 and occludin, down-regulation of ZO-1 and up-regulation of myosin light chain kinase (MLCK) mRNA expression, and increased MAPK isoforms phosphorylation. In Caco-2 monolayers, ethanol increased permeability, induced redistribution of the junctional proteins and F-actin, and MAPK and MLCK activation, as indicated by phosphorylation of MAPK isoforms and myosin light chain (MLC), respectively, which could be reversed by pretreatment with either MAPK inhibitors or the anti-oxidant L-cysteine. CONCLUSIONS Administration of moderate ethanol dosage can increase both small and colon permeability. Furthermore, the data indicate a pivotal role for MAPK and its crosstalk with MLCK in ethanol-induced intestinal barrier disruption. TRIAL REGISTRATION ClinicalTrials.gov NCT00928733.
Collapse
Affiliation(s)
- Elhaseen Elamin
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Ad Masclee
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Freddy Troost
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Harm-Jan Pieters
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Daniel Keszthelyi
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Katarina Aleksa
- Division of Clinical Pharmacology and Toxicology, Hospital for Sick Children, Toronto, Canada
| | - Jan Dekker
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Host microbe interactomics, Department of Animal Sciences, Wageningen University, Wageningen, the Netherlands
| | - Daisy Jonkers
- Top Institute Food and Nutrition (TIFN), Wageningen, the Netherlands
- Division Gastroenterology-Hepatology, Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
148
|
Solga SF. Breath volatile organic compounds for the gut-fatty liver axis: Promise, peril, and path forward. World J Gastroenterol 2014; 20:9017-9025. [PMID: 25083075 PMCID: PMC4112861 DOI: 10.3748/wjg.v20.i27.9017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/15/2014] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
The worldwide interest in the gut microbiome and its impact on the upstream liver highlight a critical upside to breath research: it can uniquely measure otherwise unmeasurable biology. Bacteria make gases [volatile organic compounds (VOCs)] that are directly relevant to pathophysiology of the fatty liver and associated conditions, including obesity. Measurement of these VOCs and their metabolites in the exhaled breath, therefore, present an opportunity to safely and easily evaluate, on both a personal and a population level, some of our most pressing public health threats. This is an opportunity that must be pursued. To date, however, breath analysis remains a slowly evolving field which only occasionally impacts clinical research or patient care. One major obstacle to progress is that breath analysis is inherently and emphatically mutli-disciplinary: it connects engineering, chemistry, breath mechanics, biology and medicine. Unbalanced or incomplete teams may produce inconsistent and often unsatisfactory results. A second impediment is the lack of a well-known stepwise structure for the development of non-invasive diagnostics. As a result, the breath research landscape is replete with orphaned single-center pilot studies. Often, important hypotheses and key observations have not been pursued to maturation. This paper reviews the rationale and requirements for breath VOC research applied to the gut-fatty liver axis and offers some suggestions for future development.
Collapse
|
149
|
Vonlaufen A, Spahr L, Apte MV, Frossard JL. Alcoholic pancreatitis: A tale of spirits and bacteria. World J Gastrointest Pathophysiol 2014; 5:82-90. [PMID: 24891979 PMCID: PMC4025076 DOI: 10.4291/wjgp.v5.i2.82] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 04/29/2014] [Indexed: 02/06/2023] Open
Abstract
Alcohol is a major cause of chronic pancreatitis. About 5% of alcoholics will ever suffer from pancreatitis, suggesting that additional co-factors are required to trigger an overt disease. Experimental work has implicated lipopolysaccharide, from gut-derived bacteria, as a potential co-factor of alcoholic pancreatitis. This review discusses the effects of alcohol on the gut flora, the gut barrier, the liver-and the pancreas and proposes potential interventional strategies. A better understanding of the interaction between the gut, the liver and the pancreas may provide valuable insight into the pathophysiology of alcoholic pancreatitis.
Collapse
|
150
|
Schnabl B, Brenner DA. Interactions between the intestinal microbiome and liver diseases. Gastroenterology 2014; 146:1513-24. [PMID: 24440671 PMCID: PMC3996054 DOI: 10.1053/j.gastro.2014.01.020] [Citation(s) in RCA: 742] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/29/2013] [Accepted: 01/07/2014] [Indexed: 02/08/2023]
Abstract
The human intestine harbors a diverse community of microbes that promote metabolism and digestion in their symbiotic relationship with the host. Disturbance of its homeostasis can result in disease. We review factors that disrupt intestinal homeostasis and contribute to nonalcoholic fatty liver disease, steatohepatitis, alcoholic liver disease, and cirrhosis. Liver disease has long been associated with qualitative and quantitative (overgrowth) dysbiotic changes in the intestinal microbiota. Extrinsic factors, such as the Western diet and alcohol, contribute to these changes. Dysbiosis results in intestinal inflammation, a breakdown of the intestinal barrier, and translocation of microbial products in animal models. However, the contribution of the intestinal microbiome to liver disease goes beyond simple translocation of bacterial products that promote hepatic injury and inflammation. Microbial metabolites produced in a dysbiotic intestinal environment and host factors are equally important in the pathogenesis of liver disease. We review how the combination of liver insult and disruptions in intestinal homeostasis contribute to liver disease.
Collapse
Affiliation(s)
- Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, California.
| | | |
Collapse
|