201
|
Waluga M, Kukla M, Żorniak M, Bacik A, Kotulski R. From the stomach to other organs: Helicobacter pylori and the liver. World J Hepatol 2015; 7:2136-2146. [PMID: 26328025 PMCID: PMC4550868 DOI: 10.4254/wjh.v7.i18.2136] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 07/23/2015] [Accepted: 08/21/2015] [Indexed: 02/06/2023] Open
Abstract
Many recent studies have examined the importance of Helicobacter pylori (H. pylori) infection in the pathogenesis of the diseases outside the stomach and explored the significance of this bacterium in the pathogenesis of some metabolic and cardiovascular diseases. Recent studies have provided evidence that H. pylori is also involved in the pathogenesis of some liver diseases. Many observations have proved that H. pylori infection is important in the development of insulin resistance, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis, liver fibrosis and cirrhosis. The worsening of liver inflammation of different origins also occurs during H. pylori infection. Some studies have indicated that H. pylori infection induces autoimmunological diseases in the liver and biliary tract. The potential significance of this bacterium in carcinogenesis is unclear, but it is within the scope of interest of many studies. The proposed mechanisms through which H. pylori impacts the development of hepatobiliary diseases are complex and ambiguous. The importance of other Helicobacter species in the development of hepatobiliary diseases is also considered because they could lead to the development of inflammatory, fibrotic and necrotic injuries of the liver and, consequently, to hepatocellular carcinoma. However, many contrary viewpoints indicate that some evidence is not convincing, and further studies of the subject are needed. This review presents the current knowledge about the importance of H. pylori in the pathogenesis of liver and in biliary diseases.
Collapse
|
202
|
Abstract
Oxygen is used by eukaryotic cells for metabolic transformations and energy production in mitochondria. Under physiological conditions, there is a constant endogenous production of intermediates of reactive oxygen (ROI) and nitrogen species (RNI) that interact as signaling molecules in physiological mechanisms. When these species are not eliminated by antioxidants or are produced in excess, oxidative stress arises. Oxidative stress can damage proteins, lipids, DNA, and organelles. It is a process directly linked to inflammation; in fact, inflammatory cells secrete a large number of cytokines and chemokines responsible for the production of ROI and RNI in phagocytic and nonphagocytic cells through the activation of protein kinases signaling. Currently, there is a wide variety of diseases capable of producing inflammatory manifestations. While, in the short term, most of these diseases are not fatal they have a major impact on life quality. Since there is a direct relationship between chronic inflammation and many emerging disorders like cancer, oral diseases, kidney diseases, fibromyalgia, gastrointestinal chronic diseases or rheumatics diseases, the aim of this review is to describe the use and role of melatonin, a hormone secreted by the pineal gland, that works directly and indirectly as a free radical scavenger, like a potent antioxidant.
Collapse
Affiliation(s)
- Aroha Sánchez
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Barcelona, Joan XXIII Avenue, Barcelona 08028, Spain.
| | - Ana Cristina Calpena
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Barcelona, Joan XXIII Avenue, Barcelona 08028, Spain.
| | - Beatriz Clares
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Granada, Campus of Cartuja Street, Granada 18071, Spain.
| |
Collapse
|
203
|
The Influence of the Gut Microbiome on Obesity, Metabolic Syndrome and Gastrointestinal Disease. Clin Transl Gastroenterol 2015; 6:e91. [PMID: 26087059 PMCID: PMC4816244 DOI: 10.1038/ctg.2015.16] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 04/13/2015] [Indexed: 12/11/2022] Open
Abstract
There is a fine balance in the mutual relationship between the intestinal microbiota and its mammalian host. It is thought that disruptions in this fine balance contribute/account for the pathogenesis of many diseases. Recently, the significance of the relationship between gut microbiota and its mammalian host in the pathogenesis of obesity and the metabolic syndrome has been demonstrated. Emerging data has linked intestinal dysbiosis to several gastrointestinal diseases including inflammatory bowel disease, irritable bowel syndrome, nonalcoholic fatty liver disease, and gastrointestinal malignancy. This article is intended to review the role of gut microbiota maintenance/alterations of gut microbiota as a significant factor as a significant factor discriminating between health and common diseases. Based on current available data, the role of microbial manipulation in disease management remains to be further defined and a focus for further clinical investigation.
Collapse
|
204
|
Sellmann C, Priebs J, Landmann M, Degen C, Engstler AJ, Jin CJ, Gärttner S, Spruss A, Huber O, Bergheim I. Diets rich in fructose, fat or fructose and fat alter intestinal barrier function and lead to the development of nonalcoholic fatty liver disease over time. J Nutr Biochem 2015; 26:1183-92. [PMID: 26168700 DOI: 10.1016/j.jnutbio.2015.05.011] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/08/2015] [Accepted: 05/13/2015] [Indexed: 02/07/2023]
Abstract
General overnutrition but also a diet rich in certain macronutrients, age, insulin resistance and an impaired intestinal barrier function may be critical factors in the development of nonalcoholic fatty liver disease (NAFLD). Here the effect of chronic intake of diets rich in different macronutrients, i.e. fructose and/or fat on liver status in mice, was studied over time. C57BL/6J mice were fed plain water, 30% fructose solution, a high-fat diet or a combination of both for 8 and 16 weeks. Indices of liver damage, toll-like receptor 4 (TLR-4) signaling cascade, macrophage polarization and insulin resistance in the liver and intestinal barrier function were analyzed. Chronic exposure to a diet rich in fructose and/or fat was associated with the development of hepatic steatosis that progressed with time to steatohepatitis in mice fed a combination of macronutrients. The development of NAFLD was also associated with a marked reduction of the mRNA expression of insulin receptor, whereas hepatic expressions of TLR-4, myeloid differentiation primary response gene 88 and markers of M1 polarization of macrophages were induced in comparison to controls. Bacterial endotoxin levels in portal plasma were found to be increased while levels of the tight junction protein occludin and zonula occludens 1 were found to be significantly lower in the duodenum of all treated groups after 8 and 16 weeks. Our data suggest that chronic intake of fructose and/or fat may lead to the development of NAFLD over time and that this is associated with an increased translocation of bacterial endotoxin.
Collapse
Affiliation(s)
- Cathrin Sellmann
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Josephine Priebs
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Marianne Landmann
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Christian Degen
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Anna Janina Engstler
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Cheng Jun Jin
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Stefanie Gärttner
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany
| | - Astrid Spruss
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - Otmar Huber
- Department of Biochemistry II, University Hospital Jena, Friedrich-Schiller-University Jena, Jena, Germany; Center of Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Ina Bergheim
- Institute of Nutritional Sciences, Model Systems of Molecular Nutrition, Friedrich-Schiller-University Jena, Jena, Germany.
| |
Collapse
|
205
|
Chronic inflammation aggravates metabolic disorders of hepatic fatty acids in high-fat diet-induced obese mice. Sci Rep 2015; 5:10222. [PMID: 25974206 PMCID: PMC4431481 DOI: 10.1038/srep10222] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/07/2015] [Indexed: 02/06/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) increases with increasing body mass index (BMI). However, approximately 40–50% of obese adults do not develop hepatic steatosis. The level of inflammatory biomarkers is higher in obese subjects with NAFLD compared to BMI-matched subjects without hepatic steatosis. We used a casein injection in high-fat diet (HFD)-fed C57BL/6J mice to induce inflammatory stress. Although mice on a HFD exhibited apparent phenotypes of obesity and hyperlipidemia regardless of exposure to casein injection, only the HFD+Casein mice showed increased hepatic vacuolar degeneration accompanied with elevated inflammatory cytokines in the liver and serum, compared to mice on a normal chow diet. The expression of genes related to hepatic fatty acid synthesis and oxidation were upregulated in the HFD-only mice. The casein injection further increased baseline levels of lipogenic genes and decreased the levels of oxidative genes in HFD-only mice. Inflammatory stress induced both oxidative stress and endoplasmic reticulum stress in HFD-fed mice livers. We conclude that chronic inflammation precedes hepatic steatosis by disrupting the balance between fatty acid synthesis and oxidation in the livers of HFD-fed obese mice. This mechanism may operate in obese individuals with chronic inflammation, thus making them more prone to NAFLD.
Collapse
|
206
|
Role of gut barrier function in the pathogenesis of nonalcoholic Fatty liver disease. Gastroenterol Res Pract 2015; 2015:287348. [PMID: 25945084 PMCID: PMC4402198 DOI: 10.1155/2015/287348] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/28/2015] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common forms of chronic liver disease, and its incidence is increasing year by year. Many efforts have been made to investigate the pathogenesis of this disease. Since 1998 when Marshall proposed the conception of "gut-liver axis," more and more researchers have paid close attention to the role of gut barrier function in the pathogenesis of NAFLD. The four aspects of gut barrier function, including physical, chemical, biological, and immunological barriers, are interrelated closely and related to NAFLD. In this paper, we present a summary of research findings on the relationship between gut barrier dysfunction and the development of NAFLD, aiming at illustrating the role of gut barrier function in the pathogenesis of this disease.
Collapse
|
207
|
Luther J, Garber JJ, Khalili H, Dave M, Bale SS, Jindal R, Motola DL, Luther S, Bohr S, Jeoung SW, Deshpande V, Singh G, Turner JR, Yarmush ML, Chung RT, Patel SJ. Hepatic Injury in Nonalcoholic Steatohepatitis Contributes to Altered Intestinal Permeability. Cell Mol Gastroenterol Hepatol 2015; 1:222-232. [PMID: 26405687 PMCID: PMC4578658 DOI: 10.1016/j.jcmgh.2015.01.001] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/09/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Emerging data suggest that changes in intestinal permeability and increased gut microbial translocation contribute to the inflammatory pathway involved in nonalcoholic steatohepatitis (NASH) development. Numerous studies have investigated the association between increased intestinal permeability and NASH. Our meta-analysis of this association investigates the underlying mechanism. METHODS A meta-analysis was performed to compare the rates of increased intestinal permeability in patients with NASH and healthy controls. To further address the underlying mechanism of action, we studied changes in intestinal permeability in a diet-induced (methionine-and-choline-deficient; MCD) murine model of NASH. In vitro studies were also performed to investigate the effect of MCD culture medium at the cellular level on hepatocytes, Kupffer cells, and intestinal epithelial cells. RESULTS Nonalcoholic fatty liver disease (NAFLD) patients, and in particular those with NASH, are more likely to have increased intestinal permeability compared with healthy controls. We correlate this clinical observation with in vivo data showing mice fed an MCD diet develop intestinal permeability changes after an initial phase of liver injury and tumor necrosis factor-α (TNFα) induction. In vitro studies reveal that MCD medium induces hepatic injury and TNFα production yet has no direct effect on intestinal epithelial cells. Although these data suggest a role for hepatic TNFα in altering intestinal permeability, we found that mice genetically resistant to TNFα-myosin light chain kinase (MLCK)-induced intestinal permeability changes fed an MCD diet still develop increased permeability and liver injury. CONCLUSIONS Our clinical and experimental results strengthen the association between intestinal permeability increases and NASH and also suggest that an early phase of hepatic injury and inflammation contributes to altered intestinal permeability in a fashion independent of TNFα and MLCK.
Collapse
Affiliation(s)
- Jay Luther
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John J. Garber
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hamed Khalili
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maneesh Dave
- Division of Gastroenterology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Shyam Sundhar Bale
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, and the Shriners Burns Hospital, Boston, Massachusetts
| | - Rohit Jindal
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, and the Shriners Burns Hospital, Boston, Massachusetts
| | - Daniel L. Motola
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sanjana Luther
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stefan Bohr
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, and the Shriners Burns Hospital, Boston, Massachusetts
| | - Soung Won Jeoung
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vikram Deshpande
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gurminder Singh
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | - Martin L. Yarmush
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, and the Shriners Burns Hospital, Boston, Massachusetts
| | - Raymond T. Chung
- Gastrointestinal Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Suraj J. Patel
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, and the Shriners Burns Hospital, Boston, Massachusetts
| |
Collapse
|
208
|
The Gut Microbiota, Intestinal Permeability, Bacterial Translocation, and Nonalcoholic Fatty Liver Disease: What Comes First? Cell Mol Gastroenterol Hepatol 2015; 1:129-130. [PMID: 28210672 PMCID: PMC5301143 DOI: 10.1016/j.jcmgh.2015.01.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
209
|
Tomita K, Hokari R. [Gut Microbiota and Internal Diseases: Update Information. Topics: III. NASH/NAFLD and gut microbiota]. NIHON NAIKA GAKKAI ZASSHI. THE JOURNAL OF THE JAPANESE SOCIETY OF INTERNAL MEDICINE 2015; 104:48-56. [PMID: 26571774 DOI: 10.2169/naika.104.48] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
|
210
|
Brandl K, Schnabl B. Is intestinal inflammation linking dysbiosis to gut barrier dysfunction during liver disease? Expert Rev Gastroenterol Hepatol 2015; 9:1069-76. [PMID: 26088524 PMCID: PMC4828034 DOI: 10.1586/17474124.2015.1057122] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Changes in the intestinal microbiota composition contribute to the pathogenesis of many disorders including gastrointestinal and liver diseases. Recent studies have broadened our understanding of the "gut-liver" axis. Dietary changes, other environmental and genetic factors can lead to alterations in the microbiota. Dysbiosis can further disrupt the integrity of the intestinal barrier leading to pathological bacterial translocation and the initiation of an inflammatory response in the liver. In this article, the authors dissect the different steps involved in disease pathogenesis to further refine approaches for the medical management of liver diseases. The authors will specifically discuss the role of dysbiosis in inducing intestinal inflammation and increasing intestinal permeability.
Collapse
Affiliation(s)
- Katharina Brandl
- Skaggs School of Pharmacy, University of California San Diego, 9500 Gilman Drive La Jolla, California 92093-0675, 858-822-6853,
| | - Bernd Schnabl
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA,Department of Medicine, 9500 Gilman Drive, La Jolla, CA 92093-0063, University of California San Diego, 858-534-9484,
| |
Collapse
|
211
|
Burcelin R, Courtney M, Amar J. Gut Microbiota and Metabolic Diseases: From Pathogenesis to Therapeutic Perspective. MOLECULAR AND INTEGRATIVE TOXICOLOGY 2015:199-234. [DOI: 10.1007/978-1-4471-6539-2_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
212
|
Arslan N. Obesity, fatty liver disease and intestinal microbiota. World J Gastroenterol 2014; 20:16452-16463. [PMID: 25469013 PMCID: PMC4248188 DOI: 10.3748/wjg.v20.i44.16452] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 05/14/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disorder that is increasing in prevalence with the worldwide epidemic of obesity. NAFLD is the hepatic manifestation of the metabolic syndrome. The term NAFLD describes a spectrum of liver pathology ranges from simple steatosis to steatosis with inflammation nonalcoholic steatohepatitis and even cirrhosis. Metabolic syndrome and NAFLD also predict hepatocellular carcinoma. Many genetic and environmental factors have been suggested to contribute to the development of obesity and NAFLD, but the exact mechanisms are not known. Intestinal ecosystem contains trillions of microorganisms including bacteria, Archaea, yeasts and viruses. Several studies support the relationship between the intestinal microbial changes and obesity and also its complications, including insulin resistance and NAFLD. Given that the gut and liver are connected by the portal venous system, it makes the liver more vulnerable to translocation of bacteria, bacterial products, endotoxins or secreted cytokines. Altered intestinal microbiota (dysbiosis) may stimulate hepatic fat deposition through several mechanisms: regulation of gut permeability, increasing low-grade inflammation, modulation of dietary choline metabolism, regulation of bile acid metabolism and producing endogenous ethanol. Regulation of intestinal microbial ecosystem by diet modifications or by using probiotics and prebiotics as a treatment for obesity and its complications might be the issue of further investigations.
Collapse
|
213
|
Shanmugam NKN, Trebicka E, Fu LL, Shi HN, Cherayil BJ. Intestinal inflammation modulates expression of the iron-regulating hormone hepcidin depending on erythropoietic activity and the commensal microbiota. THE JOURNAL OF IMMUNOLOGY 2014; 193:1398-407. [PMID: 24973448 DOI: 10.4049/jimmunol.1400278] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
States of chronic inflammation such as inflammatory bowel disease are often associated with dysregulated iron metabolism and the consequent development of an anemia that is caused by maldistribution of iron. Abnormally elevated expression of the hormone hepcidin, the central regulator of systemic iron homeostasis, has been implicated in these abnormalities. However, the mechanisms that regulate hepcidin expression in conditions such as inflammatory bowel disease are not completely understood. To clarify this issue, we studied hepcidin expression in mouse models of colitis. We found that dextran sulfate sodium-induced colitis inhibited hepcidin expression in wild-type mice but upregulated it in IL-10-deficient animals. We identified two mechanisms contributing to this difference. Firstly, erythropoietic activity, as indicated by serum erythropoietin concentrations and splenic erythropoiesis, was higher in the wild-type mice, and pharmacologic inhibition of erythropoiesis prevented colitis-associated hepcidin downregulation in these animals. Secondly, the IL-10 knockout mice had higher expression of multiple inflammatory genes in the liver, including several controlled by STAT3, a key regulator of hepcidin. The results of cohousing and fecal transplantation experiments indicated that the microbiota was involved in modulating the expression of hepcidin and other STAT3-dependent hepatic genes in the context of intestinal inflammation. Our observations thus demonstrate the importance of erythropoietic activity and the microbiota in influencing hepcidin expression during colitis and provide insight into the dysregulated iron homeostasis seen in inflammatory diseases.
Collapse
Affiliation(s)
- Nanda Kumar N Shanmugam
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129; and
| | - Estela Trebicka
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129; and
| | - Ling-Lin Fu
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129; and Food Safety Key Laboratory of Zhejiang Province, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310035, People's Republic of China
| | - Hai Ning Shi
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129; and
| | - Bobby J Cherayil
- Department of Pediatrics, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129; and
| |
Collapse
|
214
|
Miura K, Ohnishi H. Role of gut microbiota and Toll-like receptors in nonalcoholic fatty liver disease. World J Gastroenterol 2014; 20:7381-7391. [PMID: 24966608 PMCID: PMC4064083 DOI: 10.3748/wjg.v20.i23.7381] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/07/2014] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Emerging data have shown a close association between compositional changes in gut microbiota and the development of nonalcoholic fatty liver disease (NAFLD). The change in gut microbiota may alter nutritional absorption and storage. In addition, gut microbiota are a source of Toll-like receptor (TLR) ligands, and their compositional change can also increase the amount of TLR ligands delivered to the liver. TLR ligands can stimulate liver cells to produce proinflammatory cytokines. Therefore, the gut-liver axis has attracted much interest, particularly regarding the pathogenesis of NAFLD. The abundance of the major gut microbiota, including Firmicutes and Bacteroidetes, has been considered a potential underlying mechanism of obesity and NAFLD, but the role of these microbiota in NAFLD remains unknown. Several reports have demonstrated that certain gut microbiota are associated with the development of obesity and NAFLD. For instance, a decrease in Akkermansia muciniphila causes a thinner intestinal mucus layer and promotes gut permeability, which allows the leakage of bacterial components. Interventions to increase Akkermansia muciniphila improve the metabolic parameters in obesity and NAFLD. In children, the levels of Escherichia were significantly increased in nonalcoholic steatohepatitis (NASH) compared with those in obese control. Escherichia can produce ethanol, which promotes gut permeability. Thus, normalization of gut microbiota using probiotics or prebiotics is a promising treatment option for NAFLD. In addition, TLR signaling in the liver is activated, and its downstream molecules, such as proinflammatory cytokines, are increased in NAFLD. To data, TLR2, TLR4, TLR5, and TLR9 have been shown to be associated with the pathogenesis of NAFLD. Therefore, gut microbiota and TLRs are targets for NAFLD treatment.
Collapse
|
215
|
Myles IA. Fast food fever: reviewing the impacts of the Western diet on immunity. Nutr J 2014; 13:61. [PMID: 24939238 PMCID: PMC4074336 DOI: 10.1186/1475-2891-13-61] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 05/23/2014] [Indexed: 02/08/2023] Open
Abstract
While numerous changes in human lifestyle constitute modern life, our diet has been gaining attention as a potential contributor to the increase in immune-mediated diseases. The Western diet is characterized by an over consumption and reduced variety of refined sugars, salt, and saturated fat. Herein our objective is to detail the mechanisms for the Western diet's impact on immune function. The manuscript reviews the impacts and mechanisms of harm for our over-indulgence in sugar, salt, and fat, as well as the data outlining the impacts of artificial sweeteners, gluten, and genetically modified foods; attention is given to revealing where the literature on the immune impacts of macronutrients is limited to either animal or in vitro models versus where human trials exist. Detailed attention is given to the dietary impact on the gut microbiome and the mechanisms by which our poor dietary choices are encoded into our gut, our genes, and are passed to our offspring. While today's modern diet may provide beneficial protection from micro- and macronutrient deficiencies, our over abundance of calories and the macronutrients that compose our diet may all lead to increased inflammation, reduced control of infection, increased rates of cancer, and increased risk for allergic and auto-inflammatory disease.
Collapse
Affiliation(s)
- Ian A Myles
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike Building 33, Room 2W10A, Bethesda, MD, 20892, Maryland.
| |
Collapse
|
216
|
Zarepoor L, Lu JT, Zhang C, Wu W, Lepp D, Robinson L, Wanasundara J, Cui S, Villeneuve S, Fofana B, Tsao R, Wood GA, Power KA. Dietary flaxseed intake exacerbates acute colonic mucosal injury and inflammation induced by dextran sodium sulfate. Am J Physiol Gastrointest Liver Physiol 2014; 306:G1042-55. [PMID: 24763556 DOI: 10.1152/ajpgi.00253.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Flaxseed (FS), a dietary oilseed, contains a variety of anti-inflammatory bioactives, including fermentable fiber, phenolic compounds (lignans), and the n-3 polyunsaturated fatty acid (PUFA) α-linolenic acid. The objective of this study was to determine the effects of FS and its n-3 PUFA-rich kernel or lignan- and soluble fiber-rich hull on colitis severity in a mouse model of acute colonic inflammation. C57BL/6 male mice were fed a basal diet (negative control) or a basal diet supplemented with 10% FS, 6% kernel, or 4% hull for 3 wk prior to and during colitis induction via 5 days of 2% (wt/vol) dextran sodium sulfate (DSS) in their drinking water (n = 12/group). An increase in anti-inflammatory metabolites (hepatic n-3 PUFAs, serum mammalian lignans, and cecal short-chain fatty acids) was associated with consumption of all FS-based diets, but not with anti-inflammatory effects in DSS-exposed mice. Dietary FS exacerbated DSS-induced acute colitis, as indicated by a heightened disease activity index and an increase in colonic injury and inflammatory biomarkers [histological damage, apoptosis, myeloperoxidase, inflammatory cytokines (IL-6 and IL-1β), and NF-κB signaling-related genes (Nfkb1, Ccl5, Bcl2a1a, Egfr, Relb, Birc3, and Atf1)]. Additionally, the adverse effect of the FS diet was extended systemically, as serum cytokines (IL-6, IFNγ, and IL-1β) and hepatic cholesterol levels were increased. The adverse effects of FS were not associated with alterations in fecal microbial load or systemic bacterial translocation (endotoxemia). Collectively, this study demonstrates that although consumption of a 10% FS diet enhanced the levels of n-3 PUFAs, short-chain polyunsaturated fatty acids, and lignans in mice, it exacerbated DSS-induced colonic injury and inflammation.
Collapse
Affiliation(s)
- Leila Zarepoor
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Jenifer T Lu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Claire Zhang
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | - Wenqing Wu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Dion Lepp
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Lindsay Robinson
- Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada
| | | | - Steve Cui
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | | | - Bourlaye Fofana
- Crops and Livestock Research Centre, AAFC, Charlottetown, Prince Edward Island, Canada; and
| | - Rong Tsao
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Krista A Power
- Guelph Food Research Centre, Agriculture and Agri-Food Canada (AAFC), Guelph, Ontario, Canada; Department of Human Health and Nutritional Science, University of Guelph, Guelph, Ontario, Canada;
| |
Collapse
|
217
|
Lei S, Cheng T, Guo Y, Li C, Zhang W, Zhi F. Somatostatin ameliorates lipopolysaccharide-induced tight junction damage via the ERK-MAPK pathway in Caco2 cells. Eur J Cell Biol 2014; 93:299-307. [PMID: 24950815 DOI: 10.1016/j.ejcb.2014.05.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 05/16/2014] [Accepted: 05/21/2014] [Indexed: 12/21/2022] Open
Abstract
Dysfunction of the epithelial barrier is an important pathogenic factor of inflammatory bowel disease and other inflammatory conditions of the gut. Somatostatin (SST) has been demonstrated to reduce local and systemic inflammation reactions and maintain the integrity of the blood-brain barrier (BBB). To determine the beneficial effect of SST on lipopolysaccharide (LPS)-induced damage of the tight junction (TJ) and its mechanisms, Caco2 cells pretreated with SST (1nM) or MEK inhibitor U0126 (10μM) were exposed to LPS. LPS significantly reduced the expression of TJ proteins in a dose-dependent way. LPS (100μg/ml) greatly induced Caco2 monolayer barrier dysfunction by decreasing transepithelial resistance and increasing epithelial permeability. Pretreatment with SST effectively improved the barrier dysfunction of Caco2 cells. SST significantly increased the expression of TJ proteins occludin and ZO-1 and inhibited the redistribution of TJ proteins due to LPS stimulation. Furthermore, SST decreased the LPS-induced phosphorylation of ERK1/2, and a selective MEK inhibitor markedly protected the barrier function against LPS disturbance by blocking the activation of the ERK-MAPK pathway in Caco2 cells. Besides, LPS significantly increased the mRNA level of SSTR5, which was partly inhibited by pretreatment with SST. In conclusion, the present study indicates that SST protects the Caco2 monolayer barrier against LPS-induced tight junction breakdown by down-regulating the activation of the ERK-MAPK pathway and suppression the activation of SSTR5.
Collapse
Affiliation(s)
- Shan Lei
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tianming Cheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yandong Guo
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wendi Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
218
|
Giorgio V, Miele L, Principessa L, Ferretti F, Villa MP, Negro V, Grieco A, Alisi A, Nobili V. Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with liver disease severity. Dig Liver Dis 2014; 46:556-60. [PMID: 24631029 DOI: 10.1016/j.dld.2014.02.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2013] [Revised: 02/06/2014] [Accepted: 02/12/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Increased intestinal permeability seems to play a major role in non-alcoholic liver disease development and progression. AIM To investigate the prevalence of altered intestinal permeability in children with non-alcoholic fatty liver disease, and to study its potential association with the stage of liver disease. METHODS We performed a case-control study examining intestinal permeability in children using the lactulose-mannitol bowel permeability test. RESULTS Overall, 39 consecutive patients (30 males, median age 12 years) and 21 controls (14 males, median age 11.8 years) were included. The lactulose/mannitol ratio resulted impaired in 12/39 patients (31%) and none of the controls. Intestinal permeability was higher in children with non-alcoholic fatty liver disease (lactulose/mannitol ratios: 0.038±0.037 vs. 0.008±0.007, p<0.05). Within the non-alcoholic fatty liver disease group, intestinal permeability was increased in children with steatohepatitis compared to those with steatosis only (0.05±0.04 vs. 0.03 vs. 0.03, p<0.05). Pathological lactulose/mannitol ratio correlated with portal inflammation (p=0.02), fibrosis (p=0.0002), and ballooning of hepatocytes (p=0.003). Blood lipopolysaccharides levels were higher in children with steatohepatitis (2.27±0.68 vs. 2.80±0.35, p<0.05). CONCLUSIONS Intestinal permeability is increased in children with non-alcoholic fatty liver disease, and correlates with the severity of the disease.
Collapse
Affiliation(s)
- Valentina Giorgio
- Hepato-metabolic Disease Unit, Bambino Gesù Children Hospital, Rome, Italy
| | - Luca Miele
- Clinical Division of Internal Medicine, Gastroenterology and Liver Unit, Complesso Integrato Columbus Hospital, Catholic University of Rome, Italy; Institute of Internal Medicine, Catholic University of Rome, Italy
| | | | - Francesca Ferretti
- Hepato-metabolic Disease Unit, Bambino Gesù Children Hospital, Rome, Italy
| | - Maria Pia Villa
- Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Valentina Negro
- Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Antonio Grieco
- Clinical Division of Internal Medicine, Gastroenterology and Liver Unit, Complesso Integrato Columbus Hospital, Catholic University of Rome, Italy
| | - Anna Alisi
- Hepato-metabolic Disease Unit, Bambino Gesù Children Hospital, Rome, Italy
| | - Valerio Nobili
- Hepato-metabolic Disease Unit, Bambino Gesù Children Hospital, Rome, Italy.
| |
Collapse
|
219
|
Melatonin in the regulation of liver steatosis following prenatal glucocorticoid exposure. BIOMED RESEARCH INTERNATIONAL 2014; 2014:942172. [PMID: 24822223 PMCID: PMC4005100 DOI: 10.1155/2014/942172] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 03/25/2014] [Indexed: 12/13/2022]
Abstract
Nonalcoholic fatty liver disease patients are characterized by hepatic steatosis. Prenatal glucocorticoid overexposure can result in steatosis. In this study, we aimed to determine the mechanism and cellular apoptosis of prenatal glucocorticoid overexposure in rats and whether melatonin can rescue the prenatal glucocorticoid-induced steatosis and apoptosis in neonatal rats. Pregnant Sprague-Dawley rats at gestational days 14 to 21 were administered dexamethasone. Acute effects of prenatal programming liver were assessed at postnatal day 7. The expression of proteins involved in the apoptotic and methylation pathways was analyzed by RT-PCR and Western blotting. Apoptosis and steatosis were examined by histology staining. The liver steatosis and apoptosis were increased in prenatal glucocorticoid group more than in control group and decreased in melatonin group. The expression of leptin decreased in prenatal glucocorticoid and increased in melatonin group by liver RT-PCR and Western blot study. Caspase 3, TNF-α proteins expression, and TUNEL stains increased in prenatal glucocorticoid compared with control and decreased in melatonin group. The liver histone deacetylase, DNA methyltransferase activity, and DNA methylation were increased in prenatal glucocorticoid and decreased in melatonin group. The present study showed that the prenatal glucocorticoid induced programming liver steatosis at day 7 after delivery, possibly via altered leptin expression. Melatonin can reverse the methylation of leptin and decreased liver steatosis.
Collapse
|
220
|
Seidel D, Eickmeier I, Kühl AA, Hamann A, Loddenkemper C, Schott E. CD8 T cells primed in the gut-associated lymphoid tissue induce immune-mediated cholangitis in mice. Hepatology 2014; 59:601-11. [PMID: 24038154 DOI: 10.1002/hep.26702] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 08/18/2013] [Indexed: 12/19/2022]
Abstract
UNLABELLED The pathogenesis of primary sclerosing cholangitis (PSC) remains poorly understood. Since PSC predominantly occurs in patients with inflammatory bowel disease, autoimmunity triggered by activated T cells migrating from the gut to the liver is a possible mechanism. We hypothesized that T cells primed in the gut-associated lymphoid tissue (GALT) by a specific antigen migrate to the liver and cause cholangitis when they recognize the same antigen on cholangiocytes. We induced ovalbumin-dependent colitis in mice that express ovalbumin in biliary epithelia (ASBT-OVA mice) and crossed ASBT-OVA mice with mice that express ovalbumin in enterocytes (iFABP-OVA mice). We analyzed T-cell activation in the GALT and crossreactivity to the same antigen in the liver as well as the effects of colitis per se on antigen-presentation and T-cell activation in the liver. Intrarectal application of ovalbumin followed by transfer of CD8 OT-I T cells led to antigen-dependent colitis. CD8 T cells primed in the GALT acquired effector function and the capability to migrate to the liver, where they caused cholangitis in a strictly antigen-dependent manner. Likewise, cholangitis developed in mice expressing ovalbumin simultaneously in biliary epithelia and enterocytes after transfer of OT-I T cells. Dextran sodium sulfate colitis led to increased levels of inflammatory cytokines in the portal venous blood, induced activation of resident liver dendritic cells, and promoted the induction of T-cell-dependent cholangitis. CONCLUSION Our data strengthen the notion that immune-mediated cholangitis is caused by T cells primed in the GALT and provide the first link between colitis and cholangitis in an antigen-dependent mouse model.
Collapse
Affiliation(s)
- Daniel Seidel
- Department of Hepatology and Gastroenterology, Charité Universitätsmedizin, CVK, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
221
|
Abstract
There are trillions of microorganisms in the human intestine collectively called gut microbiota. Obesity may be affected by the gut microbiota through energy harvesting and fat storage by the bacteria. Small intestinal bacterial overgrowth is also responsible for endotoxemia, systemic inflammation, and its consequences including obesity and nonalcoholic fatty liver disease (NAFLD). Relationship between gut microbiota and NAFLD is also dependent on altered choline and bile acid metabolism and endogenous alcohol production by gut bacteria. Further evidence linking gut microbiota with obesity and NAFLD comes from studies showing usefulness of probiotics in animals and patients with NAFLD. This article reviews the relationship among gut microbiota, obesity, and NAFLD.
Collapse
Affiliation(s)
- Ajay Duseja
- Department of Hepatology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh 160012, India
| | | |
Collapse
|
222
|
Mikami Y, Mizuno S, Nakamoto N, Hayashi A, Sujino T, Sato T, Kamada N, Matsuoka K, Hisamatsu T, Ebinuma H, Hibi T, Yoshimura A, Kanai T. Macrophages and dendritic cells emerge in the liver during intestinal inflammation and predispose the liver to inflammation. PLoS One 2014; 9:e84619. [PMID: 24392145 PMCID: PMC3879334 DOI: 10.1371/journal.pone.0084619] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022] Open
Abstract
The liver is a physiological site of immune tolerance, the breakdown of which induces immunity. Liver antigen-presenting cells may be involved in both immune tolerance and activation. Although inflammatory diseases of the liver are frequently associated with inflammatory bowel diseases, the underlying immunological mechanisms remain to be elucidated. Here we report two murine models of inflammatory bowel disease: RAG-2−/− mice adoptively transferred with CD4+CD45RBhigh T cells; and IL-10−/− mice, accompanied by the infiltration of mononuclear cells in the liver. Notably, CD11b−CD11clowPDCA-1+ plasmacytoid dendritic cells (DCs) abundantly residing in the liver of normal wild-type mice disappeared in colitic CD4+CD45RBhigh T cell-transferred RAG-2−/− mice and IL-10−/− mice in parallel with the emergence of macrophages (Mφs) and conventional DCs (cDCs). Furthermore, liver Mφ/cDCs emerging during intestinal inflammation not only promote the proliferation of naïve CD4+ T cells, but also instruct them to differentiate into IFN-γ-producing Th1 cells in vitro. The emergence of pathological Mφ/cDCs in the liver also occurred in a model of acute dextran sulfate sodium (DSS)-induced colitis under specific pathogen-free conditions, but was canceled in germ-free conditions. Last, the Mφ/cDCs that emerged in acute DSS colitis significantly exacerbated Fas-mediated hepatitis. Collectively, intestinal inflammation skews the composition of antigen-presenting cells in the liver through signaling from commensal bacteria and predisposes the liver to inflammation.
Collapse
Affiliation(s)
- Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Shinta Mizuno
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Hayashi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- Research Laboratory, Miyarisan Pharmaceutical, Tokyo, Japan
| | - Tomohisa Sujino
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Nobuhiko Kamada
- Department of Pathology and Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Katsuyoshi Matsuoka
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Tadakazu Hisamatsu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hirotoshi Ebinuma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Toshifumi Hibi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- * E-mail: (TK); (AY)
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
- * E-mail: (TK); (AY)
| |
Collapse
|
223
|
Zhao WX, Wang L, Yang JL, Li LZ, Xu WM, Li T. Caffeic acid phenethyl ester attenuates pro-inflammatory and fibrogenic phenotypes of LPS-stimulated hepatic stellate cells through the inhibition of NF-κB signaling. Int J Mol Med 2013; 33:687-94. [PMID: 24378685 DOI: 10.3892/ijmm.2013.1613] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/05/2013] [Indexed: 11/06/2022] Open
Abstract
Hepatic stellate cells (HSCs) are the major cell type involved in liver fibrosis. Lipopolysaccharide (LPS)-mediated signaling through Τoll-like receptor 4 (TLR4) in HSCs has been identified as a key event in liver fibrosis, and as the molecular link between inflammation and liver fibrosis. In this study, we investigated the effects of caffeic acid phenethyl ester (CAPE), one of the main medicinal components of propolis, on the pro-inflammatory and fibrogenic phenotypes of LPS-stimulated HSCs. HSCs from rats were isolated and cultured in Dulbecco's modified Eagle's medium (DMEM). Following treatment with LPS, HSCs showed a strong pro-inflammatory phenotype with an upregulation of pro-inflammatory mediators, and a fibrogenic phenotype with enhanced collagen synthesis, mediated by transforming growth factor-β1 (TGF-β1). CAPE significantly and dose-dependently reduced LPS-induced nitrite production, as well as the transcription and protein synthesis of monocyte chemoattractant protein-1 (MCP-1), interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS), as determined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting and enzyme-linked immunosorbent assays (ELISA). CAPE further reduced the TGF-β1-induced transcription and translation (protein synthesis) of the gene coding for collagen type I α1 (col1A1), in LPS-stimulated HSCs. Following LPS stimulation, the phosphorylation of the nuclear factor-κB (NF-κB) inhibitor IκBα and consequently, the nuclear translocation of NF-κB, were markedly increased in the HSCs, and these changes were reversed by pre-treatment with CAPE. In conclusion, CAPE attenuates the pro-inflammatory phenotype of LPS-stimulated HSCs, as well as the LPS-induced sensitization of HSCs to fibrogenic cytokines by inhibiting NF-κB signaling. Our results provide new insight into the treatment of hepatic fibrosis through regulation of the TLR4 signaling pathway.
Collapse
Affiliation(s)
- Wen-Xing Zhao
- Department of Pathology, Kunming General Hospital of PLA, Kunming, Yunnan 650032, P.R. China
| | - Li Wang
- Department of Pathology, Kunming General Hospital of PLA, Kunming, Yunnan 650032, P.R. China
| | - Ju-Lun Yang
- Department of Pathology, Kunming General Hospital of PLA, Kunming, Yunnan 650032, P.R. China
| | - Lian-Zhen Li
- School of Life Sciences of Yunnan University, Kunming, Yunnan 650091, P.R. China
| | - Wen-Mang Xu
- Department of Pathology, Kunming General Hospital of PLA, Kunming, Yunnan 650032, P.R. China
| | - Tao Li
- Department of Pathology, Kunming General Hospital of PLA, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
224
|
Trivedi PP, Jena GB. Melatonin reduces ulcerative colitis-associated local and systemic damage in mice: investigation on possible mechanisms. Dig Dis Sci 2013; 58:3460-74. [PMID: 23975342 DOI: 10.1007/s10620-013-2831-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 07/27/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Ulcerative colitis (UC) is a chronic gastrointestinal disorder. Substantial research reveals that melatonin has beneficial effects in ulcerative colitis both experimentally and clinically. We have previously reported that ulcerative colitis was associated with local and systemic damage in mice. The purpose of this study was to reveal the novel targets of melatonin in its protective mechanism against ulcerative colitis in mice. We also wished to determine whether or not melatonin protected against ulcerative colitis-induced systemic damage in mice. METHODS Ulcerative colitis was induced in mice by use of 3% (w/v) dextran sulfate sodium for two cycles. One cycle comprised 7 days of DSS-treated water followed by 14 days of normal drinking water. Melatonin was administered at doses of 2, 4, or 8 mg/kg bw/day, po throughout. The effect of melatonin in mice with UC was evaluated by use of biochemical data, histological evaluation, comet and micronucleus assays, immunohistochemistry, and western blot analysis. RESULTS The results indicated that melatonin treatment ameliorated the severity of ulcerative colitis by modulating a variety of molecular targets, for example nuclear factor kappa B, cyclooxygenase-2, interleukin 17, signal transducer and activator of transcription 3, nuclear erythroid 2-related factor 2, matrix metalloproteinase-9, and connective tissue growth factor. Further, ulcerative colitis increased gut permeability, plasma lipopolysaccharide level, systemic inflammation, and genotoxicity. Melatonin treatment led to mucosal healing and reduced ulcerative colitis-induced elevated gut permeability and reduced the plasma LPS level, systemic inflammation, and genotoxicity. CONCLUSION Melatonin ameliorated ulcerative colitis-associated local and systemic damage in mice.
Collapse
Affiliation(s)
- P P Trivedi
- Department of Pharmacology and Toxicology, Facility for Risk Assessment and Intervention Studies, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab, 160062, India,
| | | |
Collapse
|
225
|
Li M, Shen Z, Li YM. Potential role of Helicobacter pylori infection in nonalcoholic fatty liver disease. World J Gastroenterol 2013; 19:7024-7031. [PMID: 24222944 PMCID: PMC3819536 DOI: 10.3748/wjg.v19.i41.7024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/05/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence has implicated Helicobacter pylori (H. pylori) infection in extragastrointestinal diseases, including obesity, type 2 diabetes mellitus, cardiovascular disease, and liver disease. Recently, there has been a special focus on H. pylori infection as a risk factor for the development of nonalcoholic fatty liver disease (NAFLD). NAFLD is currently considered to be the most common liver disorder in western countries, and is rapidly becoming a serious threat to public health. The mechanisms of pathogenesis underlying NAFLD remain unclear at present and therapeutic options are limited. The growing awareness of the role of H. pylori in NAFLD is thus important to aid the development of novel intervention and prevention strategies, because the eradication of H. pylori is easy and much less expensive than long-term treatment of the other risk factors. H. pylori infection is involved in the pathogenesis of insulin resistance (IR), which is closely linked with NAFLD. It provides a new insight into the pathogenesis of NAFLD. This review probes the possible relationship between H. pylori and NAFLD, from the perspective of the potential mechanism of how H. pylori infection brings about IR and other aspects concerning this correlation.
Collapse
|
226
|
Influence of oxidative stress on the level of genes expression TGFB1 and HGF in rat liver upon long-term gastric hypochlorhydria and administration of multiprobiotic Symbiter. UKRAINIAN BIOCHEMICAL JOURNAL 2013. [DOI: 10.15407/ubj85.05.114] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
227
|
Steib CJ, Gmelin L, Pfeiler S, Schewe J, Brand S, Göke B, Gerbes AL. Functional relevance of the cannabinoid receptor 2 - heme oxygenase pathway: a novel target for the attenuation of portal hypertension. Life Sci 2013; 93:543-51. [PMID: 24007798 DOI: 10.1016/j.lfs.2013.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 12/17/2022]
Abstract
AIMS In liver cirrhosis, inflammation triggers portal hypertension. Kupffer cells (KC) produce vasoconstrictors upon activation by bacterial constituents. Here, we hypothesize that the anti-inflammatory action of the cannabinoid receptor 2 (CB2) agonists JWH-133 and GP 1a attenuate portal hypertension. MAIN METHODS In vivo measurements of portal pressures and non-recirculating liver perfusions were performed in rats 4weeks after bile duct ligation (BDL). Zymosan (150μg/ml, isolated liver perfusion) or LPS (4mg/kgb.w., in vivo) was infused to activate the KC in the absence or presence of JWH-133 (10mg/kgb.w.), GP 1a (2.5mg/kgb.w.) or ZnPP IX (1μM). Isolated KC were treated with Zymosan (0.5mg/ml) in addition to JWH-133 (5μM). The thromboxane (TX) B2 levels in the perfusate and KC media were determined by ELISA. Heme oxygenase-1 (HO-1) and CB2 were analyzed by Western blot or confocal microscopy. KEY FINDINGS JWH-133 or GP 1a pre-treatment attenuated portal pressures following KC activation in all experimental settings. In parallel, HO-1 expression increased with JWH-133 pre-treatment. However, the inhibition of HO-1 enhanced portal hypertension, indicating the functional role of this novel pathway. In isolated KC, the expression of CB2 and HO-1 increased with Zymosan, LPS and JWH-133 treatment while TXB2 production following KC activation was attenuated by JWH-133 pre-treatment. SIGNIFICANCE JWH-133 or GP 1a treatment attenuates portal hypertension. HO-1 induction by JWH-133 plays a functional role. Therefore, the administration of JWH-133 or GP 1a represents a promising new treatment option for portal hypertension triggered by microbiological products.
Collapse
Key Words
- (6aR,10aR)-3-(1,1-dimethylbutyl)-6a,7,10,10a-tetrahydro-6,6,9-trimethyl-6H-dibenzo[b,d]pyran
- 2-Chloro-5-nitro-N-phenylbenzamide
- BDL
- Bile duct ligation (BDL)
- CB(2)
- GW 9662
- Gp 1a
- HO
- Heme oxygenase (HO)
- JWH-133
- KC
- Kupffer cell
- LDH
- LPS
- N-(Piperidin-1-yl)-1-(2,4-dichlorophenyl)-1,4-dihydro-6-methylindeno[1,2-c]pyrazole-3-carboxamide
- TLR
- TX
- Thromboxane
- Zy
- Zymosan A
- b. w
- bile duct ligation
- body weight
- cannabinoid receptor 2
- heme oxygenase
- lactate dehydrogenase
- lipopolysaccharide
- thromboxane
- toll like receptor
Collapse
Affiliation(s)
- Christian J Steib
- Department of Medicine II (Gastroenterology and Hepatology), Liver Center Munich, University of Munich, Grosshadern, Munich, Germany.
| | | | | | | | | | | | | |
Collapse
|
228
|
Trivedi P, Jena G. Role of α-lipoic acid in dextran sulfate sodium-induced ulcerative colitis in mice: Studies on inflammation, oxidative stress, DNA damage and fibrosis. Food Chem Toxicol 2013; 59:339-55. [DOI: 10.1016/j.fct.2013.06.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/07/2013] [Accepted: 06/12/2013] [Indexed: 01/08/2023]
|
229
|
Myles IA, Fontecilla NM, Janelsins BM, Vithayathil PJ, Segre JA, Datta SK. Parental dietary fat intake alters offspring microbiome and immunity. THE JOURNAL OF IMMUNOLOGY 2013; 191:3200-9. [PMID: 23935191 DOI: 10.4049/jimmunol.1301057] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mechanisms underlying modern increases in prevalence of human inflammatory diseases remain unclear. The hygiene hypothesis postulates that decreased microbial exposure has, in part, driven this immune dysregulation. However, dietary fatty acids also influence immunity, partially through modulation of responses to microbes. Prior reports have described the direct effects of high-fat diets on the gut microbiome and inflammation, and some have additionally shown metabolic consequences for offspring. Our study sought to expand on these previous observations to identify the effects of parental diet on offspring immunity using mouse models to provide insights into challenging aspects of human health. To test the hypothesis that parental dietary fat consumption during gestation and lactation influences offspring immunity, we compared pups of mice fed either a Western diet (WD) fatty acid profile or a standard low-fat diet. All pups were weaned onto the control diet to specifically test the effects of early developmental fat exposure on immune development. Pups from WD breeders were not obese or diabetic, but still had worse outcomes in models of infection, autoimmunity, and allergic sensitization. They had heightened colonic inflammatory responses, with increased circulating bacterial LPS and muted systemic LPS responsiveness. These deleterious impacts of the WD were associated with alterations of the offspring gut microbiome. These results indicate that parental fat consumption can leave a "lard legacy" impacting offspring immunity and suggest inheritable microbiota may contribute to the modern patterns of human health and disease.
Collapse
Affiliation(s)
- Ian A Myles
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Natalia M Fontecilla
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Brian M Janelsins
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Paul J Vithayathil
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Julia A Segre
- Epithelial Biology Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Sandip K Datta
- Bacterial Pathogenesis Unit, Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
230
|
Frasinariu OE, Ceccarelli S, Alisi A, Moraru E, Nobili V. Gut-liver axis and fibrosis in nonalcoholic fatty liver disease: an input for novel therapies. Dig Liver Dis 2013; 45:543-51. [PMID: 23280158 DOI: 10.1016/j.dld.2012.11.010] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Revised: 11/08/2012] [Accepted: 11/23/2012] [Indexed: 02/06/2023]
Abstract
Non-alcoholic fatty liver disease is a multifactorial condition, ranging from simple steatosis to non-alcoholic steatohepatitis with or without fibrosis. In non-alcoholic fatty liver disease, alteration of gut microbiota and increased intestinal permeability increase exposure of the liver to gut-derived bacterial products: lipopolysaccharides and unmethylated CpG DNA. These products stimulate innate immune receptors, namely Toll-like receptors, which activate signalling pathways involved in liver inflammation and fibrogenesis. Currently, there are several studies on the involvement of lipopolysaccharide-activated Toll-like receptor 4 signalling in non-alcoholic fatty liver disease pathogenesis. There has been widespread interest in the study of the involvement of resident hepatic stellate cells and Kupffer cells activation in liver fibrogenesis upon TLR4 stimulation. Although the best evidence to support a role for gut microbiota in non-alcoholic fatty liver disease-induced fibrosis comes largely from animal models, data from human studies are accumulating and could lead to new therapeutic approaches. Therapeutic modulation of gut microflora may be an alternative strategy to develop an anti-fibrotic therapy. In this review, we discuss the relevant role of gut-liver axis in non-alcoholic liver disease-associated liver fibrosis and discuss the evidence on novel anti-fibrotic therapeutic approaches.
Collapse
Affiliation(s)
- Otilia E Frasinariu
- Discipline of Pediatrics, Department of Mother and Child Medicine, University of Medicine and Pharmacy, Grigore T. Popa, Iasi, Iasi, Romania
| | | | | | | | | |
Collapse
|
231
|
Huebener P, Schwabe RF. Regulation of wound healing and organ fibrosis by toll-like receptors. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1832:1005-17. [PMID: 23220258 PMCID: PMC3848326 DOI: 10.1016/j.bbadis.2012.11.017] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/22/2012] [Accepted: 11/23/2012] [Indexed: 02/06/2023]
Abstract
Chronic injury often triggers maladaptive wound healing responses leading to the development of tissue fibrosis and subsequent organ malfunction. Inflammation is a key component of the wound healing process and promotes the development of organ fibrosis. Here, we review the contribution of Toll-like receptors (TLRs) to wound healing with a particular focus on their role in liver, lung, kidney, skin and myocardial fibrosis. We discuss the role of TLRs on distinct cell populations that participate in the repair process following tissue injury, and the contribution of exogenous and endogenous TLR ligands to the wound healing response. Systemic review of the literature shows that TLRs promote tissue repair and fibrosis in many settings, albeit with profound differences between organs. In particular, TLRs exert a pronounced effect on fibrosis in organs with higher exposure to bacterial TLR ligands, such as the liver. Targeting TLR signaling at the ligand or receptor level may represent a novel strategy for the prevention of maladaptive wound healing and fibrosis in chronically injured organs. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Peter Huebener
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
| | - Robert F. Schwabe
- Department of Medicine, Columbia University, College of Physicians and Surgeons, New York, NY 10032, USA
- Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| |
Collapse
|
232
|
Abstract
PURPOSE OF REVIEW Interactions of the gut microbiome with the host are important in health and disease. Microbial translocation releases bacterial products that play a key role in progression of chronic liver disease by promoting hepatic injury and inflammation. Although this has long been recognized, we are just beginning to understand the circumstances under which the gut becomes leaky and to discover bacterial metabolites that promote liver disease. In this review, we will summarize recent findings from the last 2 years. RECENT FINDINGS Chronic liver disease is associated with an altered microbiome with both qualitative (dysbiosis) and quantitative (overgrowth) differences. This can be viewed as a loss of the symbiotic relationship between the microflora and the host. An imbalanced intestinal homeostasis results in a breach of the gut barrier and subsequent microbial translocation. However, the contribution of the intestinal microflora is beyond simple microbial translocation as a pathogenic factor. Bacterial metabolites resulting from an imbalanced homeostasis and dysbiosis play also a crucial role in liver disease. SUMMARY A combination between an initiating liver insult and a disturbance of the gut-host symbiosis synergize in progression of liver disease.
Collapse
|
233
|
Aron-Wisnewsky J, Gaborit B, Dutour A, Clement K. Gut microbiota and non-alcoholic fatty liver disease: new insights. Clin Microbiol Infect 2013; 19:338-48. [DOI: 10.1111/1469-0691.12140] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 12/21/2012] [Indexed: 12/20/2022]
|
234
|
Nakano M, Murohisa T, Imai Y, Tamano M, Hiraishi H. Association Between Appendectomy and Fibrosis Progression in Nonalcoholic Fatty Liver Disease. Gastroenterology Res 2013; 6:17-25. [PMID: 27785221 PMCID: PMC5051115 DOI: 10.4021/gr513w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2013] [Indexed: 12/17/2022] Open
Abstract
Background A two-hit theory explaining the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH) and fibrosis is widely accepted. Endotoxins entering the portal vein from the gut are thought to be one cause of this second hit, and the literature frequently mentions associations between gut-derived endotoxins and progression of fibrosis in NAFLD. The appendix regulates intestinal immunity to protect the gut from the invasion of bacteria and antigens. Appendectomy may thus contribute to progression of fibrosis in NAFLD, but this association has not yet been clarified. We therefore investigated the association between appendectomy and progression of fibrosis in NAFLD. Methods Fifty two patients with NAFLD who underwent liver biopsy in our department were included in this study. Based on Brunt’s scores, patients with NAFLD were classified into a mild fibrosis group and advanced fibrosis group. Results History of appendectomy was found to be significantly more frequent in patients with advanced fibrosis than in patients with mild fibrosis (P = 0.014). Multivariate logistic analysis was conducted with age, sex, albumin, platelet count, steatosis grade, and history of appendectomy as covariates and advanced fibrosis as the dependent variable. Significant differences were identified for platelet count and history of appendectomy, identifying these as independent risk factors for advanced fibrosis in NAFLD patients. The odds ratio for appendectomy history was 39.415 (P = 0.044). Conclusions History of appendectomy was significantly more frequent in NAFLD patients with advanced fibrosis, suggesting that appendectomy may represent a risk factor for advanced fibrosis in NAFLD.
Collapse
Affiliation(s)
- Masakazu Nakano
- Department of Gastroenterology, Dokkyo Medical University, 880 Kitakobayashi, Mibu-machi, Simotsuga-gun, Tochigi, Japan
| | - Toshimitsu Murohisa
- Department of Gastroenterology, Dokkyo Medical University, 880 Kitakobayashi, Mibu-machi, Simotsuga-gun, Tochigi, Japan
| | - Yasuo Imai
- Department of Pathology, Dokkyo Medical University Koshigaya Hospital, 2-1-50 Minamikoshigaya, Koshigaya, Saitama, Japan
| | - Masaya Tamano
- Department of Gastroenterology, Dokkyo Medical University Koshigaya Hospital, 2-1-50 Minamikoshigaya, Koshigaya, Saitama, Japan
| | - Hideyuki Hiraishi
- Department of Gastroenterology, Dokkyo Medical University, 880 Kitakobayashi, Mibu-machi, Simotsuga-gun, Tochigi, Japan
| |
Collapse
|
235
|
|
236
|
Trivedi PP, Jena GB. Ulcerative colitis-induced hepatic damage in mice: studies on inflammation, fibrosis, oxidative DNA damage and GST-P expression. Chem Biol Interact 2012; 201:19-30. [PMID: 23261717 DOI: 10.1016/j.cbi.2012.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 11/01/2012] [Accepted: 12/08/2012] [Indexed: 12/12/2022]
Abstract
There exists a close relationship between ulcerative colitis and various hepatic disorders. The present study was aimed to evaluate the hepatocellular damage in experimental colitis model. Ulcerative colitis was induced in Swiss mice by cyclic treatment with 3% w/v dextran sulfate sodium in drinking water. The severity of colitis was assessed on the basis of disease activity index and colon histology. The effect of ulcerative colitis on the liver was assessed using various biochemical parameters, histological evaluation, sirius red staining, immunohistochemical staining with peroxisome proliferator-activated receptor γ, 8-oxo-7,8-dihydro-2'-deoxyguanosine and placental glutathione S-transferase, comet assay (alkaline and modified), Terminal Deoxynucleotidyl Transferase-mediated dUTP Nick End Labeling assay and western blot analysis to detect the protein expression of nuclear factor kappa B, cyclooxygenase-2, nuclear erythroid 2-related factor 2 and NADPH: quinone oxidoreductase-1. Dextran sulfate sodium induced severe colitis in mice as evident from an elevated disease activity index and histological abnormalities. Ulcerative colitis increased the permeability of colon as apparent from a significant reduction in the expression of tight junction protein, occludin. Further, the bacterial translocation assay as well as the analysis of lipopolysaccharide level revealed the existence of various bacterial species in the liver of ulcerative colitis-induced mice. There was a significant increase in the plasma alanine and aspartate transaminases and liver triglyceride levels, expression of peroxisome proliferator-activated receptor γ, inflammatory markers, oxidative stress, fibrosis, oxidative DNA damage and apoptosis in the liver of mice. Moreover, there was an increase in the expression of nuclear factor kappa B and cyclooxygenase-2 and a reduction in the expression of nuclear erythroid 2-related factor 2 and NADPH: quinone oxidoreductase-1 in the liver of severe ulcerative colitis-induced mice. The results of the present study provide evidence that ulcerative colitis is accompanied with hepatic damage in mice.
Collapse
Affiliation(s)
- P P Trivedi
- Facility for Risk Assessment and Intervention Studies, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S. Nagar, Punjab 160062, India.
| | | |
Collapse
|
237
|
Gut microbiome and intestinal barrier failure--the "Achilles heel" in hepatology? J Hepatol 2012; 56:1221-3. [PMID: 22406521 DOI: 10.1016/j.jhep.2012.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/02/2012] [Indexed: 12/14/2022]
|
238
|
Yang L, Seki E. Toll-like receptors in liver fibrosis: cellular crosstalk and mechanisms. Front Physiol 2012; 3:138. [PMID: 22661952 PMCID: PMC3357552 DOI: 10.3389/fphys.2012.00138] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Accepted: 04/24/2012] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors that distinguish conserved microbial products, also known as pathogen-associated molecular patterns (PAMPs), from host molecules. Liver is the first filter organ between the gastrointestinal tracts and the rest of the body through portal circulation. Thus, the liver is a major organ that must deal with PAMPs and microorganisms translocated from the intestine and to respond to the damage associated molecular patterns (DAMPs) released from injured organs. These PAMPs and DAMPs preferentially activate TLR signaling on various cell types in the liver inducing the production of inflammatory and fibrogenic cytokines that initiate and prolong liver inflammation, thereby leading to fibrosis. We summarize recent findings on the role of TLRs, ligands, and intracellular signaling in the pathophysiology of liver fibrosis due to different etiology, as well as to highlight the potential role of TLR signaling in liver fibrosis associated with hepatitis C infection, non-alcoholic and alcoholic steatoheoatitis, primary biliary cirrhosis, and cystic fibrosis.
Collapse
Affiliation(s)
- Ling Yang
- Division of Gastroenterology, Department of Medicine, University of California San Diego, School of Medicine La Jolla, CA, USA
| | | |
Collapse
|
239
|
Dai X, Lv ZS. Role of gut barrier function in the pathogenesis of nonalcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2012; 20:656-661. [DOI: 10.11569/wcjd.v20.i8.656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common forms of chronic liver disease, and its incidence is increasing year by year. Many efforts have been made to investigate the pathogenesis of this disease. Since 1998 when Marshall proposed the conception of "gut-liver axis", more and more researchers have paid close attention to the role of gut barrier function in the pathogenesis of NAFLD. The four aspects of gut barrier function, including physical, chemical, biological and immunological barriers are interrelated closely and related to NAFLD. In this paper, we present a summary of research findings on the relationship between gut barrier dysfunction and the occurrence and development of NAFLD, aiming at illustrating the role of gut barrier function in the pathogenesis of this disease.
Collapse
|
240
|
Niu GC, Liu L, Zhang XL. Hepatobiliary complications of inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2012; 20:662-669. [DOI: 10.11569/wcjd.v20.i8.662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic idiopathic immune-mediated inflammatory disease of the gastrointestinal tract, including ulcerative colitis (UC) and Crohn's disease (CD). Extraintestinal manifestations (EIMs) are frequently encountered in IBD patients. Hepatobiliary manifestations are common extraintestinal manifestations of IBD, including primary sclerosing cholangitis (PSC), nonalcoholic fatty liver, cholelithiasis, primary biliary cirrhosis (PBC), small-duct PSC, IgG4-associated cholangitis (IAC), granulomatous hepatitis, amyloidosis, autoimmune hepatitis (AIH), PSC/AIH overlap syndrome, and portal vein thrombosis. In this paper, we describe the progress in understanding the hepatobiliary complications associated with IBD.
Collapse
|
241
|
Raczynski AR, Muthupalani S, Schlieper K, Fox JG, Tannenbaum SR, Schauer DB. Enteric infection with Citrobacter rodentium induces coagulative liver necrosis and hepatic inflammation prior to peak infection and colonic disease. PLoS One 2012; 7:e33099. [PMID: 22427959 PMCID: PMC3302869 DOI: 10.1371/journal.pone.0033099] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Accepted: 02/09/2012] [Indexed: 01/07/2023] Open
Abstract
Acute and chronic forms of inflammation are known to affect liver responses and susceptibility to disease and injury. Furthermore, intestinal microbiota has been shown critical in mediating inflammatory host responses in various animal models. Using C. rodentium, a known enteric bacterial pathogen, we examined liver responses to gastrointestinal infection at various stages of disease pathogenesis. For the first time, to our knowledge, we show distinct liver pathology associated with enteric infection with C. rodentium in C57BL/6 mice, characterized by increased inflammation and hepatitis index scores as well as prominent periportal hepatocellular coagulative necrosis indicative of thrombotic ischemic injury in a subset of animals during the early course of C. rodentium pathogenesis. Histologic changes in the liver correlated with serum elevation of liver transaminases, systemic and liver resident cytokines, as well as signal transduction changes prior to peak bacterial colonization and colonic disease. C. rodentium infection in C57BL/6 mice provides a potentially useful model to study acute liver injury and inflammatory stress under conditions of gastrointestinal infection analogous to enteropathogenic E. coli infection in humans.
Collapse
Affiliation(s)
- Arkadiusz R Raczynski
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America.
| | | | | | | | | | | |
Collapse
|
242
|
Etiopathogenesis of nonalcoholic steatohepatitis: role of obesity, insulin resistance and mechanisms of hepatotoxicity. Int J Hepatol 2012; 2012:212865. [PMID: 22792473 PMCID: PMC3389710 DOI: 10.1155/2012/212865] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Revised: 02/26/2012] [Accepted: 04/25/2012] [Indexed: 12/14/2022] Open
Abstract
Incidence of nonalcoholic fatty liver disease is increasing with an estimated prevalence of 20-30% in developed nations. This is leading to increased incidence of chronic liver disease, cirrhosis, and hepatocellular cancer. It is critical to understand the etiology and pathogenesis of any disease to create therapeutic targets and develop new treatments. In this paper we discuss the etiology and pathogenesis of nonalcoholic steatohepatitis with special focus on obesity, role of insulin resistance, and molecular mechanisms of hepatotoxicity.
Collapse
|