1
|
Berköz M, Aslan A, Yunusoğlu O, Krośniak M, Francik R. Hepatoprotective potentials of Usnea longissima Ach. and Xanthoparmelia somloensis (Gyelnik) Hale extracts in ethanol-induced liver injury. Drug Chem Toxicol 2024:1-14. [PMID: 39322224 DOI: 10.1080/01480545.2024.2407867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/09/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
In our study, the antioxidant and anti-inflammatory effects of different lichen applications were investigated in rats using an experimental ethanol toxicity model. 48 rats were used in the study and they were divided into 6 groups with 8 rats in each group. These groups were: control, ethanol (2 g/kg), ethanol + Usnea longissima Ach. (200 mg/kg), ethanol + Usnea longissima Ach. (400 mg/kg), ethanol + Xanthoparmelia somloensis (Gyelnik) Hale (100 mg/kg) and ethanol + Xanthoparmelia somloensis (Gyelnik) Hale (200 mg/kg). The experimental work continued for 21 days. Lichen extracts and ethanol were administered by gavage to rats divided into groups. According to the experimental protocol, the experimental animals were sacrificed and their liver tissues were isolated. Biochemical parameters in serum, histological examinations, oxidative stress and inflammation parameters both at biochemical and molecular level in liver tissues were performed. Oxidative stress and inflammatory response were increased in the liver tissue of rats treated with ethanol for 21 days, and liver functions were impaired. It was found that U. longissima and X. somloensis extracts showed good antioxidant activity and conferred protective effects against ethanol-induced oxidative stress and inflammation. This could be attributed to the presence of secondary metabolites in the extract, which act as natural antioxidants and could be responsible for increasing the defence mechanisms against free radical production induced by ethanol administration.
Collapse
Affiliation(s)
- Mehmet Berköz
- Department of Biochemistry, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, Turkey
| | - Ali Aslan
- Department of Pharmacology, Faculty of Pharmacy, Van Yuzuncu Yil University, Van, Turkey
- Department of Biology, Faculty of Science, Kyrgyz-Turkish Manas University, Bishkek, Kyrgyzstan
| | - Oruç Yunusoğlu
- Department of Medical Pharmacology, Faculty of Medicine, Bolu Abant İzzet Baysal University, Bolu, Turkey
| | - Mirosław Krośniak
- Department of Food Chemistry and Nutrition, Medical College, Jagiellonian University, Cracow, Poland
| | - Renata Francik
- Department of Bioorganic Chemistry, Medical College, Jagiellonian University, Cracow, Poland
| |
Collapse
|
2
|
Wickramasinghe PB, Qian S, Langley LE, Liu C, Jia L. Hepatocyte Toll-Like Receptor 4 Mediates Alcohol-Induced Insulin Resistance in Mice. Biomolecules 2023; 13:454. [PMID: 36979389 PMCID: PMC10046504 DOI: 10.3390/biom13030454] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Accumulating evidence has demonstrated the association between alcohol overconsumption and the development of insulin resistance. However, the underlying mechanisms are not completely understood. To investigate the requirement and sufficiency of hepatocyte toll-like receptor 4 (TLR4) in alcohol-induced insulin resistance, we used two mouse models (Tlr4fl/fl and Tlr4LoxTB) that allow ablation of TLR4 only in hepatocytes (Tlr4LKO) and restoration of endogenous TLR4 expression in hepatocytes on a TLR4-null background (Tlr4LoxTB × Alb-Cre), respectively. A Lieber-DeCarli feeding model was used to induce glucose intolerance and insulin resistance in mice. Glucose tolerance test, insulin tolerance test, and insulin signaling experiments were performed to examine systemic and tissue-specific insulin sensitivity. We found that alcohol-fed hepatocyte TLR4 deficient mice (Tlr4LKO) had lower blood glucose levels in response to intraperitoneal injection of insulin. Moreover, increased phosphorylation of glycogen synthase kinase-3β (GSK3β) was observed in the liver of Tlr4LKO mice after chronic alcohol intake. In contrast, when hepatic TLR4 was reactivated in mice (Tlr4LoxTB × Alb-Cre), alcohol feeding caused glucose intolerance in these mice compared with littermate controls (Tlr4LoxTB). In addition, AKT phosphorylation was dramatically reduced in the liver and epididymal white adipose tissue (eWAT) of alcohol-fed Tlr4LoxTB × Alb-Cre mice, which was similar to that of mice with whole-body TLR4 reactivation (Tlr4LoxTB × Zp3-Cre). Collectively, these findings suggest that hepatocyte TLR4 is both required and sufficient in the development of insulin resistance induced by alcohol overconsumption.
Collapse
Affiliation(s)
- Piumi B. Wickramasinghe
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Shuwen Qian
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lyndsey E. Langley
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| | - Chen Liu
- Center for Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Jia
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
3
|
Foster TC. Animal models for studies of alcohol effects on the trajectory of age-related cognitive decline. Alcohol 2023; 107:4-11. [PMID: 35504438 DOI: 10.1016/j.alcohol.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
There is growing interest in understanding how ethanol use interacts with advancing age to influence the brain and cognition. Animal models are employed to investigate the cellular and molecular mechanisms of brain aging and age-related neurodegenerative diseases that underlie cognitive decline. However, all too often research on problems and diseases of the elderly are conducted in healthy young animals, providing little clinical relevance. The validity of animal models is discussed, and confounds due to age-related differences in anxiety, sensory-motor function, and procedural learning are highlighted in order to enhance the successful translation of preclinical results into clinical settings. The mechanism of action of ethanol on brain aging will depend on the dose, acute or chronic treatment, or withdrawal from treatment and the age examined. Due to the fact that humans experience alcohol use throughout life, important questions concern the effects of the dose and duration of ethanol treatment on the trajectory of cognitive function. Central to this research will be questions of the specificity of alcohol effects on cognitive functions and related brain regions that decline with age, as well as the interaction of alcohol with mechanisms or biomarkers of brain aging. Alternatively, moderate alcohol use may provide a source of reserve and resilience against brain aging. Longitudinal studies have the advantage of being sensitive to detecting the effects of treatment on the emergence of cognitive impairment in middle age and can minimize effects of stress/anxiety associated with the novelty of alcohol exposure and behavioral testing, which disproportionately influence aged animals. Finally, the effect of alcohol on senescent neurophysiology and biomarkers of brain aging are discussed. In particular, the interaction of age and effects of alcohol on inflammation, oxidative stress, N-methyl-d-aspartate receptor function, and the balance of excitatory and inhibitory synaptic transmission are highlighted.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
4
|
Marković Filipović J, Karan J, Ivelja I, Matavulj M, Stošić M. Acrylamide and Potential Risk of Diabetes Mellitus: Effects on Human Population, Glucose Metabolism and Beta-Cell Toxicity. Int J Mol Sci 2022; 23:6112. [PMID: 35682790 PMCID: PMC9181725 DOI: 10.3390/ijms23116112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus is a frequent endocrine disorder characterized by hyperglycemia. Acrylamide (AA) is food contaminant formed during the high-temperature processing of food rich in carbohydrates and low in proteins. Recent human epidemiological studies have shown a potential association between AA exposure and the prevalence of diabetes in the general population. In male rats, AA treatment promoted pancreatic islet remodeling, which was determined by alpha-cell expansion and beta-cell reduction, while in female rats AA caused hyperglycemia and histopathological changes in pancreatic islets. In vitro and in vivo rodent model systems have revealed that AA induces oxidative stress in beta cells and that AA impairs glucose metabolism and the insulin signaling pathway. Animal studies have shown that diabetic rodents are more sensitive to acrylamide and that AA aggravates the diabetic state. In this review, we provide an overview of human epidemiological studies that examined the relation between AA exposure and glucose disorders. In addition, the effects of AA treatment on pancreatic islet structure, beta-cell function and glucose metabolism in animal models are comprehensively analyzed with an emphasis on sex-related responses. Furthermore, oxidative stress as a putative mechanism of AA-induced toxicity in beta cells is explored. Finally, we discuss the effects of AA on diabetics in a rodent model system.
Collapse
Affiliation(s)
- Jelena Marković Filipović
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (J.K.); (I.I.); (M.M.)
| | - Jelena Karan
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (J.K.); (I.I.); (M.M.)
| | - Ivana Ivelja
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (J.K.); (I.I.); (M.M.)
| | - Milica Matavulj
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (J.K.); (I.I.); (M.M.)
| | - Milena Stošić
- Department of Environmental Engineering and Occupational Safety and Health, Faculty of Technical Science, University of Novi Sad, Trg Dositeja Obradovića 6, 21000 Novi Sad, Serbia;
| |
Collapse
|
5
|
Marković Filipović J, Miler M, Kojić D, Karan J, Ivelja I, Čukuranović Kokoris J, Matavulj M. Effect of Acrylamide Treatment on Cyp2e1 Expression and Redox Status in Rat Hepatocytes. Int J Mol Sci 2022; 23:6062. [PMID: 35682741 PMCID: PMC9181519 DOI: 10.3390/ijms23116062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 12/16/2022] Open
Abstract
Acrylamide (AA) toxicity is associated with oxidative stress. During detoxification, AA is either coupled to gluthatione or biotransformed to glycidamide by the enzyme cytochrome P450 2E1 (CYP2E1). The aim of our study was to examine the hepatotoxicity of AA in vivo and in vitro. Thirty male Wistar rats were treated with 25 or 50 mg/kg b.w. of AA for 3 weeks. Qualitative and quantitative immunohistochemical evaluation of inducible nitric oxide synthase (iNOS), CYP2E1, catalase (CAT), superoxide dismutase 1 (SOD1), and SOD2 expression in liver was carried out. Bearing in mind that the liver is consisted mainly of hepatocytes, in a parallel study, we used the rat hepatoma cell line H4IIE to investigate the effects of AA at IC20 and IC50 concentrations on the redox status and the activity of CAT, SOD, and glutathione-S-transferase (GST), their gene expression, and CYP2E1 and iNOS expression. Immunohistochemically stained liver sections showed that treatment with AA25mg induced a significant decrease of CYP2E1 protein expression (p < 0.05), while treatment with AA50mg led to a significant increase of iNOS protein expression (p < 0.05). AA treatment dose-dependently elevated SOD2 protein expression (p < 0.05), while SOD1 protein expression was significantly increased only at AA50mg (p < 0.05). CAT protein expression was not significantly affected by AA treatments (p > 0.05). In AA-treated H4IIE cells, a concentration-dependent significant increase in lipid peroxidation and nitrite levels was observed (p < 0.05), while GSH content and SOD activity significantly decreased in a concentration-dependent manner (p < 0.05). AA IC50 significantly enhanced GST activity (p < 0.05). The level of mRNA significantly increased in a concentration-dependent manner for iNOS, SOD2, and CAT in AA-treated H4IIE cells (p < 0.05). AA IC50 significantly increased the transcription of SOD1, GSTA2, and GSTP1 genes (p < 0.05), while AA IC20 significantly decreased mRNA for CYP2E1 in H4IIE cells (p < 0.05). Obtained results indicate that AA treatments, both in vivo and in vitro, change hepatocytes; drug-metabolizing potential and disturb its redox status.
Collapse
Affiliation(s)
- Jelena Marković Filipović
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (D.K.); (J.K.); (I.I.); (M.M.)
| | - Marko Miler
- Department of Cytology, Institute for Biological Research “Siniša Stanković”-National Institute of Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11060 Belgrade, Serbia;
| | - Danijela Kojić
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (D.K.); (J.K.); (I.I.); (M.M.)
| | - Jelena Karan
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (D.K.); (J.K.); (I.I.); (M.M.)
| | - Ivana Ivelja
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (D.K.); (J.K.); (I.I.); (M.M.)
| | - Jovana Čukuranović Kokoris
- Department of Anatomy, Faculty of Medicine, University of Niš, Blvd. Dr Zorana Djindjica 81, 18000 Niš, Serbia;
| | - Milica Matavulj
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia; (D.K.); (J.K.); (I.I.); (M.M.)
| |
Collapse
|
6
|
Koga T, Peters JM. Targeting Peroxisome Proliferator-Activated Receptor-β/δ (PPARβ/δ) for the Treatment or Prevention of Alcoholic Liver Disease. Biol Pharm Bull 2021; 44:1598-1606. [PMID: 34719638 DOI: 10.1248/bpb.b21-00486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Excessive, chronic alcohol consumption can lead to alcoholic liver disease. The etiology of alcoholic liver disease is multifactorial and is influenced by alterations in gene expression and changes in fatty acid metabolism, oxidative stress, and insulin resistance. These events can lead to steatosis, fibrosis, and eventually to cirrhosis and liver cancer. Many of these functions are regulated by peroxisome proliferator-activated receptors (PPARs). Thus, it is not surprising that PPARs can modulate the mechanisms that cause alcoholic liver disease. While the roles of PPARα and PPARγ are clearer, the role of PPARβ/δ in alcoholic liver disease requires further clarification. This review summarizes the current understanding based on recent studies that indicate that PPARβ/δ can likely be targeted for the treatment and/or the prevention of alcoholic liver disease.
Collapse
Affiliation(s)
- Takayuki Koga
- Laboratory of Hygienic Chemistry, Department of Health Science and Hygiene, Daiichi University of Pharmacy
| | - Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, The Pennsylvania State University
| |
Collapse
|
7
|
Sauton P, Deschamps C, Jeanblanc V, Pierrefiche O, Jeanblanc J, Naassila M. Interstrain differences in voluntary binge-like drinking behavior and in two acute ethanol injections-induced synaptic plasticity deficits in rats. Addict Biol 2021; 26:e12992. [PMID: 33331070 DOI: 10.1111/adb.12992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/14/2022]
Abstract
Propensity to drink alcohol and to initiate binge drinking behavior is driven by genetic factors. Recently, we proposed an original animal model useful in the study of voluntary binge-like drinking (BD) in outbred Long-Evans rats by combining intermittent access to 20% ethanol in a two-bottle choice (IA2BC) paradigm to 15-min daily sessions of 20% ethanol operant self-administration. We sought to compare three strains of outbred rats (Long-Evans, Sprague-Dawley, and Wistar) in our BD model. Because we found different propensity to BD between strains, we also sought to test interstrain differences using another procedure of two acute ethanol exposures known to alter long-term depression of hippocampal synaptic plasticity. Our results demonstrate that in both IA2BC and operant procedures, the Long-Evans strain consumed the highest, Wistar the lowest amount of ethanol, and the Sprague-Dawley was intermediate. Long-Evans rats were also the fastest consuming with the shortest time to reach 50% of their maximum consumption in 15 min. When we tested the acute effects of ethanol, long-term depression in hippocampus was abolished specifically in Long-Evans rats with no impact in the two other strains. Thus, our study reveals that the Long-Evans strain is the ideal strain in our recently developed animal model useful in the study of BD. In addition, with the other paradigm of forced acute ethanol exposure, the Long-Evans strain displayed an increase in sensitivity to the deleterious effect of BD on hippocampal synaptic plasticity. Further studies are needed in order to investigate why Long-Evans rats are more prone to BD.
Collapse
Affiliation(s)
- Pierre Sauton
- INSERM UMR 1247 – Research Group on Alcohol & Pharmacodependences (GRAP) Université de Picardie Jules Verne Amiens France
| | - Chloé Deschamps
- INSERM UMR 1247 – Research Group on Alcohol & Pharmacodependences (GRAP) Université de Picardie Jules Verne Amiens France
| | - Virginie Jeanblanc
- Animal Facility PlatAnN Université de Picardie Jules Verne Amiens France
| | - Olivier Pierrefiche
- INSERM UMR 1247 – Research Group on Alcohol & Pharmacodependences (GRAP) Université de Picardie Jules Verne Amiens France
| | - Jérôme Jeanblanc
- INSERM UMR 1247 – Research Group on Alcohol & Pharmacodependences (GRAP) Université de Picardie Jules Verne Amiens France
| | - Mickael Naassila
- INSERM UMR 1247 – Research Group on Alcohol & Pharmacodependences (GRAP) Université de Picardie Jules Verne Amiens France
| |
Collapse
|
8
|
Liu SX, Du YC, Zeng T. A mini-review of the rodent models for alcoholic liver disease: shortcomings, application, and future prospects. Toxicol Res (Camb) 2021; 10:523-530. [PMID: 34141166 DOI: 10.1093/toxres/tfab042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/12/2021] [Accepted: 04/15/2021] [Indexed: 12/19/2022] Open
Abstract
Rodents are the most common models in studies of alcoholic liver disease (ALD). Although several rodents ALD models have been established and multiple mechanisms have been elucidated based on them, these models have some non-negligible shortcomings, specifically only inducing early stage (mainly steatosis, slight to moderate steatohepatitis) but not the whole spectrum of human ALD. The resistance of rodents to advanced ALD has been suggested to be due to the physiological differences between rodents and human beings. Previous studies have reported significant interstrain differences in the susceptibility to ethanol-induced liver injury and in the manifestation of ALD (such as different alteration of lipid profiles). Therefore, it would be interesting to characterize the manifestation of ethanol-induced liver damage in various rodents, which may provide a recommendation to investigators of ALD. Furthermore, more severe ALD models need to be established for the study of serious ALD forms, which may be achieved by using genetic modified rodents.
Collapse
Affiliation(s)
- Shi-Xuan Liu
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yan-Chao Du
- Jinan Institute for Product Quality Inspection, 1311 Longao Bei Road, Jinan, Shandong, 250102, China
| | - Tao Zeng
- Institute of Toxicology, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| |
Collapse
|
9
|
Kang K, Sun Y, Pan D, Sang LX, Sun MJ, Li YL, Chang B. Distinctive gut microbial dysbiosis between chronic alcoholic fatty liver disease and metabolic-associated fatty liver disease in mice. Exp Ther Med 2021; 21:418. [PMID: 33777186 PMCID: PMC7967804 DOI: 10.3892/etm.2021.9862] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota, which may affect normal physiological and biochemical functions, has an important role in the development of human liver diseases. The aim of the present study was to investigate differences in the gut microbiota between chronic alcoholic fatty liver disease (AFLD) and metabolic-associated fatty liver disease (MAFLD). AFLD was induced by chronic alcohol administration and MAFLD was induced by a Western-style diet in C57BL/6 mice. After 8 weeks, the levels of plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), triglyceride (TG), total cholesterol (TC), lipopolysaccharide (LPS), tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β and IL-10 were assessed and H&E staining of mouse liver tissue was performed. High-throughput sequencing of 16S ribosomal DNA from the intestinal contents was used to analyze the different effects of AFLD and MAFLD on the gut microbiota. Differences in the gut microbiota composition were assessed by the t-test. The results revealed increases in LPS, ALT, AST, TG, IL-1β and TNF-α in the AFLD group. Compared with those in the MAFLD control group, the MAFLD group exhibited increased plasma ALT, TG, TC, IL-6, IL-1β and TNF-α levels and decreased plasma IL-10 levels. In addition, the α- and β-diversities revealed that the AFLD and MAFLD groups exhibited obvious changes in the gut structure (with an increase in abundance in the AFLD group and a decrease in abundance in the MAFLD group). In comparison to the AFLD control group, Enterococcaceae were the most abundant bacteria at the family level and Enterococcus and Streptococcus were the most abundant bacteria at the genus level in the AFLD group. However, in the MAFLD group, Lachnospiraceae was the most abundant at the family level, with increases in Erysipelatoclostridium, Gordonibacter and Streptococcus at the genus level and a decrease in the genus Bifidobacterium. In conclusion, the present study confirmed that the AFLD and MAFLD groups harbored differences in the gut microbiota. The marked differences in the gut microbiota at the family and genus levels may contribute to the development process of FLD.
Collapse
Affiliation(s)
- Kai Kang
- Department of Gastroenterology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yue Sun
- Department of Gastroenterology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Dan Pan
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Li-Xuan Sang
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ming-Jun Sun
- Department of Gastroenterology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi-Ling Li
- Department of Gastroenterology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Bing Chang
- Department of Gastroenterology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
10
|
Hartmann P, Chu H, Duan Y, Schnabl B. Gut microbiota in liver disease: too much is harmful, nothing at all is not helpful either. Am J Physiol Gastrointest Liver Physiol 2019; 316:G563-G573. [PMID: 30767680 PMCID: PMC6580239 DOI: 10.1152/ajpgi.00370.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/15/2019] [Accepted: 02/15/2019] [Indexed: 01/31/2023]
Abstract
The intestinal microbiome plays a major role in the pathogenesis of liver disease, with a hallmark event being dysbiosis, or an imbalance of pathobionts and beneficial bacteria with the associated deleterious effects on their host. Reducing the number of intestinal bacteria with antibiotic treatment is generally advantageous in experimental liver diseases. Complete absence of intestinal microbiota as in germ-free rodents can be protective in autoimmune hepatitis and hepatic tumors induced by chemicals, or it can exacerbate disease as in acute toxic liver injury and liver fibrosis/cirrhosis. In alcoholic liver disease, nonalcoholic fatty liver disease, and autoimmune cholangiopathies, germ-free status can be associated with worsened or improved hepatic phenotype depending on the experimental model and type of rodent. Some of the unexpected outcomes can be explained by the limitations of rodents raised in a germ-free environment including a deficient immune system and an altered metabolism of lipids, cholesterol, xenobiotics/toxins, and bile acids. Given these limitations and to advance understanding of the interactions between host and intestinal microbiota, simplified model systems such as humanized gnotobiotic mice, or gnotobiotic mice monoassociated with a single bacterial strain or colonized with a defined set of microbes, are unique and useful models for investigation of liver disease in a complex ecosystem.
Collapse
Affiliation(s)
- Phillipp Hartmann
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology , Wuhan , China
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Yi Duan
- Department of Medicine, University of California, San Diego, La Jolla, California
- Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, California
- Department of Medicine, Veterans Affairs San Diego Healthcare System, San Diego, California
| |
Collapse
|
11
|
Lamas-Paz A, Hao F, Nelson LJ, Vázquez MT, Canals S, Gómez del Moral M, Martínez-Naves E, Nevzorova YA, Cubero FJ. Alcoholic liver disease: Utility of animal models. World J Gastroenterol 2018; 24:5063-5075. [PMID: 30568384 PMCID: PMC6288648 DOI: 10.3748/wjg.v24.i45.5063] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is a major cause of acute and chronic liver injury. Extensive evidence has been accumulated on the pathological process of ALD during the past decades. However, effective treatment options for ALD are very limited due to the lack of suitable in vivo models that recapitulate the full spectrum of ALD. Experimental animal models of ALD, particularly rodents, have been used extensively to mimic human ALD. An ideal animal model should recapitulate all aspects of the ALD process, including significant steatosis, hepatic neutrophil infiltration, and liver injury. A better strategy against ALD depends on clear diagnostic biomarkers, accurate predictor(s) of its progression and new therapeutic approaches to modulate stop or even reverse the disease. Numerous models employing rodent animals have been established in the last decades to investigate the effects of acute and chronic alcohol exposure on the initiation and progression of ALD. Although significant progress has been made in gaining better knowledge on the mechanisms and pathology of ALD, many features of ALD are unknown, and require further investigation, ideally with improved animal models that more effectively mimic human ALD. Although differences in the degree and stages of alcoholic liver injury inevitably exist between animal models and human ALD, the acquisition and translational relevance will be greatly enhanced with the development of new and improved animal models of ALD.
Collapse
Affiliation(s)
- Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Fengjie Hao
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Leonard J Nelson
- Institute for Bioengineering (IBioE), School of Engineering, Faraday Building, The University of Edinburgh, Edinburgh EH9 3 JL, Scotland, United Kingdom
| | - Maria Teresa Vázquez
- Department of Human Anatomy and Embryology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante 03550, Spain
| | - Manuel Gómez del Moral
- Department of Cell Biology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Yulia A Nevzorova
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Universidad Complutense, Madrid 28040, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen 52062, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
| |
Collapse
|
12
|
Ma L, Huo CY, Zhang XY, Qin CQ, Ren DF, Lu J. Protective effect of Letinous edodes foot peptides against ethanol‑induced liver injury in L02 cells. Mol Med Rep 2018; 18:1858-1866. [PMID: 29845248 DOI: 10.3892/mmr.2018.9093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 05/09/2018] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to evaluate the protective effect and mechanism of Letinous edodes foot peptides on ethanol‑induced L02 cells. A cell model of ethanol‑induced damage was established in vitro to study the effects of the Letinous edodes foot peptides on human L02 hepatocytes. The expression and activity of superoxide dismutase (SOD), malondialdehyde (MDA), aspartate aminotransferase (AST), alanine aminotransferase (ALT), alcohol dehydrogenase (ADH) and acetaldehyde dehydrogenase (ALDH), following treatment were examined to determine the anti‑alcoholism and hepatoprotective functions of Letinous edodes foot peptides. Taking Letinous edodes foot peptides prior to ethanol exposure was more beneficial, which significantly increased SOD activity and the mRNA expression of ADH and ALDH suppressed by ethanol. In addition, the intracellular MDA content, and AST and ALT activity decreased in ethanol‑induced L02 cells pretreated with the peptides, when compared with the control. Furthermore, Letinous edodes foot peptides inhibited the ethanol‑induced activation of the proinflammatory cytokines, interleukin‑6 and tumor necrosis factor‑α, and promoted the metabolic regulation factors, AMP‑activated protein kinase‑α2 and peroxisome proliferator‑activated receptor‑α.
Collapse
Affiliation(s)
- Lin Ma
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Chun-Yan Huo
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Xiao-Yu Zhang
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Chen-Qiang Qin
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Di-Feng Ren
- Beijing Key Laboratory of Forest Food Processing and Safety, College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing 100083, P.R. China
| | - Jun Lu
- Beijing Engineering Research Center of Protein and Functional Peptides, China National Research Institute of Food and Fermentation Industries, Beijing 100015, P.R. China
| |
Collapse
|
13
|
Papp-Peka A, Tong M, Kril JJ, De La Monte SM, Sutherland GT. The Differential Effects of Alcohol and Nicotine-Specific Nitrosamine Ketone on White Matter Ultrastructure. Alcohol Alcohol 2017; 52:165-171. [PMID: 28182194 PMCID: PMC6075461 DOI: 10.1093/alcalc/agw067] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/15/2016] [Accepted: 08/29/2016] [Indexed: 01/04/2023] Open
Abstract
Aims The chronic consumption of alcohol is known to result in neurodegeneration and impairment of cognitive function. Pathological and neuroimaging studies have confirmed that brain atrophy in alcoholics is mainly due to widespread white matter (WM) loss with neuronal loss restricted to specific regions, such as the prefrontal cortex. Neuroimaging studies of cigarette smokers also suggest that chronic inhalation of tobacco smoke leads to brain atrophy, although the neurotoxic component is unknown. As a high proportion of chronic alcoholics also smoke cigarettes it has been hypothesized that at least some alcohol-related brain damage is due to tobacco smoke exposure. Methods 39 Long Evans rats were subjected to 8 weeks exposure to alcohol and/or 5 weeks co-exposure to nicotine-specific nitrosamine ketone (NNK), a proxy for tobacco smoke. Their frontal WM was then assayed with transmission electron microscopy. Results NNK and ethanol co-exposure had a synergistic effect in decreasing myelinated fibre density. Furthermore, NNK treatment led to a greater reduction in myelin sheath thickness than ethanol whereas only the ethanol-treated animals showed a decrease in unmyelinated fibre density. Conclusion These data suggest that NNK causes WM degeneration, an effect that is exacerbated by alcohol, but unlike alcohol, it has little impact on the neuronal components of the brain.
Collapse
Affiliation(s)
- A Papp-Peka
- Charles Perkins Centre, Discipline of Pathology, School of Medical Sciences, The University of Sydney, Johns Hopkins Drive, Camperdown NSW 2050, Australia
| | | | - J J Kril
- Charles Perkins Centre, Discipline of Pathology, School of Medical Sciences, The University of Sydney, Johns Hopkins Drive, Camperdown NSW 2050, Australia
| | | | - G T Sutherland
- Charles Perkins Centre, Discipline of Pathology, School of Medical Sciences, The University of Sydney, Johns Hopkins Drive, Camperdown NSW 2050, Australia
| |
Collapse
|
14
|
Deochand C, Tong M, Agarwal AR, Cadenas E, de la Monte SM. Tobacco Smoke Exposure Impairs Brain Insulin/IGF Signaling: Potential Co-Factor Role in Neurodegeneration. J Alzheimers Dis 2016; 50:373-86. [PMID: 26682684 DOI: 10.3233/jad-150664] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Human studies suggest tobacco smoking is a risk factor for cognitive impairment and neurodegeneration, including Alzheimer's disease (AD). However, experimental data linking tobacco smoke exposures to underlying mediators of neurodegeneration, including impairments in brain insulin and insulin-like growth factor (IGF) signaling in AD are lacking. OBJECTIVE This study tests the hypothesis that cigarette smoke (CS) exposures can impair brain insulin/IGF signaling and alter expression of AD-associated proteins. METHODS Adult male A/J mice were exposed to air for 8 weeks (A8), CS for 4 or 8 weeks (CS4, CS8), or CS8 followed by 2 weeks recovery (CS8+R). Gene expression was measured by qRT-PCR analysis and proteins were measured by multiplex bead-based or direct binding duplex ELISAs. RESULTS CS exposure effects on insulin/IGF and insulin receptor substrate (IRS) proteins and phosphorylated proteins were striking compared with the mRNA. The main consequences of CS4 or CS8 exposures were to significantly reduce insulin R, IGF-1R, IRS-1, and tyrosine phosphorylated insulin R and IGF-1R proteins. Paradoxically, these effects were even greater in the CS8+R group. In addition, relative levels of S312-IRS-1, which inhibits downstream signaling, were increased in the CS4, CS8, and CS8+R groups. Correspondingly, CS and CS8+R exposures inhibited expression of proteins and phosphoproteins required for signaling through Akt, PRAS40, and/or p70S6K, increased AβPP-Aβ, and reduced ASPH protein, which is a target of insulin/IGF-1 signaling. CONCLUSION Secondhand CS exposures caused molecular and biochemical abnormalities in brain that overlap with the findings in AD, and many of these effects were sustained or worsened despite short-term CS withdrawal.
Collapse
Affiliation(s)
- Chetram Deochand
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Divisions of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Divisions of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Amit R Agarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Suzanne M de la Monte
- Liver Research Center, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Divisions of Gastroenterology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Divisions of Neuropathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA.,Department of Neurosurgery, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
15
|
Colman K. Impact of the Genetics and Source of Preclinical Safety Animal Models on Study Design, Results, and Interpretation. Toxicol Pathol 2016; 45:94-106. [DOI: 10.1177/0192623316672743] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
It has been long established that not only the species but also the strain and supplier of rodents used in preclinical safety studies can have a significant impact on the outcome of studies due to variability in their genetic background and thus spontaneous pathologic findings. In addition, local husbandry, housing, and other environmental conditions may have effects on the development and expression of comorbidities, particularly in longer-term or chronic studies. More recently, similar effects related to the source, including genetic and environmental variability, have been recognized in cynomolgus macaques ( Macaca fascicularis). The increased use of cynomolgus macaques from various sources of captive-bred animals (including nonnative, U.S./European Union-based breeding facilities or colonies) can affect study design and study results and outcome. It is important to acknowledge and understand the impact of this variability on the results and interpretation of research studies. This review includes recent examples where variability of preclinical animal models (rats and monkeys) affected the postmortem observations highlighting its relevance to study design or interpretation in safety studies.
Collapse
Affiliation(s)
- Karyn Colman
- Novartis Pharmaceuticals Corp., East Hanover, New Jersey, USA
| |
Collapse
|
16
|
Wang HJ, Murray GJ, Jung MK. Host homeostatic responses to alcohol-induced cellular stress in animal models of alcoholic liver disease. Expert Rev Gastroenterol Hepatol 2016; 9:1193-205. [PMID: 26293978 DOI: 10.1586/17474124.2015.1069705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Humans develop various clinical phenotypes of severe alcoholic liver disease, including alcoholic hepatitis and cirrhosis, generally after decades of heavy drinking. In such individuals, following each episode of drinking, their livers experience heightened intracellular and extracellular stresses that are closely associated with alcohol consumption and alcohol metabolism. This article focuses on the latest advances made in animal models on evolutionarily conserved homeostatic mechanisms for coping with and resolving these stress conditions. The mechanisms discussed include the stress-activated protein kinase JNK, energy regulator AMPK, autophagy and the inflammatory response. Over time, the host may respond variably to stress with protective mechanisms that are critical in determining an individual's vulnerability to developing severe alcoholic liver disease. A systematic review of these mechanisms and their temporal changes in animal models provides the basis for general conclusions, and raises questions for future studies. The relevance of these data to human conditions is also discussed.
Collapse
Affiliation(s)
- He Joe Wang
- a Division of Metabolism and Health Effect, National Institute of Alcohol Abuse and Alcoholism/NIH, 5635 Fishers Lane, MSC 9304, Bethesda, MD 20892-9304, USA
| | | | | |
Collapse
|
17
|
Liver-Specific Deletion of Phosphatase and Tensin Homolog Deleted on Chromosome 10 Significantly Ameliorates Chronic EtOH-Induced Increases in Hepatocellular Damage. PLoS One 2016; 11:e0154152. [PMID: 27124661 PMCID: PMC4849752 DOI: 10.1371/journal.pone.0154152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/08/2016] [Indexed: 12/20/2022] Open
Abstract
Alcoholic liver disease is a significant contributor to global liver failure. In murine models, chronic ethanol consumption dysregulates PTEN/Akt signaling. Hepatospecific deletion of phosphatase and tensin homolog deleted on chromosome 10 (PTENLKO) mice possess constitutive activation of Akt(s) and increased de novo lipogenesis resulting in increased hepatocellular steatosis. This makes PTENLKO a viable model to examine the effects of ethanol in an environment of preexisting steatosis. The aim of this study was to determine the impact of chronic ethanol consumption and the absence of PTEN (PTENLKO) compared to Alb-Cre control mice (PTENf/f) on hepatocellular damage as evidenced by changes in lipid accumulation, protein carbonylation and alanine amino transferase (ALT). In the control PTENf/f animals, ethanol significantly increased ALT, liver triglycerides and steatosis. In contrast, chronic ethanol consumption in PTENLKO mice decreased hepatocellular damage when compared to PTENLKO pair-fed controls. Consumption of ethanol elevated protein carbonylation in PTENf/f animals but had no effect in PTENLKO animals. In PTENLKO mice, overall hepatic mRNA expression of genes that contribute to GSH homeostasis as well as reduced glutathione (GSH) and oxidized glutathione (GSSG) concentrations were significantly elevated compared to respective PTENf/f counterparts. These data indicate that during conditions of constitutive Akt activation and steatosis, increased GSH homeostasis assists in mitigation of ethanol-dependent induction of oxidative stress and hepatocellular damage. Furthermore, data herein suggest a divergence in EtOH-induced hepatocellular damage and increases in steatosis due to polyunsaturated fatty acids downstream of PTEN.
Collapse
|
18
|
Andreani T, Tong M, Gundogan F, Silbermann E, de la Monte SM. Differential Effects of 3rd Trimester-Equivalent Binge Ethanol and Tobacco-Specific Nitrosamine Ketone Exposures on Brain Insulin Signaling in Adolescence. JOURNAL OF DIABETES AND RELATED DISORDERS 2016; 1:105. [PMID: 29242853 PMCID: PMC5726776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Fetal alcohol spectrum disorder (FASD) is associated with impairments in insulin and insulin-like growth factor (IGF) signaling through Akt pathways and altered expression of neuro-glial proteins needed for structural and functional integrity of the brain. However, alcohol abuse correlates with smoking, and tobacco smoke contains 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), which like other nitrosamines, impairs insulin and IGF signaling. HYPOTHESIS NNK exposure can serve as a co-factor in mediating long-term neuro-developmental abnormalities associated with FASD. DESIGN Long Evans rat pups were IP administered ethanol (2 g/kg) on postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7, simulating third trimester human exposures. Temporal lobes from P30 rats (young adolescent) were used to measure signaling through the insulin/IGF-1/Akt pathways by multiplex ELISAs, and expression of neuroglial proteins by duplex ELISAs. RESULTS Ethanol, NNK, and ethanol + NNK exposures significantly inhibited insulin receptor tyrosine phosphorylation, and IRS-1 and myelin-associated glycoprotein expression. However, the major long-term adverse effects on Akt pathway downstream signaling and its targeted proteins including choline acetyltransferase, Tau, pTau, ubiquitin, and aspartate-β-hydroxylase were due to NNK rather than ethanol. CONCLUSION Alcohol and tobacco exposures can both contribute to long-term brain abnormalities currently regarded fetal ethanol effects. However, the findings suggest that many of the adverse effects on brain function are attributable to smoking, including impairments in signaling through survival and metabolic pathways, and altered expression of genes that regulate myelin synthesis, maturation and integrity and synaptic plasticity. Therefore, public health measures should address both substances of abuse to prevent "FASD".
Collapse
Affiliation(s)
- Tomas Andreani
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| | - Fusun Gundogan
- Department of Pathology, Women and Infants Hospital of Rhode Island,
Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| | | | - Suzanne M. de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver
Research Center Rhode Island Hospital, Providence, RI, USA
- Departments of Pathology and Neurology, and the Division of
Neuropathology, Rhode Island Hospital, Providence, RI, USA
- Warren Alpert Medical School of Brown University, Providence, RI,
USA
| |
Collapse
|
19
|
Steiner JL, Crowell KT, Lang CH. Impact of Alcohol on Glycemic Control and Insulin Action. Biomolecules 2015; 5:2223-46. [PMID: 26426068 PMCID: PMC4693236 DOI: 10.3390/biom5042223] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 08/24/2015] [Accepted: 09/21/2015] [Indexed: 02/06/2023] Open
Abstract
Alcohol has profound effects on tissue and whole-body fuel metabolism which contribute to the increased morbidity and mortality in individuals with alcohol use disorder. This review focuses on the glucose metabolic effects of alcohol, primarily in the muscle, liver and adipose tissue, under basal postabsorptive conditions and in response to insulin stimulation. While there is a relatively extensive literature in this area, results are often discordant and extrapolating between models and tissues is fraught with uncertainty. Comparisons between data generated in experimental cell and animals systems will be contrasted with that obtained from human subjects as often times results differ. Further, the nutritional status is also an important component of the sometimes divergent findings pertaining to the effects of alcohol on the regulation of insulin and glucose metabolism. This work is relevant as the contribution of alcohol intake to the development or exacerbation of type 2 diabetes remains ill-defined and a multi-systems approach is likely needed as both alcohol and diabetes affect multiple targets within the body.
Collapse
Affiliation(s)
- Jennifer L Steiner
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Kristen T Crowell
- Department of Surgery, Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA.
- Department of Surgery, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
20
|
Tong M, Yu R, Deochand C, de la Monte SM. Differential Contributions of Alcohol and the Nicotine-Derived Nitrosamine Ketone (NNK) to Insulin and Insulin-Like Growth Factor Resistance in the Adolescent Rat Brain. Alcohol Alcohol 2015; 50:670-9. [PMID: 26373814 DOI: 10.1093/alcalc/agv101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/17/2015] [Indexed: 12/11/2022] Open
Abstract
AIMS Since epidemiologic studies suggest that tobacco smoke toxins, e.g. the nicotine-derived nitrosamine ketone (NNK) tobacco-specific nitrosamine, can be a co-factor in alcohol-related brain disease (ARBD), we examined the independent and additive effects of alcohol and NNK exposures on spatial learning/memory, and brain insulin/IGF signaling, neuronal function and oxidative stress. METHODS Adolescent Long Evans rats were fed liquid diets containing 0 or 26% caloric ethanol for 8 weeks. During weeks 3-8, rats were treated with i.p. NNK (2 mg/kg, 3×/week) or saline. In weeks 7-8, ethanol groups were binge-administered ethanol (2 g/kg; 3×/week). In week 8, at 12 weeks of age, rats were subjected to Morris Water Maze tests. Temporal lobes were used to assess molecular indices of insulin/IGF resistance, oxidative stress and neuronal function. RESULTS Ethanol and NNK impaired spatial learning, and NNK ± ethanol impaired memory. Linear trend analysis demonstrated worsening performance from control to ethanol, to NNK, and then ethanol + NNK. Ethanol ± NNK, caused brain atrophy, inhibited insulin signaling through the insulin receptor and Akt, activated GSK-3β, increased protein carbonyl and 3-nitrotyrosine, and reduced acetylcholinesterase. NNK increased NTyr. Ethanol + NNK had synergistic stimulatory effects on 8-iso-PGF-2α, inhibitory effects on p-p70S6K, tau and p-tau and trend effects on insulin-like growth factor type 1 (IGF-1) receptor expression and phosphorylation. CONCLUSIONS Ethanol, NNK and combined ethanol + NNK exposures that begin in adolescence impair spatial learning and memory in young adults. The ethanol and/or NNK exposures differentially impair insulin/IGF signaling through neuronal growth, survival and plasticity pathways, increase cellular injury and oxidative stress and reduce expression of critical proteins needed for neuronal function.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rosa Yu
- Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI, USA
| | - Chetram Deochand
- Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
21
|
Bhopale KK, Kondraganti S, Fernando H, Boor PJ, Kaphalia BS, Shakeel Ansari GA. Alcoholic Steatosis in Different Strains of Rat: A Comparative Study. ACTA ACUST UNITED AC 2015; 4. [PMID: 27213081 PMCID: PMC4874529 DOI: 10.4303/jdar/235912] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Different strains of rats have been used to study alcoholic liver disease (ALD) while the reason for selecting a particular rat strain was not apparent. Purpose The aim of our study was to compare outbred (Wistar) and inbred (Fischer) strains to evaluate pathological, biochemical changes, and gene expression differences associated with ethanol-induced early hepatic steatosis. Study Design Male Wistar and Fischer-344 rats were pair-fed for 6 weeks with or without 5% ethanol in Lieber-DeCarli liquid diet. Livers were analyzed for histological and lipid-related differences. Results Hepatic midzonal steatosis was mainly found in Wistar rats while Fischer rats showed mostly pericentral steatosis. Increased hepatic steatosis in ethanol-fed Wistar rats is supported by increases in lipids with related genes and transcription factors involved in fatty acid and triglyceride synthesis. Conclusion Our data showed that Fischer rats are relatively less prone to ethanol-mediated steatosis with pericentral lipid deposition pattern in the liver which is similar to humans and show no trace level of lipid accumulation in pair-fed controls as observed in Wistar (outbred) strain. Therefore, Fischer rats are better suited for lipid studies in an early development of ALD.
Collapse
Affiliation(s)
- Kamlesh K Bhopale
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shakuntala Kondraganti
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Harshica Fernando
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Paul J Boor
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bhupendra S Kaphalia
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| | - G A Shakeel Ansari
- Department of Pathology, The University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
22
|
Carr RM, Correnti J. Insulin resistance in clinical and experimental alcoholic liver disease. Ann N Y Acad Sci 2015; 1353:1-20. [PMID: 25998863 DOI: 10.1111/nyas.12787] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alcoholic liver disease (ALD) is the number one cause of liver failure worldwide; its management costs billions of healthcare dollars annually. Since the advent of the obesity epidemic, insulin resistance (IR) and diabetes have become common clinical findings in patients with ALD; and the development of IR predicts the progression from simple steatosis to cirrhosis in ALD patients. Both clinical and experimental data implicate the impairment of several mediators of insulin signaling in ALD, and experimental data suggest that insulin-sensitizing therapies improve liver histology. This review explores the contribution of impaired insulin signaling in ALD and summarizes the current understanding of the synergistic relationship between alcohol and nutrient excess in promoting hepatic inflammation and disease.
Collapse
Affiliation(s)
- Rotonya M Carr
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jason Correnti
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
23
|
Zabala V, Tong M, Yu R, Ramirez T, Yalcin EB, Balbo S, Silbermann E, Deochand C, Nunez K, Hecht S, de la Monte SM. Potential contributions of the tobacco nicotine-derived nitrosamine ketone (NNK) in the pathogenesis of steatohepatitis in a chronic plus binge rat model of alcoholic liver disease. Alcohol Alcohol 2015; 50:118-31. [PMID: 25618784 DOI: 10.1093/alcalc/agu083] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIMS Alcoholic liver disease (ALD) is linked to binge drinking and cigarette smoking. Heavy chronic ± binge alcohol, or low-level exposures to dietary nitrosamines cause steatohepatitis with insulin resistance and oxidative stress in animal models. This study examines hepatotoxic effects of sub-mutagenic exposures to tobacco-specific nitrosamine (NNK) in relation to ALD. METHODS Long Evans rats were fed liquid diets containing 0 or 26% (caloric) ethanol (EtOH) for 8 weeks. In Weeks 3 through 8, rats were treated with NNK (2 mg/kg) or saline by i.p. injection, 3×/week, and in Weeks 7 and 8, EtOH-fed rats were binge-administered 2 g/kg EtOH 3×/week; controls were given saline. RESULTS EtOH ± NNK caused steatohepatitis with necrosis, disruption of the hepatic cord architecture, ballooning degeneration, early fibrosis, mitochondrial cytopathy and ER disruption. Severity of lesions was highest in the EtOH+NNK group. EtOH and NNK inhibited insulin/IGF signaling through Akt and activated pro-inflammatory cytokines, while EtOH promoted lipid peroxidation, and NNK increased apoptosis. O(6)-methyl-Guanine adducts were only detected in NNK-exposed livers. CONCLUSION Both alcohol and NNK exposures contribute to ALD pathogenesis, including insulin/IGF resistance and inflammation. The differential effects of EtOH and NNK on adduct formation are critical to ALD progression among alcoholics who smoke.
Collapse
Affiliation(s)
- Valerie Zabala
- Department of Medicine, Division of Gastroenterology, and The Liver Research Center, Rhode Island Hospital, Providence, RI, USA Department of Molecular Pharmacology and Physiology, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Division of Gastroenterology, and The Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rosa Yu
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Teresa Ramirez
- Department of Molecular Pharmacology and Physiology, Brown University, Providence, RI, USA
| | - Emine B Yalcin
- Department of Medicine, Division of Gastroenterology, and The Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN 55455, USA
| | | | - Chetram Deochand
- Biotechnology Graduate Program, Brown University, Providence, RI, USA
| | - Kavin Nunez
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Stephen Hecht
- Masonic Cancer Center, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN 55455, USA
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and The Liver Research Center, Rhode Island Hospital, Providence, RI, USA Warren Alpert Medical School of Brown University, Providence, RI, USA Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
24
|
Carr RM, Peralta G, Yin X, Ahima RS. Absence of perilipin 2 prevents hepatic steatosis, glucose intolerance and ceramide accumulation in alcohol-fed mice. PLoS One 2014; 9:e97118. [PMID: 24831094 PMCID: PMC4022498 DOI: 10.1371/journal.pone.0097118] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/14/2014] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Perilipin 2 (Plin2) is a lipid droplet protein that has roles in both lipid and glucose homeostasis. An increase in Plin2 in liver is associated with the development of steatosis, glucose intolerance, and ceramide accumulation in alcoholic liver disease. We investigated the role of Plin2 on energy balance and glucose and lipid homeostasis in wildtype and Plin2 knockout (Plin2KO) mice chronically fed a Lieber-DeCarli liquid ethanol or control diet for six weeks. METHODS We performed in vivo measurements of energy intake and expenditure; body composition; and glucose tolerance. After sacrifice, liver was dissected for histology and lipid analysis. RESULTS We found that neither genotype nor diet had a significant effect on final weight, body composition, or energy intake between WT and Plin2KO mice fed alcohol or control diets. Additionally, alcohol feeding did not affect oxygen consumption or carbon dioxide production in Plin2KO mice. We performed glucose tolerance testing and observed that alcohol feeding failed to impair glucose tolerance in Plin2KO mice. Most notably, absence of Plin2 prevented hepatic steatosis and ceramide accumulation in alcohol-fed mice. These changes were related to downregulation of genes involved in lipogenesis and triglyceride synthesis. CONCLUSIONS Plin2KO mice chronically fed alcohol are protected from hepatic steatosis, glucose intolerance, and hepatic ceramide accumulation, suggesting a critical pathogenic role of Plin2 in experimental alcoholic liver disease.
Collapse
Affiliation(s)
- Rotonya M. Carr
- University of Pennsylvania, Perelman School of Medicine, Department of Medicine, Gastroenterology Division, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Giselle Peralta
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| | - Xiaoyan Yin
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| | - Rexford S. Ahima
- University of Pennsylvania, Perelman School of Medicine, Institute for Diabetes, Obesity and Metabolism, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
25
|
Tong M, Longato L, Ramirez T, Zabala V, Wands JR, de la Monte SM. Therapeutic reversal of chronic alcohol-related steatohepatitis with the ceramide inhibitor myriocin. Int J Exp Pathol 2013; 95:49-63. [PMID: 24456332 DOI: 10.1111/iep.12052] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2013] [Accepted: 08/18/2013] [Indexed: 12/13/2022] Open
Abstract
Alcohol-related liver disease (ALD) is associated with steatohepatitis and insulin resistance. Insulin resistance impairs growth and disrupts lipid metabolism in hepatocytes. Dysregulated lipid metabolism promotes ceramide accumulation and oxidative stress, leading to lipotoxic states that activate endoplasmic reticulum (ER) stress pathways and worsen inflammation and insulin resistance. In a rat model of chronic alcohol feeding, we characterized the effects of a ceramide inhibitor, myriocin, on the histopathological and ultrastructural features of steatohepatitis, and the biochemical and molecular indices of hepatic steatosis, insulin resistance and ER stress. Myriocin reduced the severity of alcohol-related steatohepatitis including the abundance and sizes of lipid droplets and mitochondria, inflammation and architectural disruption of the ER. In addition, myriocin-mediated reductions in hepatic lipid and ceramide levels were associated with constitutive enhancement of insulin signalling through the insulin receptor and IRS-2, reduced hepatic oxidative stress and modulation of ER stress signalling mechanisms. In conclusion, ceramide accumulation in liver mediates tissue injury, insulin resistance and lipotoxicity in ALD. Reducing hepatic ceramide levels can help restore the structural and functional integrity of the liver in chronic ALD due to amelioration of insulin resistance and ER stress. However, additional measures are needed to protect the liver from alcohol-induced necroinflammatory responses vis-à-vis continued alcohol abuse.
Collapse
Affiliation(s)
- Ming Tong
- Liver Research Center, Division of Gastroenterology, Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | | | | | | | | |
Collapse
|
26
|
Carr RM, Dhir R, Yin X, Agarwal B, Ahima RS. Temporal effects of ethanol consumption on energy homeostasis, hepatic steatosis, and insulin sensitivity in mice. Alcohol Clin Exp Res 2013; 37:1091-9. [PMID: 23398239 DOI: 10.1111/acer.12075] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 11/20/2012] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alcoholic liver disease (ALD) progresses from steatosis to inflammation, fibrosis, and cirrhosis. Although ALD has been associated with insulin resistance, it is unclear whether insulin resistance coincides with the development of steatosis. METHODS We studied the temporal relationship of steatosis and glucose homeostasis in mice fed a Lieber-DeCarli liquid control or ethanol (EtOH) diet for 2, 4, or 8 weeks. We studied the effects of alcohol consumption on energy balance, body composition, and hepatic lipids. Glucose tolerance test was performed, and insulin sensitivity was evaluated with hyperinsulinemic-euglycemic clamp. RESULTS EtOH-fed mice developed hepatic steatosis over time as compared with control-fed mice despite similar energy intake and expenditure, and gain in body weight and fat. EtOH-fed mice developed glucose intolerance as early as 2 weeks, while insulin resistance developed at 4 weeks. A hyperinsulinemic clamp study at 8 weeks revealed both hepatic and peripheral insulin resistance in EtOH-fed mice. Insulin resistance was associated with hepatic steatosis, increased ceramide levels, and Perilipin 2 expression. CONCLUSIONS Chronic EtOH consumption leads to the development of hepatic steatosis, impaired glucose tolerance, and insulin resistance. These changes are independent of energy intake or expenditure, weight, whole body fat content, and inflammation. A better understanding of the processes linking EtOH-induced steatosis and abnormal glucose homeostasis may lead to novel therapies targeting the progression of ALD.
Collapse
Affiliation(s)
- Rotonya M Carr
- Department of Medicine, Gastroenterology Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
27
|
Ramirez T, Tong M, Chen WC, Nguyen QG, Wands JR, de la Monte SM. Chronic alcohol-induced hepatic insulin resistance and endoplasmic reticulum stress ameliorated by peroxisome-proliferator activated receptor-δ agonist treatment. J Gastroenterol Hepatol 2013; 28:179-87. [PMID: 22988930 PMCID: PMC4406771 DOI: 10.1111/j.1440-1746.2012.07256.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/10/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Chronic alcoholic liver disease is associated with hepatic insulin resistance, dysregulated lipid metabolism with increased toxic lipid (ceramide) accumulation, lipid peroxidation, and oxidative and endoplasmic reticulum (ER) stress. Peroxisome-proliferator activated receptor (PPAR) agonists are insulin sensitizers that can restore hepatic insulin responsiveness in both alcohol and non-alcohol-related steatohepatitis. Herein, we demonstrate that treatment with a PPAR-δ agonist enhances insulin signaling and reduces the severities of ER stress and ceramide accumulation in an experimental model of ethanol-induced steatohepatitis. METHODS Adult male Long Evans rats were pair fed with isocaloric liquid diets containing 0% or 37% ethanol (caloric) for 8 weeks. After 3 weeks on the diets, rats were treated with vehicle or PPAR-δ agonist twice weekly by i.p. injection. RESULTS Ethanol-fed rats developed steatohepatitis with inhibition of signaling through the insulin and insulin-like growth factor-1 receptors, and Akt activated pathways. Despite continued ethanol exposure, PPAR-δ agonist co-treatments increased Akt activation, reduced multiple molecular indices of ER stress and steatohepatitis. CONCLUSIONS These results suggest that PPAR-δ agonist rescue of chronic alcoholic liver disease is mediated by enhancement of insulin signaling through Akt/metabolic pathways that reduce lipotoxicity and ER stress.
Collapse
Affiliation(s)
- Teresa Ramirez
- Liver Research Center and Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island 02903, USA
| | | | | | | | | | | |
Collapse
|
28
|
Brandon-Warner E, Schrum LW, Schmidt CM, McKillop IH. Rodent models of alcoholic liver disease: of mice and men. Alcohol 2012; 46:715-25. [PMID: 22960051 DOI: 10.1016/j.alcohol.2012.08.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 07/18/2012] [Accepted: 08/14/2012] [Indexed: 02/07/2023]
Abstract
Alcoholic liver disease (ALD) is a major cause of acute and chronic liver disease worldwide. The progressive nature of ALD is well described; however, the complex interactions under which these pathologies evolve remain to be fully elucidated. Clinically there are no clear biomarkers or universally accepted, effective treatment strategies for ALD. Experimental models of ALD are an important component in identifying underlying mechanisms of alcohol-induced injury to develop better diagnostic markers, predictors of disease progression, and therapeutic targets to manage, halt, or reverse disease progression. Rodents remain the most accessible model for studying ALD pathology. Effective rodent models must mimic the natural history of ALD while allowing examination of complex interactions between multiple hepatic, and non-hepatic, cell types in the setting of altered metabolic or oxidative/nitrosative stress, inflammatory responses, and sensitivity to cytotoxic stress. Additionally, mode and duration of alcohol delivery influence hepatic response and present unique challenges in understanding disease pathology. This review provides an overview of rodent models of ALD, their strengths and weaknesses relative to human disease states, and provides insight of the potential to develop novel rodent models to simulate the course of human ALD.
Collapse
|
29
|
Ramirez T, Longato L, Dostalek M, Tong M, Wands JR, de la Monte SM. Insulin resistance, ceramide accumulation and endoplasmic reticulum stress in experimental chronic alcohol-induced steatohepatitis. Alcohol Alcohol 2012; 48:39-52. [PMID: 22997409 DOI: 10.1093/alcalc/ags106] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIMS Chronic alcohol abuse causes steatohepatitis with insulin resistance, which impairs hepatocellular growth, survival and metabolism. However, growing evidence supports the concept that progressive alcohol-related liver injury may be mediated by concurrent mal-signaling through other networks that promote insulin resistance, e.g. pro-inflammatory, pro-ceramide and endoplasmic reticulum (ER) stress cascades. METHODS Using the Long Evans rat model of chronic ethanol feeding, we characterized the histopathologic and ultrastructural features of steatohepatitis in relation to biochemical and molecular indices of tissue injury, inflammation, insulin resistance, dysregulated lipid metabolism and ER stress. RESULTS Chronic steatohepatitis with early chicken-wire fibrosis was associated with enlargement of mitochondria and disruption of ER structure by electron microscopy, elevated indices of lipid storage, lipid peroxidation and DNA damage, increased activation of pro-inflammatory cytokines, impaired signaling through the insulin receptor (InR), InR substrate-1, Akt, ribosomal protein S6 kinase and proline-rich Akt substrate 40 kDa, glycogen synthase kinase 3β activation and constitutive up-regulation of ceramide and ER stress-related genes. Liquid chromatography coupled with tandem mass spectrometry demonstrated altered ceramide profiles with higher levels of C14 and C18, and reduced C16 species in ethanol-exposed livers. CONCLUSION The histopathologic and ultrastructural abnormalities in chronic alcohol-related steatohepatitis are associated with persistent hepatic insulin resistance and pro-inflammatory cytokine activation, dysregulated lipid metabolism with altered ceramide profiles and both ER and oxidative stress. Corresponding increases in lipid peroxidation, DNA damage and protein carbonylation may have contributed to the chronicity and progression of disease. The findings herein suggest that multi-pronged therapeutic strategies may be needed for effective treatment of chronic alcoholic liver disease in humans.
Collapse
Affiliation(s)
- Teresa Ramirez
- Liver Research Center, Divisions of Gastroenterology and Neuropathology, Departments of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Claverick Street, Providence, RI 02903, USA
| | | | | | | | | | | |
Collapse
|
30
|
Sharda DR, Miller-Lee JL, Kanski GM, Hunter JC, Lang CH, Kennett MJ, Korzick DH. Comparison of the agar block and Lieber-DeCarli diets to study chronic alcohol consumption in an aging model of Fischer 344 female rats. J Pharmacol Toxicol Methods 2012; 66:257-63. [PMID: 22951285 DOI: 10.1016/j.vascn.2012.08.166] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Post-menopausal women have a greater risk of developing alcoholic complications compared to age-matched men. Unfortunately, animal models of chronic ethanol consumption with estrogen deficiency are lacking. Here, we characterize the ability of the agar block and Lieber-DeCarli models of chronic ethanol consumption to produce elevated blood alcohol content (BAC) and liver pathology in the F344 postmenopausal animal model of aging. METHODS Adult (3 mo) and aged (18 mo) F344 ovary-intact or ovariectomized rats were administered ethanol for 14-20 weeks as follows: diet 1, standard chow access, 10% ethanol in drinking water, and 40% ethanol in agar blocks; diet 2, diet 1 plus low phytoestrogen chow (known to affect ethanol metabolism) for the final 4 weeks; diet 3, Lieber-DeCarli all liquid diet with 36% kcal ethanol. Control animals were matched isocalorically with dextrin. RESULTS For the agar block diet, average BAC was 13±4 mg/dL across groups. BAC was unaffected by reducing dietary phytoestrogen content (12±4 mg/dL), which is known to interfere with ethanol metabolism. Liver pathology was unaffected by the agar block diet. In contrast, the Lieber-DeCarli diet resulted in BAC of 45±5 mg/dL in conjunction with more severe hepatopathology.223 DISCUSSION We conclude that the Lieber-DeCarli diet produces greater BAC and hepatopathology to study the effects of chronic ethanol administration in the F344 postmenopausal rodent model of aging when compared to an ethanol agar block diet.
Collapse
Affiliation(s)
- Daniel R Sharda
- Department of Kinesiology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Impaired insulin/IGF signaling in experimental alcohol-related myopathy. Nutrients 2012; 4:1058-75. [PMID: 23016132 PMCID: PMC3448087 DOI: 10.3390/nu4081058] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 08/07/2012] [Accepted: 08/13/2012] [Indexed: 12/13/2022] Open
Abstract
Alcohol-related myopathy (Alc-M) is highly prevalent among heavy drinkers, although its pathogenesis is not well understood. We hypothesize that Alc-M is mediated by combined effects of insulin/IGF resistance and oxidative stress, similar to the effects of ethanol on liver and brain. We tested this hypothesis using an established model in which adult rats were pair-fed for 8 weeks with isocaloric diets containing 0% (N = 8) or 35.5% (N = 13) ethanol by caloric content. Gastrocnemius muscles were examined by histology, morphometrics, qRT-PCR analysis, and ELISAs. Chronic ethanol feeding reduced myofiber size and mRNA expression of IGF-1 polypeptide, insulin, IGF-1, and IGF-2 receptors, IRS-1, and IRS-2. Multiplex ELISAs demonstrated ethanol-associated inhibition of insulin, IRS-1, Akt, and p70S6K signaling, and increased activation of GSK-3β. In addition, ethanol-exposed muscles had increased 4-hydroxy-2-nonenal immunoreactivity, reflecting lipid peroxidation, and reduced levels of mitochondrial Complex IV, Complex V, and acetylcholinesterase. These results demonstrate that experimental Alc-M is associated with inhibition of insulin/IGF/IRS and downstream signaling that mediates metabolism and cell survival, similar to findings in alcoholic liver and brain degeneration. Moreover, the increased oxidative stress, which could be mediated by mitochondrial dysfunction, may have led to inhibition of acetylcholinesterase, which itself is sufficient to cause myofiber atrophy and degeneration.
Collapse
|
32
|
Nguyen VA, Le T, Tong M, Mellion M, Gilchrist J, de la Monte SM. Experimental alcohol-related peripheral neuropathy: role of insulin/IGF resistance. Nutrients 2012; 4:1042-57. [PMID: 23016131 PMCID: PMC3448086 DOI: 10.3390/nu4081042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 07/30/2012] [Accepted: 08/02/2012] [Indexed: 01/01/2023] Open
Abstract
The mechanisms of alcohol-related peripheral neuropathy (ALPN) are poorly understood. We hypothesize that, like alcohol-related liver and brain degeneration, ALPN may be mediated by combined effects of insulin/IGF resistance and oxidative stress. Adult male Long Evans rats were chronically pair-fed with diets containing 0% or 37% ethanol (caloric), and subjected to nerve conduction studies. Chronic ethanol feeding slowed nerve conduction in the tibial (p = 0.0021) motor nerve, and not plantar sensory nerve, but it did not affect amplitude. Histological studies of the sciatic nerve revealed reduced nerve fiber diameters with increased regenerative sprouts, and denervation myopathy in ethanol-fed rats. qRT-PCR analysis demonstrated reduced mRNA levels of insulin, IGF-1, and IGF-2 polypeptides, IGF-1 receptor, and IRS2, and ELISAs revealed reduced immunoreactivity for insulin and IGF-1 receptors, IRS-1, IRS-4, myelin-associated glycoprotein, and tau in sciatic nerves of ethanol-fed rats (all p < 0.05 or better). The findings suggest that ALPN is characterized by (1) slowed conduction velocity with demyelination, and a small component of axonal degeneration; (2) impaired trophic factor signaling due to insulin and IGF resistance; and (3) degeneration of myelin and axonal cytoskeletal proteins. Therefore, ALPN is likely mediated by molecular and signal transduction abnormalities similar to those identified in alcoholic liver and brain degeneration.
Collapse
Affiliation(s)
- Van Anh Nguyen
- Department of Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, 55 Claverick Street, Providence RI 02903, USA; (V.A.N.); (T.L.); (M.T.)
| | - Tran Le
- Department of Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, 55 Claverick Street, Providence RI 02903, USA; (V.A.N.); (T.L.); (M.T.)
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, 55 Claverick Street, Providence RI 02903, USA; (V.A.N.); (T.L.); (M.T.)
| | - Michelle Mellion
- Department of Neurology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence RI 02903, USA; (M.M.); (J.G.)
| | - James Gilchrist
- Department of Neurology, Rhode Island Hospital, Alpert Medical School of Brown University, 593 Eddy Street, Providence RI 02903, USA; (M.M.); (J.G.)
| | - Suzanne M. de la Monte
- Departments of Neuropathology/Pathology, Neurology, Neurosurgery, and Medicine, Rhode Island Hospital, Alpert Medical School of Brown University, 55 Claverick Street, Providence RI 02903, USA
| |
Collapse
|
33
|
Insulin resistance, ceramide accumulation, and endoplasmic reticulum stress in human chronic alcohol-related liver disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2012; 2012:479348. [PMID: 22577490 PMCID: PMC3347750 DOI: 10.1155/2012/479348] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 12/28/2011] [Accepted: 01/10/2012] [Indexed: 12/14/2022]
Abstract
Background. Chronic alcohol-related liver disease (ALD) is mediated by insulin resistance, mitochondrial dysfunction, inflammation, oxidative stress, and DNA damage. Recent studies suggest that dysregulated lipid metabolism with accumulation of ceramides, together with ER stress potentiate hepatic insulin resistance and may cause steatohepatitis to progress. Objective. We examined the degree to which hepatic insulin resistance in advanced human ALD is correlated with ER stress, dysregulated lipid metabolism, and ceramide accumulation. Methods. We assessed the integrity of insulin signaling through the Akt pathway and measured proceramide and ER stress gene expression, ER stress signaling proteins, and ceramide profiles in liver tissue. Results. Chronic ALD was associated with increased expression of insulin, IGF-1, and IGF-2 receptors, impaired signaling through IGF-1R and IRS1, increased expression of multiple proceramide and ER stress genes and proteins, and higher levels of the C14, C16, C18, and C20 ceramide species relative to control. Conclusions. In human chronic ALD, persistent hepatic insulin resistance is associated with dysregulated lipid metabolism, ceramide accumulation, and striking upregulation of multiple ER stress signaling molecules. Given the role of ceramides as mediators of ER stress and insulin resistance, treatment with ceramide enzyme inhibitors may help reverse or halt progression of chronic ALD.
Collapse
|
34
|
Liangpunsakul S, Rahmini Y, Ross RA, Zhao Z, Xu Y, Crabb DW. Imipramine blocks ethanol-induced ASMase activation, ceramide generation, and PP2A activation, and ameliorates hepatic steatosis in ethanol-fed mice. Am J Physiol Gastrointest Liver Physiol 2012; 302:G515-23. [PMID: 22194417 PMCID: PMC3311438 DOI: 10.1152/ajpgi.00455.2011] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 12/18/2011] [Indexed: 01/31/2023]
Abstract
Our previous data showed the inhibitory effect of ethanol on AMP-activated protein kinase phosphorylation, which appears to be mediated, in part, through increased levels of hepatic ceramide and activation of protein phosphatase 2A (Liangpunsakul S, Sozio MS, Shin E, Zhao Z, Xu Y, Ross RA, Zeng Y, Crabb DW. Am J Physiol Gastrointest Liver Physiol 298: G1004-G1012, 2010). The effect of ethanol on AMP-activated protein kinase phosphorylation was reversed by imipramine, suggesting that the generation of ceramide via acid sphingomyelinase (ASMase) is stimulated by ethanol. In this study, we determined the effects of imipramine on the development of hepatic steatosis, the generation of ceramide, and downstream effects of ceramide on inflammatory, insulin, and apoptotic signaling pathways, in ethanol-fed mice. The effect of ethanol and imipramine (10 μg/g body wt ip) on ceramide levels, as well as inflammatory, insulin, and apoptotic signaling pathways, was studied in C57BL/6J mice fed the Lieber-DeCarli diet. Ethanol-fed mice developed the expected steatosis, and cotreatment with imipramine for the last 2 wk of ethanol feeding resulted in improvement in hepatic steatosis. Ethanol feeding for 4 wk induced impaired glucose tolerance compared with controls, and this was modestly improved with imipramine treatment. There was a significant decrease in total ceramide concentrations in response to imipramine in ethanol-fed mice treated with and without imipramine (287 ± 11 vs. 348 ± 12 pmol/mg tissue). The magnitude and specificity of inhibition on each ceramide species differed. A significant decrease was observed for C16 (28 ± 3 vs. 33 ± 2 pmol/mg tissue) and C24 (164 ± 9 vs. 201 ± 4 pmol/mg tissue) ceramide. Ethanol feeding increased the levels of the phosphorylated forms of ERK slightly and increased phospho-p38 and phospho-JNK substantially. The levels of phospho-p38 and phospho-JNK were reduced by treatment with imipramine. The activation of ASMase and generation of ceramide in response to ethanol feeding may underlie several effects of ethanol. ASMase inhibitors may be considered as a therapeutic target for alcohol-induced hepatic steatosis and activation of stress kinases.
Collapse
Affiliation(s)
- Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Ramirez T, Tong M, Ayala CA, Monfils PR, McMillan PN, Zabala V, Wands JR, de la Monte SM. Structural Correlates of PPAR Agonist Rescue of Experimental Chronic Alcohol-Induced Steatohepatitis. ACTA ACUST UNITED AC 2012; 2. [PMID: 26339530 PMCID: PMC4554760 DOI: 10.4172/2161-0681.1000114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Chronic alcoholic liver disease is associated with hepatic insulin resistance, inflammation, oxidative and ER stress, mitochondrial dysfunction, and DNA damage. Peroxisome-proliferator activated receptor (PPAR) agonists are insulin sensitizers that have anti-inflammatory/anti-oxidant effects. We previously showed that PPAR agonists can restore hepatic insulin responsiveness in chronic ethanol-fed rats with steatohepatitis. Herein, we furthered our investigations by characterizing the histological and ultrastructural changes mediated by PPAR agonist rescue of alcohol-induced steatohepatitis. Adult male Long Evans rats were pair fed with isocaloric liquid diets containing 0% or 37% ethanol (caloric) for 8 weeks. After 3 weeks on the diets, rats were treated with vehicle, or a PPAR-α, PPAR-δ, or PPAR-γ agonist twice weekly by i.p. injection. Ethanol-fed rats developed steatohepatitis with disordered hepatic chord architecture, mega-mitochondria, disruption of the RER, increased apoptosis, and increased 4-hydroxynonenal (HNE) and 3-nitrotyrosine (NTyr) immunoreactivity. PPAR-δ and PPAR-γ agonists reduced the severity of steatohepatitis, and restored the hepatic chord-like architectural, mitochondrial morphology, and RER organization, and the PPAR-δ agonist significantly reduced hepatic HNE. On the other hand, prominent RER tubule dilation, which could reflect ER stress, persisted in ethanol-exposed, PPAR-γ treated but not PPAR-δ treated livers. The PPAR-α agonist exacerbated both steatohepatitis and formation of mega-mitochondria, and it failed to restore RER architecture or lower biochemical indices of oxidative stress. In conclusion, improved hepatic insulin responsiveness and decreased inflammation resulting from PPAR-δ or PPAR-γ agonist treatments of alcohol-induced steatohepatitis are likely mediated by enhanced signaling through metabolic pathways with attendant reductions in ER stress, oxidative stress, and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Teresa Ramirez
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Carol A Ayala
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Paul R Monfils
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Paul N McMillan
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Valerie Zabala
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jack R Wands
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Suzanne M de la Monte
- Liver Research Center and Departments of Medicine, Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
36
|
Abstract
BACKGROUND/AIMS We have compared dendritic cell (DC) function derived from the alcoholic liver disease (ALD) sensitive Long-Evans (LE) and resistant Fischer rat strains to determine if the influence of ethanol on DCs was dependent on ALD. METHODS The LE and Fischer rats were fed an ethanol-containing or isocaloric control liquid diet for 8 weeks and comparisons were made to LE rats injected with thioacetamide as a liver disease control. DCs were isolated from the spleen after expansion with human Fms-like tyrosine kinase receptor 3 ligand plasmid. Maturation markers CD86, CD80, CD40 and MHC-II were analysed by flow cytometry with or without lipopolysaccharide and poly I:C stimulation. Production of tumour necrosis factor-α (TNF-α), interferon-γ (IFN-γ), interleukin (IL)-12p40 and IL-10 cytokines and the antigen presentation ability of DCs was determined. RESULTS Only LE rats developed ALD characterized by liver injury, elevated alanine aminotransferase levels and steatosis; CD86 and CD40 expression was decreased in LE but not Fischer rats. Reduced TNF-α, IFN-γ, IL-12, proinflammatory and enhanced IL-10 cytokine production was found in DCs isolated from ethanol-fed LE but not Fischer rats. Allostimulatory activity was reduced in LE compared with the Fischer strain. In contrast, DCs isolated from thioacetamide-induced liver damage displayed a reduction only in IL-12p40; TNF-α, IL-10 and IFN-α production as well as antigen presenting ability remained intact compared with controls. CONCLUSIONS ALD sensitive LE rats exhibited characteristics of a suppressed DC phenotype that was not observed following thioacetamide-induced liver disease, which suggests an important role for ALD in altering the host cellular and humoral immune responses.
Collapse
Affiliation(s)
- Dechun Feng
- The Department of Medicine, Liver Research Center, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | | | | |
Collapse
|
37
|
de la Monte SM, Pang M, Chaudhry R, Duan K, Longato L, Carter J, Ouh J, Wands JR. Peroxisome proliferator-activated receptor agonist treatment of alcohol-induced hepatic insulin resistance. Hepatol Res 2011; 41:386-98. [PMID: 21426453 PMCID: PMC3399907 DOI: 10.1111/j.1872-034x.2011.00775.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Chronic ethanol exposure impairs insulin signaling in the liver. Peroxisome-proliferator activated receptor (PPAR) agonists function as insulin sensitizers and are used to treat type 2 diabetes mellitus. We examined the therapeutic effectiveness of PPAR agonists in reducing alcoholic hepatitis and hepatic insulin resistance in a model of chronic ethanol feeding. METHODS Adult male Long Evans rats were pair fed with isocaloric liquid diets containing 0% (control) or 37% ethanol (caloric content; 9.2% v/v) for 8 weeks. After 3 weeks on the diets, the rats were treated with vehicle, or a PPAR-α, PPAR-δ or PPAR-γ agonist twice weekly by i.p. injection. Livers were harvested for histopathological, gene expression (reverse transcription polymerase chain reaction), protein (western and ELISA) and receptor binding studies. RESULTS Ethanol-fed rats developed steatohepatitis with disordered hepatic chord architecture, increased hepatocellular apoptosis, reduced binding to the insulin, insulin-like growth factor (IGF)-1 and IGF-2 receptors, and decreased expression of glyceraldehyde-3-phosphate dehydrogenase and aspartyl-(asparaginyl)-β-hydroxylase (mediating remodeling), which are regulated by insulin/IGF signaling. PPAR-α, PPAR-δ or PPAR-γ agonist treatments reduced the severity of ethanol-mediated liver injury, including hepatic architectural disarray and steatosis. In addition, PPAR-δ and PPAR-γ agonists reduced insulin/IGF resistance and increased insulin/IGF-responsive gene expression. CONCLUSION PPAR agonists may help reduce the severity of chronic ethanol-induced liver injury and insulin/IGF resistance, even in the context of continued high-level ethanol consumption.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Liver Research Center and Departments of Medicine and Pathology, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | | | | | | | | | | | | | | |
Collapse
|