1
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [PMID: 37482263 DOI: 10.1016/j.jep.2023.116933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/15/2023] [Accepted: 07/17/2023] [Indexed: 07/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellaria-coptis herb couple (SC) is a classic herbal pair used in many Traditional Chinese Medicine (TCM) formulations in the treatment of endocrine and metabolic deseases. Diabetes mellitus and non-alcoholic steatohepatitis (NASH) are both endocrine and metabolic diseases. Previous studies have shown that SC has anti-diabetic effects. However, the effect and mechanism of SC against NASH remains unclear. AIM OF THE STUDY This study aimed to demonstrate the effect and mechanism of SC against NASH through the nuclear factor-erythroid 2-related factor 2 (Nrf2) and farnesoid X receptor (FXR) dual signaling pathways in vivo and in vitro. MATERIALS AND METHODS The high fat diet-fed rat model, and HepG2 and RAW264.7 cell models were used. Serum biochemical indexes and liver histopathological changes were examined. Metabolomics, transcriptomics, and flow cytometry were performed. RT-qPCR and western blot analysis were performed to provide expression of NRF2 and FXR pathway signal molecules during SC's anti-NASH treatment in vivo and in vitro. RESULTS SC had anti-NASH effects in vivo with significantly improvement of serum NASH biochemical index and hepatopathological structure; meanwhile, SC significantly elevated the expression levels of FXR protein in liver and intestinal tissues, and cholesterol 7a-hydroxylase (CYP7A1) protein in liver. The mRNA expression levels of Takeda G protein receptor 5 (TGR5), CYP7A1, fibroblast growth factor receptor-4 (FGFR4), FXR, small heterodimer partner (SHP), fibroblast growth factor 15/19 (FGF15/19) and glucagon-like peptide-1 (GLP-1) were significantly elevated by SC. SC reduced the levels of NorCA, isoLCA and α-MCA in the feces of NAFLD rats. In vitro, SC-containing serum (SC-CS) was found to significantly reduce intracellular lipid deposition, inhibit ROS production, reduce intracellular Malondialdehyde (MDA) and IL-1β levels, and enhance the activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px). Six differential genes closely related to oxidative stress and Nrf2 were identified by transcriptomic analysis. SC-CS up-regulated the expression of NRF2, and reduced the expression of TXNIP and Caspase-1 genes in RAW264.7 cells. In addition, SC-CS reduced the expression of Keap1 and NF-κB, and up-regulated the expression of Nrf2, heme oxygenase-1 (HO-1), quinone oxidoreductase 1 (NQO1), and SOD; SC-CS elevated the protein level of NRF2, and reduced the protein level of TXNIP in HepG2 cells. CONCLUSIONS the mechanisms of SC action against NASH was closely related to the simultaneous activations of both NRF2 and FXR signaling pathways. These findings provide a new insight into the anti-NASH application of SC in clinical settings and demonstrate the potential of SC in the treatment of NASH.
Collapse
Affiliation(s)
- Yanan Xue
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Yue Wei
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Lan Cao
- Research Center of Chinese Medicine Resources and Ethnic Medicine, Jiangxi University of Chinese Medicine, PR China
| | - Min Shi
- College of Life Science, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Junqing Sheng
- College of Life Science, Nanchang University, Nanchang, 330031, PR China
| | - Qin Xiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ziwen Cheng
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Tao Luo
- First Affiliated Hospital of Nanchang University, 330006, PR China
| | - Quanhui Jiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Ailan Wu
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Lingyun Zhong
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China.
| | - Changhua Zhang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, PR China; Nanchang Research Institute, Sun Yat-sen University, Jiangxi, 330096, PR China.
| |
Collapse
|
2
|
Xue Y, Wei Y, Cao L, Shi M, Sheng J, Xiao Q, Cheng Z, Luo T, Jiao Q, Wu A, Chen C, Zhong L, Zhang C. Protective effects of scutellaria-coptis herb couple against non-alcoholic steatohepatitis via activating NRF2 and FXR pathways in vivo and in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116933. [DOI: https:/doi.org/10.1016/j.jep.2023.116933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2024]
|
3
|
Blas-García A, Apostolova N. Novel Therapeutic Approaches to Liver Fibrosis Based on Targeting Oxidative Stress. Antioxidants (Basel) 2023; 12:1567. [PMID: 37627562 PMCID: PMC10451738 DOI: 10.3390/antiox12081567] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Chronic liver disease (CLD) constitutes a growing global health issue, with no effective treatments currently available. Oxidative stress closely interacts with other cellular and molecular processes to trigger stress pathways in different hepatic cells and fuel the development of liver fibrosis. Therefore, inhibition of reactive oxygen species (ROS)-mediated effects and modulation of major antioxidant responses to counteract oxidative stress-induced damage have emerged as interesting targets to prevent or ameliorate liver injury. Although many preclinical studies have shown that dietary supplements with antioxidant properties can significantly prevent CLD progression in animal models, this strategy has not proved effective to significantly reduce fibrosis when translated into clinical trials. Novel and more specific therapeutic approaches are thus required to alleviate oxidative stress and reduce liver fibrosis. We have reviewed the relevant literature concerning the crucial role of alterations in redox homeostasis in different hepatic cell types during the progression of CLD and discussed current pharmacological approaches to ameliorate fibrosis by reducing oxidative stress focusing on selective modulation of enzymatic oxidant sources, antioxidant systems and ROS-mediated pathogenic processes.
Collapse
Affiliation(s)
- Ana Blas-García
- Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Nadezda Apostolova
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
- CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Instituto de Salud Carlos III, Monforte de Lemos, 3-5, 28029 Madrid, Spain
- Departamento de Farmacología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain
| |
Collapse
|
4
|
Willemse L, Terburgh K, Louw R. A ketogenic diet alters mTOR activity, systemic metabolism and potentially prevents collagen degradation associated with chronic alcohol consumption in mice. Metabolomics 2023; 19:43. [PMID: 37076659 PMCID: PMC10115735 DOI: 10.1007/s11306-023-02006-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023]
Abstract
INTRODUCTION A ketogenic diet (KD), which is a high fat, low carbohydrate diet has been shown to inhibit the mammalian target of rapamycin (mTOR) pathway and alter the redox state. Inhibition of the mTOR complex has been associated with the attenuation and alleviation of various metabolic and- inflammatory diseases such as neurodegeneration, diabetes, and metabolic syndrome. Various metabolic pathways and signalling mechanisms have been explored to assess the therapeutic potential of mTOR inhibition. However, chronic alcohol consumption has also been reported to alter mTOR activity, the cellular redox- and inflammatory state. Thus, a relevant question that remains is what effect chronic alcohol consumption would have on mTOR activity and overall metabolism during a KD-based intervention. OBJECTIVES The aim of this study was to evaluate the effect of alcohol and a KD on the phosphorylation of the mTORC1 target p70S6K, systemic metabolism as well as the redox- and inflammatory state in a mouse model. METHODS Mice were fed either a control diet with/without alcohol or a KD with/without alcohol for three weeks. After the dietary intervention, samples were collected and subjected towards western blot analysis, multi-platform metabolomics analysis and flow cytometry. RESULTS Mice fed a KD exhibited significant mTOR inhibition and reduction in growth rate. Alcohol consumption alone did not markedly alter mTOR activity or growth rate but moderately increased mTOR inhibition in mice fed a KD. In addition, metabolic profiling showed alteration of several metabolic pathways as well as the redox state following consumption of a KD and alcohol. A KD was also observed to potentially prevent bone loss and collagen degradation associated with chronic alcohol consumption, as indicated by hydroxyproline metabolism. CONCLUSION This study sheds light on the influence that a KD alongside alcohol intake can exert on not just mTOR, but also their effect on metabolic reprogramming and the redox state.
Collapse
Affiliation(s)
- Luciano Willemse
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Karin Terburgh
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa
| | - Roan Louw
- Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University (Potchefstroom Campus), Private Bag X6001, Potchefstroom, South Africa.
| |
Collapse
|
5
|
Jiang ZB, Gao J, Chai YH, Li W, Luo YF, Chen YZ. Astragaloside alleviates alcoholic fatty liver disease by suppressing oxidative stress. Kaohsiung J Med Sci 2021; 37:718-729. [PMID: 33973356 DOI: 10.1002/kjm2.12390] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/24/2021] [Accepted: 03/17/2021] [Indexed: 12/17/2022] Open
Abstract
Alcoholic fatty liver disease (AFLD) is the most common liver disease and can progress to fatal liver cirrhosis and carcinoma, affecting millions of patients worldwide. The functions of astragaloside on the cardiovascular system have been elucidated. However, its role in AFLD is unclear. Ethanol-treated AML-12 cells were used as a cell model of alcoholic fatty liver. Real-time quantitative reverse transcription-PCR and Western blotting detected genes and proteins expressions. Reactive oxygen species (ROS), triglyceride, total cholesterol, low-density lipoprotein, albumin, ferritin, bilirubin, superoxide dismutase, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) were examined using commercial kits. Lipid accumulation was assessed by Oil red O staining. MTT and flow cytometry measured cell viability and apoptosis. JC-1 was used to analyze mitochondrial membrane potential. A rat model of AFLD was established by treating rats with ethanol. Astragaloside suppressed ethanol-induced lipid accumulation, oxidative stress, and the production of AST and ALT in AML-12 cells. Ethanol induced TNF-α and reduced IL-10 expression, which were reversed by astragaloside. Ethanol promoted Bax expression and cytochrome C release and inhibited Bcl-2 and ATP expression. Astragaloside hampered these apoptosis effects in AML-12 cells. Impaired mitochondrial membrane potential was recovered by astragaloside. However, all these astragaloside-mediated beneficial effects were abolished by the ROS inducer pyocyanin. Ethanol-induced activation of NF-κB signaling was suppressed by astragaloside in vitro and in vivo, suggesting that astragaloside inhibited oxidative stress by suppressing the activation of NF-κB signaling, thus improving liver function and alleviating AFLD in rats. Our study elucidates the pharmacological mechanism of astragaloside and provides potential therapeutic strategies for AFLD.
Collapse
Affiliation(s)
- Zhi-Bin Jiang
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jie Gao
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yi-Hui Chai
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wen Li
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yun-Feng Luo
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yun-Zhi Chen
- Department of Preclinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
6
|
Angi A, Chiarelli F. Obesity and Diabetes: A Sword of Damocles for Future Generations. Biomedicines 2020; 8:E478. [PMID: 33171922 PMCID: PMC7694547 DOI: 10.3390/biomedicines8110478] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023] Open
Abstract
Childhood obesity is one of the most challenging problem of the 21st century. The prevalence has increased, reaching an alarming rate. Furthermore, the problem is global and is also affecting low- and middle-income countries. This global obesity epidemic explains how the roots of cardiovascular disease, the most common cause of mortality among adults, begin in childhood. Overweight and obese children are likely to stay obese into adulthood and to develop noncommunicable diseases such as diabetes and cardiovascular diseases at a younger age. Thus, prevention should be the major goal and should start early in life. The aim of this review is to present an updated framework of the current understanding of the cardiovascular and metabolic risks in obese children and adolescents and to discuss the available therapeutic options.
Collapse
Affiliation(s)
- Alessia Angi
- Department of Pediatrics, University of Chieti, 66100 Chieti, Italy;
| | | |
Collapse
|
7
|
Tavares TB, Santos IB, de Bem GF, Ognibene DT, da Rocha APM, de Moura RS, Resende ADC, Daleprane JB, da Costa CA. Therapeutic effects of açaí seed extract on hepatic steatosis in high-fat diet-induced obesity in male mice: a comparative effect with rosuvastatin. J Pharm Pharmacol 2020; 72:1921-1932. [PMID: 32856322 DOI: 10.1111/jphp.13356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/25/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Obesity is considered a risk factor for the development of non-alcoholic fatty liver disease (NAFLD). The hydroalcoholic extract obtained from the açai seed (ASE), rich in proanthocyanidins, has been shown a potential body weight regulator with antioxidant properties. This study aimed to investigate the therapeutic effect of ASE in obesity-associated NAFLD and compare it with Rosuvastatin. METHODS Male C57BL/6 mice received a high-fat diet or standard diet for 12 weeks. The treatments with ASE (300 mg/kg per day) or rosuvastatin (20 mg/kg per day) began in the eighth week until the 12th week. KEY FINDINGS Our data show that the treatments with ASE and rosuvastatin reduced body weight and hyperglycaemia, improved lipid profile and attenuated hepatic steatosis in HFD mice. ASE and Rosuvastatin reduced HMGCoA-Reductase and SREBP-1C and increased ABGC8 and pAMPK expressions in the liver. Additionally, ASE, but not Rosuvastatin, reduced NPC1L1 and increased ABCG5 and PPAR-α expressions. ASE and rosuvastatin increased SIRT-1 expression and antioxidant defence, although only ASE was able to decrease the oxidative damage in hepatic tissue. CONCLUSIONS The therapeutic effect of ASE was similar to that of rosuvastatin in reducing dyslipidemia and hepatic steatosis but was better in reducing oxidative damage and hyperglycaemia.
Collapse
Affiliation(s)
- Thamires Barros Tavares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Angela de Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Julio Beltrame Daleprane
- Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
8
|
Zhao P, Han SN, Arumugam S, Yousaf MN, Qin Y, Jiang JX, Torok NJ, Chen Y, Mankash MS, Liu J, Li J, Iwakiri Y, Ouyang X. Digoxin improves steatohepatitis with differential involvement of liver cell subsets in mice through inhibition of PKM2 transactivation. Am J Physiol Gastrointest Liver Physiol 2019; 317:G387-G397. [PMID: 31411894 PMCID: PMC6842989 DOI: 10.1152/ajpgi.00054.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The cardiac glycoside digoxin was identified as a potent suppressor of pyruvate kinase isoform 2-hypoxia-inducible factor-α (PKM2-HIF-1α) pathway activation in liver injury mouse models via intraperitoneal injection. We have assessed the therapeutic effects of digoxin to reduce nonalcoholic steatohepatitis (NASH) by the clinically relevant oral route in mice and analyzed the cellular basis for this effect with differential involvement of liver cell subsets. C57BL/6J male mice were placed on a high-fat diet (HFD) for 10 wk and started concurrently with the gavage of digoxin (2.5, 0.5, 0.125 mg/kg twice a week) for 5 wk. Digoxin significantly reduced HFD-induced hepatic damage, steatosis, and liver inflammation across a wide dosage range. The lowest dose of digoxin (0.125 mg/kg) showed significant protective effects against liver injury and sterile inflammation. Consistently, digoxin attenuated HIF-1α sustained NLRP3 inflammasome activation in macrophages. We have reported for the first time that PKM2 is upregulated in hepatocytes with hepatic steatosis, and digoxin directly improved hepatocyte mitochondrial dysfunction and steatosis. Mechanistically, digoxin directly bound to PKM2 and inhibited PKM2 targeting HIF-1α transactivation without affecting PKM2 enzyme activation. Thus, oral digoxin showed potential to therapeutically inhibit liver injury in NASH through the regulation of PKM2-HIF-1α pathway activation with involvement of multiple cell types. Because of the large clinical experience with oral digoxin, this may have significant clinical applicability in human NASH.NEW & NOTEWORTHY This study is the first to assess the therapeutic efficacy of oral digoxin on nonalcoholic steatohepatitis (NASH) in a high-fat diet (HFD) mouse model and to determine the divergent of cell type-specific effects. Oral digoxin reduced liver damage, steatosis, and inflammation in HFD mice. Digoxin attenuated hypoxia-inducible factor (HIF)-1α axis-sustained inflammasome activity in macrophages and hepatic oxidative stress response in hepatocytes via the regulation of PKM2-HIF-1α axis pathway activation. Oral digoxin may have significant clinical applicability in human NASH.
Collapse
Affiliation(s)
- Peng Zhao
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut,5Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment and Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Sheng-Na Han
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut,6Department of Pharmacology, Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Suyavaran Arumugam
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| | - Muhammad Nadeem Yousaf
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| | - Yanqin Qin
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut,5Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment and Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Joy X. Jiang
- 2Department of Internal Medicine, University of California, Davis, Sacramento, California
| | - Natalie Julia Torok
- 3Department of Gastroenterology and Hepatology, Stanford University and Veterans Affairs, Palo Alto, Stanford, California
| | - Yonglin Chen
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| | - Mohd Salah Mankash
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| | - Junbao Liu
- 4The People’s Hospital of Henan Province, Zhengzhou, China
| | - Jiansheng Li
- 5Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment and Chinese Medicine Development of Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Yasuko Iwakiri
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| | - Xinshou Ouyang
- 1Section of Digestive Diseases, Yale University School of medicine, New Haven, Connecticut
| |
Collapse
|
9
|
Periodontitis causes abnormalities in the liver of rats. J Periodontol 2018; 90:295-305. [DOI: 10.1002/jper.18-0226] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/14/2018] [Accepted: 06/20/2018] [Indexed: 12/16/2022]
|
10
|
Ouyang X, Han SN, Zhang JY, Dioletis E, Nemeth BT, Pacher P, Feng D, Bataller R, Cabezas J, Stärkel P, Caballeria J, Pongratz RL, Cai SY, Schnabl B, Hoque R, Chen Y, Yang WH, Garcia-Martinez I, Wang FS, Gao B, Torok NJ, Kibbey RG, Mehal WZ. Digoxin Suppresses Pyruvate Kinase M2-Promoted HIF-1α Transactivation in Steatohepatitis. Cell Metab 2018; 27:339-350.e3. [PMID: 29414684 PMCID: PMC5806149 DOI: 10.1016/j.cmet.2018.01.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 03/07/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022]
Abstract
Sterile inflammation after tissue damage is a ubiquitous response, yet it has the highest amplitude in the liver. This has major clinical consequences, for alcoholic and non-alcoholic steatohepatitis (ASH and NASH) account for the majority of liver disease in industrialized countries and both lack therapy. Requirements for sustained sterile inflammation include increased oxidative stress and activation of the HIF-1α signaling pathway. We demonstrate the ability of digoxin, a cardiac glycoside, to protect from liver inflammation and damage in ASH and NASH. Digoxin was effective in maintaining cellular redox homeostasis and suppressing HIF-1α pathway activation. A proteomic screen revealed that digoxin binds pyruvate kinase M2 (PKM2), and independently of PKM2 kinase activity results in chromatin remodeling and downregulation of HIF-1α transactivation. These data identify PKM2 as a mediator and therapeutic target for regulating liver sterile inflammation, and demonstrate a novel role for digoxin that can effectively protect the liver from ASH and NASH.
Collapse
Affiliation(s)
- Xinshou Ouyang
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA.
| | - Sheng-Na Han
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Ji-Yuan Zhang
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Evangelos Dioletis
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Balazs Tamas Nemeth
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIAAA/NIH, Bethesda, MD 20892, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, NIAAA/NIH, Bethesda, MD 20892, USA
| | - Dechun Feng
- NIAAA, NIH, 5625 Fishers Lane, Bethesda, MD 20892, USA
| | - Ramon Bataller
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Joaquin Cabezas
- Division of Gastroenterology and Hepatology, Department of Medicine, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Peter Stärkel
- Department of Gastroenterology, Saint-Luc Academic Hospital and Institute of Clinical Research, Catholic University of Louvain, Brussels, Belgium
| | - Joan Caballeria
- Unidad de Hepatología, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | | | - Shi-Ying Cai
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rafaz Hoque
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Yonglin Chen
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | - Wei-Hong Yang
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA
| | | | - Fu-Sheng Wang
- Institute of Translational Hepatology, Beijing 302 Hospital, Beijing 100039, China
| | - Bin Gao
- NIAAA, NIH, 5625 Fishers Lane, Bethesda, MD 20892, USA
| | - Natalie Julia Torok
- Department of Medicine, Gastroenterology, and Hepatology, University of California, Davis, Sacramento, CA, USA
| | | | - Wajahat Zafar Mehal
- Section of Digestive Diseases, Yale University, New Haven, CT 06520, USA; West Haven Veterans Medical Center, West Haven, CT 06516, USA.
| |
Collapse
|
11
|
Zhang X, Ji R, Sun H, Peng J, Ma X, Wang C, Fu Y, Bao L, Jin Y. Scutellarin ameliorates nonalcoholic fatty liver disease through the PPARγ/PGC-1α-Nrf2 pathway. Free Radic Res 2018; 52:198-211. [PMID: 29400110 DOI: 10.1080/10715762.2017.1422602] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/26/2017] [Accepted: 12/26/2017] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterised by excessive accumulation of hepatic lipids and oxidative injury of hepatocytes. Scutellarin is a flavonoid glycoside having antioxidative stress activity. Our current study aims to investigate the molecular mechanism of scutellarin ameliorating NAFLD. Scutellarin treatment was applied to male C57BL/6 mice maintained on a high-fat diet (HFD) and HepG2 cells challenged with oleic acid. The antioxidation biochemical indicators and lipid levels in the liver and cells were detected by kits. Liver pathology was observed by light microscope, Oil Red O staining, and transmission electron microscope (TEM). In addition, quantitative real-time polymerase chain reactions (qRT-PCR) and western blot assays were employed to detect the mRNA and protein levels of various antioxidative-related genes in the presence or absence of peroxisome proliferator-activated receptor gamma (PPARγ); inhibitor GW9662. Our results showed that scutellarin could significantly reduce blood lipid levels and enhance antioxidative capacities in both the models. In addition, scutellarin treatment conspicuously activated PPARγ, peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), nuclear factor erythroid-2-related factor (Nrf2), haem oxygenase-1 (HO-1), glutathione S-transferase (GST), and NAD(P)H quinone dehydrogenase one (NQO1), while it significantly inhibited nuclear factor kappa B (NF-κB), Kelch-like ECH-associated protein 1 (Keap1) at both the mRNA and protein levels. However, after interfered by GW9662, scutellarin effect was significantly decreased. The experimental data demonstrated that scutellarin showed strong hypolipidaemic, antioxidative, and liver protective activity which could be attributed to its regulating activity in the PPARγ/PGC-1α-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Renpeng Ji
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Huijun Sun
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Jinyong Peng
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Xiaodong Ma
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - ChangYuan Wang
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yufeng Fu
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Liuchi Bao
- a College of Pharmacy , Dalian Medical University , Dalian , China
| | - Yue Jin
- a College of Pharmacy , Dalian Medical University , Dalian , China
| |
Collapse
|
12
|
Mendes IKS, Matsuura C, Aguila MB, Daleprane JB, Martins MA, Mury WV, Brunini TMC. Weight loss enhances hepatic antioxidant status in a NAFLD model induced by high-fat diet. Appl Physiol Nutr Metab 2018; 43:23-29. [PMID: 28834687 DOI: 10.1139/apnm-2017-0317] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a benign condition that can progress to more severe liver damage in a process mediated, in part, by disturbances in redox balance. Additionally, some argue that it is set to become the main cause of end-stage liver disease in the near future. Here, we investigated whether diet-induced weight loss is able to reverse hepatic lipid accumulation and reduce oxidative stress in liver from C57BL/6 mice fed a high-fat (HF) diet. Male C57BL/6 mice were divided into 4 groups: standard chow (SC; 10% energy from fat, 16 weeks); HF (50% energy from fat, 16 weeks); SC-HF (SC for 8 weeks followed by HF for 8 weeks); and HF-SC (HF for 8 weeks followed by SC for 8 weeks). The HF diet during 8 (SC-HF) and 16 weeks (HF) downregulated messenger RNA levels and protein expression of Nrf2 and endogenous antioxidant enzymes (superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase) in the liver; caused liver steatosis; affected liver function markers; increased intra-abdominal and subcutaneous adipose tissue; and induced glucose intolerance and hypercholesterolemia compared with controls (SC). Diet-induced weight loss significantly reduced the intrahepatic lipid accumulation, improved glucose tolerance, and restored both gene and protein expression of the antioxidant enzymes. Our findings suggest that a dietary intervention aimed to induce weight loss may exert protective effects in NAFLD as it can reduce hepatic oxidative stress and intrahepatic lipid accumulation, which can hinder the progression of this condition to more severe states.
Collapse
Affiliation(s)
- Iara Karise Santos Mendes
- a Department of Pharmacology and Psychobiology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Cristiane Matsuura
- a Department of Pharmacology and Psychobiology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Marcia Barbosa Aguila
- b Laboratory of Morphometry and Cardiovascular Morphology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Julio Beltrame Daleprane
- c Laboratory for Studies on Interactions between Nutrition and Genetics, Department of Basic and Experimental Nutrition, University of the State of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil
| | - Marcela Anjos Martins
- a Department of Pharmacology and Psychobiology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Wanda Vianna Mury
- a Department of Pharmacology and Psychobiology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Tatiana Marlowe Cunha Brunini
- a Department of Pharmacology and Psychobiology, University of the State of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
| |
Collapse
|
13
|
Sánchez-Ramos C, Prieto I, Tierrez A, Laso J, Valdecantos MP, Bartrons R, Roselló-Catafau J, Monsalve M. PGC-1α Downregulation in Steatotic Liver Enhances Ischemia-Reperfusion Injury and Impairs Ischemic Preconditioning. Antioxid Redox Signal 2017; 27:1332-1346. [PMID: 28269997 DOI: 10.1089/ars.2016.6836] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Liver steatosis is associated with mitochondrial dysfunction and elevated reactive oxygen species (ROS) levels together with enhanced sensitivity to ischemia-reperfusion (IR) injury and limited response to preconditioning protocols. Here, we sought to determine whether the downregulation in the steatotic liver of peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α), a master regulator of mitochondrial metabolism and ROS that is known to play a role in liver metabolic control, could be responsible for the sensitivity of the steatotic liver to ischemic damage. RESULTS PGC-1α was induced in normal liver after exposure to an IR protocol, which was concomitant with an increase in the levels of antioxidant proteins. By contrast, its induction was severely blunted in the steatotic liver, resulting in a modest induction of antioxidant proteins. Livers of PGC-1α-/- mice on a chow diet were normal, but they exhibited an enhanced sensitivity to IR injury and also a lack of response to ischemic preconditioning (IPC), a phenotype that recapitulated the features of the steatotic liver in terms of liver damage, although the inflammatory response differed between both models. Utilizing an in vitro model of IPC, we found that PGC-1α expression was downregulated in hepatic cells cultured at 1% O2; whereas it was induced after reoxygenation (3% O2), and it was responsible for the recovery of antioxidant gene expression after the ischemic period. Innovation & Conclusion: PGC-1α plays an important role in the protection against IR injury in the liver, which is likely associated with its capacity to induce antioxidant gene expression. Antioxid. Redox Signal. 27, 1332-1346.
Collapse
Affiliation(s)
| | - Ignacio Prieto
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid, Spain
| | - Alberto Tierrez
- 2 Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid, Spain
| | - Javier Laso
- 2 Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) , Madrid, Spain
| | - M Pilar Valdecantos
- 3 Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem) , ISCIII, Madrid, Spain
| | - Ramon Bartrons
- 4 Unitat de Bioquímica i Biologia Molecular, Departament de Ciències Fisiològiques, Campus de Bellvitge, IDIBELL-Universitat de Barcelona , Hospitalet, Spain
| | - Joan Roselló-Catafau
- 5 Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (CSIC) , Barcelona, Spain
| | - María Monsalve
- 1 Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid, Spain
| |
Collapse
|
14
|
Ding RB, Bao J, Deng CX. Emerging roles of SIRT1 in fatty liver diseases. Int J Biol Sci 2017; 13:852-867. [PMID: 28808418 PMCID: PMC5555103 DOI: 10.7150/ijbs.19370] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/19/2017] [Indexed: 12/11/2022] Open
Abstract
Fatty liver diseases, which are commonly associated with high-fat/calorie diet, heavy alcohol consumption and/or other metabolic disorder causes, lead to serious medical concerns worldwide in recent years. It has been demonstrated that metabolic homeostasis disruption is most likely to be responsible for this global epidemic. Sirtuins are a group of conserved nicotinamide adenine dinucleotide (NAD+) dependent histone and/or protein deacetylases belonging to the silent information regulator 2 (Sir2) family. Among seven mammalian sirtuins, sirtuin 1 (SIRT 1) is the most extensively studied one and is involved in both alcoholic and nonalcoholic fatty liver diseases. SIRT1 plays beneficial roles in regulating hepatic lipid metabolism, controlling hepatic oxidative stress and mediating hepatic inflammation through deacetylating some transcriptional regulators against the progression of fatty liver diseases. Here we summarize the latest advances of the biological roles of SIRT1 in regulating lipid metabolism, oxidative stress and inflammation in the liver, and discuss the potential of SIRT1 as a therapeutic target for treating alcoholic and nonalcoholic fatty liver diseases.
Collapse
Affiliation(s)
- Ren-Bo Ding
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Jiaolin Bao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Chu-Xia Deng
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| |
Collapse
|
15
|
Flores-Toro JA, Go KL, Leeuwenburgh C, Kim JS. Autophagy in the liver: cell's cannibalism and beyond. Arch Pharm Res 2016; 39:1050-61. [PMID: 27515049 DOI: 10.1007/s12272-016-0807-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
Chronic liver disease and its progression to liver failure are induced by various etiologies including viral infection, alcoholic and nonalcoholic hepatosteatosis. It is anticipated that the prevalence of fatty liver disease will continue to rise due to the growing incidence of obesity and metabolic disorder. Evidence is accumulating to indicate that the onset of fatty liver disease is causatively linked to mitochondrial dysfunction and abnormal lipid accumulation. Current treatment options for this disease are limited. Autophagy is an integral catabolic pathway that maintains cellular homeostasis both selectively and nonselectively. As mitophagy and lipophagy selectively remove dysfunctional mitochondria and excess lipids, respectively, stimulation of autophagy could have therapeutic potential to ameliorate liver function in steatotic patients. This review highlights our up-to-date knowledge on mechanistic roles of autophagy in the pathogenesis of fatty liver disease and its vulnerability to surgical stress, with an emphasis on mitophagy and lipophagy.
Collapse
Affiliation(s)
- Joseph A Flores-Toro
- Department of Surgery, University of Florida, R4-204 ARB, 1600 SW Archer Rd, Gainesville, FL, 32610, USA
| | - Kristina L Go
- Department of Surgery, University of Florida, R4-204 ARB, 1600 SW Archer Rd, Gainesville, FL, 32610, USA
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, 32610, USA
- Institute on Aging, University of Florida, Gainesville, FL, 32610, USA
| | - Jae-Sung Kim
- Department of Surgery, University of Florida, R4-204 ARB, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
- Institute on Aging, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
16
|
S-Propargyl-cysteine Exerts a Novel Protective Effect on Methionine and Choline Deficient Diet-Induced Fatty Liver via Akt/Nrf2/HO-1 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4690857. [PMID: 27313828 PMCID: PMC4893438 DOI: 10.1155/2016/4690857] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/23/2016] [Accepted: 05/04/2016] [Indexed: 01/06/2023]
Abstract
This study investigated the antioxidative effect of S-propargyl-cysteine (SPRC) on nonalcoholic fatty liver (NAFLD) by treating mice fed a methionine and choline deficient (MCD) diet with SPRC for four weeks. We found that SPRC significantly reduced hepatic reactive oxygen species (ROS) and methane dicarboxylic aldehyde (MDA) levels. Moreover, SPRC also increased the superoxide dismutase (SOD) activity. By Western blot, we found that this protective effect of SPRC was importantly attributed to the regulated hepatic antioxidant-related proteins, including protein kinase B (Akt), heme oxygenase-1 (HO-1), nuclear factor erythroid 2-related factor 2 (Nrf2), and cystathionine γ-lyase (CSE, an enzyme that synthesizes hydrogen sulfide). Next, we examined the detailed molecular mechanism of the SPRC protective effect using oleic acid- (OA-) induced HepG2 cells. The results showed that SPRC significantly decreased intracellular ROS and MDA levels in OA-induced HepG2 cells by upregulating the phosphorylation of Akt, the expression of HO-1 and CSE, and the translocation of Nrf2. SPRC-induced HO-1 expression and Nrf2 translocation were abolished by the phosphoinositide 3-kinase (PI3K) inhibitor LY294002. Moreover, the antioxidative effect of SPRC was abolished by CSE inhibitor DL-propargylglycine (PAG) and HO-1 siRNA. Therefore, these results proved that SPRC produced an antioxidative effect on NAFLD through the PI3K/Akt/Nrf2/HO-1 signaling pathway.
Collapse
|
17
|
Zhu J, Ren T, Zhou M, Cheng M. The combination of blueberry juice and probiotics reduces apoptosis of alcoholic fatty liver of mice by affecting SIRT1 pathway. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1649-61. [PMID: 27274198 PMCID: PMC4869661 DOI: 10.2147/dddt.s102883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Purpose To explore the effects of the combination of blueberry juice and probiotics on the apoptosis of alcoholic fatty liver disease (AFLD). Methods Healthy C57BL/6J mice were used in the control group (CG). AFLD mice models were established with Lieber–DeCarli ethanol diet and evenly assigned to six groups with different treatments: MG (model), SI (SIRT1 [sirtuin type 1] small interfering RNA [siRNA]), BJ (blueberry juice), BJSI (blueberry juice and SIRT1 siRNA), BJP (blueberry juice and probiotics), and BJPSI (blueberry juice, probiotics, and SIRT1 siRNA). Hepatic tissue was observed using hematoxylin and eosin (HE) and Oil Red O (ORO) staining. Biochemical indexes of the blood serum were analyzed. The levels of SIRT1, caspase-3, forkhead box protein O1 (FOXO1), FasL (tumor necrosis factor ligand superfamily member 6), BAX, and Bcl-2 were measured by reverse transcription-polymerase chain reaction and Western blotting. Results HE and ORO staining showed that the hepatocytes were heavily destroyed with large lipid droplets in MG and SI groups, while the severity was reduced in the CG, BJ, and BJP groups (P<0.05). The levels of superoxide dismutase (SOD), reduced glutathione (GSH), and high-density lipoprotein-cholesterol (HDL-C) were increased in BJ and BJP groups when compared with the model group (P<0.05). In contrast, the levels of aspartate aminotransferase (AST) and alanine aminotransferase (ALT), total triglycerides (TGs), total cholesterol, low-density lipoprotein-cholesterol (LDL-C), and malondialdehyde (MDA) were lower in BJ and BJP groups than in the model group (P<0.05). The level of SIRT1 was increased, while the levels of FOXO1, phosphorylated FOXO1, acetylated FOXO1, FasL, caspase-3, BAX, and Bcl-2 were decreased in CG, BJ, and BJP groups (P<0.05). Meanwhile, SIRT1 silence resulted in increase of the levels of FOXO1, phosphorylated FOXO1, acetylated FOXO1, FasL, caspase-3, BAX, and Bcl-2. Conclusion The combination of blueberry juice and probiotics reduces apoptosis in AFLD by suppressing FOXO1, phosphorylated FOXO1, acetylated FOXO1, FasL, caspase-3, BAX, and Bcl-2 via the upregulation of SIRT1.
Collapse
Affiliation(s)
- Juanjuan Zhu
- First Hospital Affiliated to Suzhou University, Suzhou, People's Republic of China; Department of Infectious Diseases, Affiliated Hospital of Guiyang Medical College, Guiyang, People's Republic of China
| | - Tingting Ren
- Biochemistry Department, Affiliated Hospital of Guiyang Medical College, Guiyang, People's Republic of China
| | - Mingyu Zhou
- Department of Infectious Diseases, Affiliated Hospital of Guiyang Medical College, Guiyang, People's Republic of China
| | - Mingliang Cheng
- Department of Infectious Diseases, Affiliated Hospital of Guiyang Medical College, Guiyang, People's Republic of China
| |
Collapse
|
18
|
de la Monte SM, Tong M, Agarwal AR, Cadenas E. Tobacco Smoke-Induced Hepatic Injury with Steatosis, Inflammation, and Impairments in Insulin and Insulin-Like Growth Factor Signaling. ACTA ACUST UNITED AC 2016; 6. [PMID: 27525191 PMCID: PMC4979551 DOI: 10.4172/2161-0681.1000269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Alcoholic liver disease (ALD) is associated with impairments in hepatic insulin and insulin-like growth factor (IGF) signaling through cell growth, survival, and metabolic pathways. Since not all heavy drinkers develop ALD, co-factors may be important. Epidemiologic data indicate that most heavy drinkers smoke tobacco and experimental data revealed that low-level nitrosamine exposures, including those from tobacco, can cause steatohepatitis with hepatic insulin/IGF resistance and exacerbate ALD. We hypothesize that cigarette smoke (CS) exposures also cause liver injury with impaired hepatic insulin/IGF signaling, and thereby contribute to ALD. Methods Adult male A/J mice were exposed to air for 8 weeks (A8), CS for 4 (CS4) or 8 (CS8) weeks, or CS for 8 weeks with 2 weeks recovery (CS8+R). Results CS exposures caused progressive liver injury with disruption of the normal hepatic chord architecture, lobular inflammation, apoptosis or necrosis, micro-steatosis, sinusoidal dilatation, and nuclear pleomorphism. Histopathological liver injury scores increased significantly from A8 to CS4 and then further to CS8 (P<0.0001). The mean histological grade was also higher in CS8+R relative to A8 (P<0.0001) but lower than in CS4, reflecting partial resolution of injury by CS withdrawal. CS exposures impaired insulin and IGF-1 signaling through IRS-1, Akt, GSK-3β, and PRAS40. Livers from CS8+R mice had normalized or elevated levels of insulin receptor, pYpY-Insulin-R, 312S-IRS-1, 473S-Akt, S9-GSK-3β, and pT246-PRAS40 relative to A8, CS4, or CS8, reflecting partial recovery. Conclusion CS-mediated liver injury and steatohepatitis with impairments in insulin/IGF signalling are reminiscent of the findings in ALD. Therefore, CS exposures (either first or second-hand) may serve as a co-factor in ALD. The persistence of several abnormalities following CS exposure cessation suggests that some aspects of CS-mediated hepatic metabolic dysfunction are not readily reversible.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Liver Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, USA; Division of Neuropathology and Departments of Pathology, Neurology, and Neurosurgery, Rhode Island Hospital and the Alpert Medical School of Brown University, USA
| | - M Tong
- Liver Research Center, Department of Medicine, Rhode Island Hospital and the Alpert Medical School of Brown University, USA
| | - A R Agarwal
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - E Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Bettaieb A, Jiang JX, Sasaki Y, Chao TI, Kiss Z, Chen X, Tian J, Katsuyama M, Yabe-Nishimura C, Xi Y, Szyndralewiez C, Schröder K, Shah A, Brandes RP, Haj FG, Török NJ. Hepatocyte Nicotinamide Adenine Dinucleotide Phosphate Reduced Oxidase 4 Regulates Stress Signaling, Fibrosis, and Insulin Sensitivity During Development of Steatohepatitis in Mice. Gastroenterology 2015; 149:468-80.e10. [PMID: 25888330 PMCID: PMC4516583 DOI: 10.1053/j.gastro.2015.04.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 03/18/2015] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Reactive oxidative species (ROS) are believed to be involved in the progression of nonalcoholic steatohepatitis (NASH). However, little is known about the sources of ROS in hepatocytes or their role in disease progression. We studied the effects of nicotinamide adenine dinucleotide phosphate reduced oxidase 4 (NOX4) in liver tissues from patients with NASH and mice with steatohepatitis. METHODS Liver biopsy samples were obtained from 5 patients with NASH, as well as 4 patients with simple steatosis and 5 patients without steatosis (controls) from the University of California, Davis Cancer Center Biorepository. Mice with hepatocyte-specific deletion of NOX4 (NOX4(hepKO)) and NOX4(floxp+/+) C57BL/6 mice (controls) were given fast-food diets (supplemented with high-fructose corn syrup) or choline-deficient l-amino acid defined diets to induce steatohepatitis, or control diets, for 20 weeks. A separate group of mice were given the NOX4 inhibitor (GKT137831). Liver tissues were collected and immunoblot analyses were performed determine levels of NOX4, markers of inflammation and fibrosis, double-stranded RNA-activated protein kinase, and phospho-eIF-2α kinase-mediated stress signaling pathways. We performed hyperinsulinemic-euglycemic clamp studies and immunoprecipitation analyses to determine the oxidation and phosphatase activity of PP1C. RESULTS Levels of NOX4 were increased in patients with NASH compared with controls. Hepatocyte-specific deletion of NOX4 reduced oxidative stress, lipid peroxidation, and liver fibrosis in mice with diet-induced steatohepatitis. A small molecule inhibitor of NOX4 reduced liver inflammation and fibrosis and increased insulin sensitivity in mice with diet-induced steatohepatitis. In primary hepatocytes, NOX4 reduced the activity of the phosphatase PP1C, prolonging activation of double-stranded RNA-activated protein kinase and phosphorylation of extracellular signal-regulated kinase-mediated stress signaling. Mice with hepatocyte-specific deletion of NOX4 and mice given GKT137831 had increased insulin sensitivity. CONCLUSIONS NOX4 regulates oxidative stress in the liver and its levels are increased in patients with NASH and mice with diet-induced steatohepatitis. Inhibitors of NOX4 reduce liver inflammation and fibrosis and increase insulin sensitivity, and might be developed for treatment of NASH.
Collapse
Affiliation(s)
- Ahmed Bettaieb
- Department of Nutrition, University of California Davis, Davis, California
| | - Joy X Jiang
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | - Yu Sasaki
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | - Tzu-I Chao
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | - Zsofia Kiss
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | - Xiangling Chen
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | - Jijing Tian
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California
| | | | | | - Yannan Xi
- Department of Nutrition, University of California Davis, Davis, California
| | | | | | - Ajay Shah
- King's College London British Heart Foundation Centre, London, UK
| | | | - Fawaz G Haj
- Department of Nutrition, University of California Davis, Davis, California
| | - Natalie J Török
- Department of Medicine, Gastroenterology and Hepatology, University of California Davis, and VA Medical Center, Sacramento, California.
| |
Collapse
|
20
|
Song BJ, Akbar M, Jo I, Hardwick JP, Abdelmegeed MA. Translational Implications of the Alcohol-Metabolizing Enzymes, Including Cytochrome P450-2E1, in Alcoholic and Nonalcoholic Liver Disease. ADVANCES IN PHARMACOLOGY 2015; 74:303-72. [PMID: 26233911 DOI: 10.1016/bs.apha.2015.04.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fat accumulation (hepatic steatosis) in alcoholic and nonalcoholic fatty liver disease is a potentially pathologic condition which can progress to steatohepatitis (inflammation), fibrosis, cirrhosis, and carcinogenesis. Many clinically used drugs or some alternative medicine compounds are also known to cause drug-induced liver injury, which can further lead to fulminant liver failure and acute deaths in extreme cases. During liver disease process, certain cytochromes P450 such as the ethanol-inducible cytochrome P450-2E1 (CYP2E1) and CYP4A isozymes can be induced and/or activated by alcohol and/or high-fat diets and pathophysiological conditions such as fasting, obesity, and diabetes. Activation of these P450 isozymes, involved in the metabolism of ethanol, fatty acids, and various drugs, can produce reactive oxygen/nitrogen species directly and/or indirectly, contributing to oxidative modifications of DNA/RNA, proteins and lipids. In addition, aldehyde dehydrogenases including the mitochondrial low Km aldehyde dehydrogenase-2 (ALDH2), responsible for the metabolism of acetaldehyde and lipid aldehydes, can be inactivated by various hepatotoxic agents. These highly reactive acetaldehyde and lipid peroxides, accumulated due to ALDH2 suppression, can interact with cellular macromolecules DNA/RNA, lipids, and proteins, leading to suppression of their normal function, contributing to DNA mutations, endoplasmic reticulum stress, mitochondrial dysfunction, steatosis, and cell death. In this chapter, we specifically review the roles of the alcohol-metabolizing enzymes including the alcohol dehydrogenase, ALDH2, CYP2E1, and other enzymes in promoting liver disease. We also discuss translational research opportunities with natural and/or synthetic antioxidants, which can prevent or delay the onset of inflammation and liver disease.
Collapse
Affiliation(s)
- Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | - Mohammed Akbar
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Inho Jo
- Department of Molecular Medicine, Ewha Womans University School of Medicine, Seoul, South Korea
| | - James P Hardwick
- Biochemistry and Molecular Pathology in Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Mohamed A Abdelmegeed
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Eid N, Ito Y, Otsuki Y. Ethanol-induced hepatic autophagy: Friend or foe? World J Hepatol 2015; 7:1154-1156. [PMID: 26019731 PMCID: PMC4438490 DOI: 10.4254/wjh.v7.i9.1154] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/14/2015] [Accepted: 03/30/2015] [Indexed: 02/06/2023] Open
Abstract
Excessive alcohol intake may induce hepatic apoptosis, steatosis, fibrosis, cirrhosis and even cancer. Ethanol-induced activation of general or selective autophagy as mitophagy or lipophagy in hepatocytes is generally considered a prosurvival mechanism. On the other side of the coin, upregulation of autophagy in non-hepatocytes as stellate cells may stimulate fibrogenesis and subsequently induce detrimental effects on the liver. The autophagic response of other non-hepatocytes as macrophages and endothelial cells is unknown yet and needs to be investigated as these cells play important roles in ethanol-induced hepatic steatosis and damage. Selective pharmacological stimulation of autophagy in hepatocytes may be of therapeutic importance in alcoholic liver disease.
Collapse
Affiliation(s)
- Nabil Eid
- Nabil Eid, Yuko Ito, Yoshinori Otsuki, Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yuko Ito
- Nabil Eid, Yuko Ito, Yoshinori Otsuki, Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| | - Yoshinori Otsuki
- Nabil Eid, Yuko Ito, Yoshinori Otsuki, Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
22
|
Molina PE, Gardner JD, Souza-Smith FM, Whitaker AM. Alcohol abuse: critical pathophysiological processes and contribution to disease burden. Physiology (Bethesda) 2015; 29:203-15. [PMID: 24789985 DOI: 10.1152/physiol.00055.2013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Alcohol abuse; the most common and costly form of drug abuse, is a major contributing factor to many disease categories. The alcohol-attributable disease burden is closely related to the average volume of alcohol consumption, with dose-dependent relationships between amount and duration of alcohol consumption and the incidence of diabetes mellitus, hypertension, cardiovascular disease, stroke, and pneumonia. The frequent occurrence of alcohol use disorders in the adult population and the significant and widespread detrimental organ system effects highlight the importance of recognizing and further investigating the pathophysiological mechanisms underlying alcohol-induced tissue and organ injury.
Collapse
Affiliation(s)
- Patricia E Molina
- Department of Physiology and Alcohol and Drug Abuse Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | | | | | | |
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 PMCID: PMC4327341 DOI: 10.1093/alcalc/agu083] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 10/14/2014] [Accepted: 10/24/2014] [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
MESH Headings
- Alcoholism
- Animals
- Binge Drinking
- Carcinogens/pharmacology
- Central Nervous System Depressants/pharmacology
- Central Nervous System Depressants/toxicity
- Disease Models, Animal
- Endoplasmic Reticulum/drug effects
- Endoplasmic Reticulum/metabolism
- Ethanol/pharmacology
- Ethanol/toxicity
- Fatty Liver, Alcoholic/etiology
- Fatty Liver, Alcoholic/metabolism
- Fatty Liver, Alcoholic/pathology
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Insulin/metabolism
- Insulin Resistance
- Insulin-Like Growth Factor I/metabolism
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver Diseases, Alcoholic/metabolism
- Liver Diseases, Alcoholic/pathology
- Mitochondria, Liver/drug effects
- Mitochondria, Liver/metabolism
- Necrosis
- Nitrosamines/pharmacology
- Rats
- Rats, Long-Evans
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/metabolism
- Signal Transduction/drug effects
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
|
Anavi S, Ni Z, Tirosh O, Fedorova M. Steatosis-induced proteins adducts with lipid peroxidation products and nuclear electrophilic stress in hepatocytes. Redox Biol 2014; 4:158-68. [PMID: 25560244 PMCID: PMC4309854 DOI: 10.1016/j.redox.2014.12.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/16/2014] [Accepted: 12/16/2014] [Indexed: 01/21/2023] Open
Abstract
Accumulating evidence suggests that fatty livers are particularly more susceptible to several pathological conditions, including hepatic inflammation, cirrhosis and liver cancer. However the exact mechanism of such susceptibility is still largely obscure. The current study aimed to elucidate the effect of hepatocytes lipid accumulation on the nuclear electrophilic stress. Accumulation of intracellular lipids was significantly increased in HepG2 cells incubated with fatty acid (FA) complex (1 mM, 2:1 oleic and palmitic acids). In FA-treated cells, lipid droplets were localized around the nucleus and seemed to induce mechanical force, leading to the disruption of the nucleus morphology. Level of reactive oxygen species (ROS) was significantly increased in FA-loaded cells and was further augmented by treatment with moderate stressor (CoCl2). Increased ROS resulted in formation of reactive carbonyls (aldehydes and ketones, derived from lipid peroxidation) with a strong perinuclear accumulation. Mass-spectroscopy analysis indicated that lipid accumulation per-se can results in modification of nuclear protein by reactive lipid peroxidation products (oxoLPP). 235 Modified proteins involved in transcription regulation, splicing, protein synthesis and degradation, DNA repair and lipid metabolism were identified uniquely in FA-treated cells. These findings suggest that steatosis can affect nuclear redox state, and induce modifications of nuclear proteins by reactive oxoLPP accumulated in the perinuclear space upon FA-treatment. Effects of fatty acids on modification of nuclear proteins in hepatocytes was studied. Lipid accumulation was associated with abnormal nuclear morphology. Lipid accumulation promoted mitochondrial activity and enhanced ROS generation. Nuclear proteins were modified by lipid peroxidation products. Data suggest nuclear stress as a mechanism for fatty liver disease progression.
Collapse
Affiliation(s)
- Sarit Anavi
- Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, 76100 Rehovot, Israel
| | - Zhixu Ni
- Faculty of Chemistry and Mineralogy, Institute of Bioanalytical Chemistry, Leipzig, Germany; Center for Biotechnology and Biomedicine, Universität Leipzig, Leipzig 04103, Germany
| | - Oren Tirosh
- Robert H. Smith Faculty of Agriculture, Food and Environment, Institute of Biochemistry, Food Science and Nutrition, The Hebrew University of Jerusalem, 76100 Rehovot, Israel.
| | - Maria Fedorova
- Faculty of Chemistry and Mineralogy, Institute of Bioanalytical Chemistry, Leipzig, Germany; Center for Biotechnology and Biomedicine, Universität Leipzig, Leipzig 04103, Germany.
| |
Collapse
|
25
|
Abstract
Oxidative stress is a common feature observed in a wide spectrum of chronic liver diseases including viral hepatitis, alcoholic, and nonalcoholic steatohepatitis. The nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are emerging as major sources of reactive oxygen species (ROS). Several major isoforms are expressed in the liver, including NOX1, NOX2, and NOX4. While the phagocytic NOX2 has been known to play an important role in Kupffer cell and neutrophil phagocytic activity and inflammation, the nonphagocytic NOX homologues are increasingly recognized as key enzymes in oxidative injury and wound healing. In this review, we will summarize the current advances in knowledge on the regulatory pathways of NOX activation, their cellular distribution, and their role in the modulation of redox signaling in liver diseases.
Collapse
|
26
|
Zou X, Yan C, Shi Y, Cao K, Xu J, Wang X, Chen C, Luo C, Li Y, Gao J, Pang W, Zhao J, Zhao F, Li H, Zheng A, Sun W, Long J, Szeto IMY, Zhao Y, Dong Z, Zhang P, Wang J, Lu W, Zhang Y, Liu J, Feng Z. Mitochondrial dysfunction in obesity-associated nonalcoholic fatty liver disease: the protective effects of pomegranate with its active component punicalagin. Antioxid Redox Signal 2014; 21:1557-70. [PMID: 24393106 PMCID: PMC4175030 DOI: 10.1089/ars.2013.5538] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Punicalagin (PU) is one of the major ellagitannins found in the pomegranate (Punica granatum), which is a popular fruit with several health benefits. So far, no studies have evaluated the effects of PU on nonalcoholic fatty liver disease (NAFLD). Our work aims at studying the effect of PU-enriched pomegranate extract (PE) on high fat diet (HFD)-induced NAFLD. RESULTS PE administration at a dosage of 150 mg/kg/day significantly inhibited HFD-induced hyperlipidemia and hepatic lipid deposition. As major contributors to NAFLD, increased expression of pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukins 1, 4, and 6 as well as augmented oxidative stress in hepatocytes followed by nuclear factor (erythroid-derived-2)-like 2 (Nrf2) activation were normalized through PE supplementation. In addition, PE treatment reduced uncoupling protein 2 (UCP2) expression, restored ATP content, suppressed mitochondrial protein oxidation, and improved mitochondrial complex activity in the liver. In contrast, mitochondrial content was not affected despite increased peroxisomal proliferator-activated receptor-gamma coactivator-1α (PGC-1α) and elevated expression of genes related to mitochondrial beta-oxidation after PE treatment. Finally, PU was identified as the predominant active component of PE with regard to the lowering of triglyceride and cholesterol content in HepG2 cells, and both PU- and PE-protected cells from palmitate induced mitochondrial dysfunction and insulin resistance. INNOVATION Our work presents the beneficial effects of PE on obesity-associated NAFLD and multiple risk factors. PU was proposed to be the major active component. CONCLUSIONS By promoting mitochondrial function, eliminating oxidative stress and inflammation, PU may be a useful nutrient for the treatment of NAFLD.
Collapse
Affiliation(s)
- Xuan Zou
- 1 The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Frontier Institute of Science and Technology, Xi'an Jiaotong University , Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The ever growing prevalence of childhood obesity is being accompanied by an increase in the pediatric population of diseases once believed to be exclusive of the adulthood such as the metabolic syndrome (MS). The MS has been defined as the link between insulin resistance, hypertension, dyslipidemia, impaired glucose tolerance, and other metabolic abnormalities associated with an increased risk of atherosclerotic cardiovascular diseases in adults. In this review, we will discuss the peculiar aspects of the pediatric MS and the role of novel molecules and biomarkers in its pathogenesis.
Collapse
|
28
|
Andringa KK, Udoh US, Landar A, Bailey SM. Proteomic analysis of 4-hydroxynonenal (4-HNE) modified proteins in liver mitochondria from chronic ethanol-fed rats. Redox Biol 2014; 2:1038-47. [PMID: 25454745 PMCID: PMC4297939 DOI: 10.1016/j.redox.2014.09.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 09/26/2014] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol-mediated oxidative stress and lipid peroxidation increases the levels of various reactive lipid species including 4-hydroxynonenal (4-HNE), which can subsequently modify proteins in the liver. It has been proposed that 4-HNE modification adversely affects the structure and/or function of mitochondrial proteins, thereby impairing mitochondrial metabolism. To determine whether chronic ethanol consumption increases levels of 4-HNE modified proteins in mitochondria, male rats were fed control and ethanol-containing diets for 5 weeks and mitochondrial samples were analyzed using complementary proteomic methods. Five protein bands (approx. 35, 45, 50, 70, and 90kDa) showed strong immunoreactivity for 4-HNE modified proteins in liver mitochondria from control and ethanol-fed rats when proteins were separated by standard 1D SDS-PAGE. Using high-resolution proteomic methods (2D IEF/SDS-PAGE and BN-PAGE) we identified several mitochondrial proteins immunoreactive for 4-HNE, which included mitofilin, dimethylglycine dehydrogenase, choline dehydrogenase, electron transfer flavoprotein α, cytochrome c1, enoyl CoA hydratase, and cytochrome c. The electron transfer flavoprotein α consistently showed increased 4-HNE immunoreactivity in mitochondria from ethanol-fed rats as compared to mitochondria from the control group. Increased 4-HNE reactivity was also detected for dimethylglycine dehydrogenase, enoyl CoA hydratase, and cytochrome c in ethanol samples when mitochondria were analyzed by BN-PAGE. In summary, this work identifies new targets of 4-HNE modification in mitochondria and provides useful information needed to better understand the molecular mechanisms underpinning chronic ethanol-induced mitochondrial dysfunction and liver injury.
Collapse
Affiliation(s)
- Kelly K Andringa
- Department of Medicine, Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Uduak S Udoh
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Aimee Landar
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shannon M Bailey
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
29
|
Reiniers MJ, van Golen RF, van Gulik TM, Heger M. Reactive oxygen and nitrogen species in steatotic hepatocytes: a molecular perspective on the pathophysiology of ischemia-reperfusion injury in the fatty liver. Antioxid Redox Signal 2014; 21:1119-42. [PMID: 24294945 PMCID: PMC4123468 DOI: 10.1089/ars.2013.5486] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/14/2013] [Accepted: 12/02/2013] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Hepatic ischemia-reperfusion (IR) injury results from the temporary deprivation of hepatic blood supply and is a common side effect of major liver surgery (i.e., transplantation or resection). IR injury, which in most severe cases culminates in acute liver failure, is particularly pronounced in livers that are affected by non-alcoholic fatty liver disease (NAFLD). In NAFLD, fat-laden hepatocytes are damaged by chronic oxidative/nitrosative stress (ONS), a state that is acutely exacerbated during IR, leading to extensive parenchymal damage. RECENT ADVANCES NAFLD triggers ONS via increased (extra)mitochondrial fatty acid oxidation and activation of the unfolded protein response. ONS is associated with widespread protein and lipid (per)oxidation, which reduces the hepatic antioxidative capacity and shifts the intracellular redox status toward an oxidized state. Moreover, activation of the transcription factor peroxisome proliferator-activated receptor α induces expression of mitochondrial uncoupling protein 2, resulting in depletion of cellular energy (ATP) reserves. The reduction in intracellular antioxidants and ATP in fatty livers consequently gives rise to severe ONS and necrotic cell death during IR. CRITICAL ISSUES Despite the fact that ONS mediates both NAFLD and IR injury, the interplay between the two conditions has never been described in detail. An integrative overview of the pathophysiology of NAFLD that renders steatotic hepatocytes more vulnerable to IR injury is therefore presented in the context of ONS. FUTURE DIRECTIONS Effective methods should be devised to alleviate ONS and the consequences thereof in NAFLD before surgery in order to improve resilience of fatty livers to IR injury.
Collapse
Affiliation(s)
- Megan J Reiniers
- Department of Surgery, Surgical Laboratory, Academic Medical Center, University of Amsterdam , Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
30
|
Mitochondrial-nuclear genome interactions in non-alcoholic fatty liver disease in mice. Biochem J 2014; 461:223-32. [PMID: 24758559 DOI: 10.1042/bj20131433] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
NAFLD (non-alcoholic fatty liver disease) involves significant changes in liver metabolism characterized by oxidative stress, lipid accumulation and fibrogenesis. Mitochondrial dysfunction and bioenergetic defects also contribute to NAFLD. In the present study, we examined whether differences in mtDNA influence NAFLD. To determine the role of mitochondrial and nuclear genomes in NAFLD, MNX (mitochondrial-nuclear exchange) mice were fed an atherogenic diet. MNX mice have mtDNA from C57BL/6J mice on a C3H/HeN nuclear background and vice versa. Results from MNX mice were compared with wild-type C57BL/6J and C3H/HeN mice fed a control or atherogenic diet. Mice with the C57BL/6J nuclear genome developed more macrosteatosis, inflammation and fibrosis compared with mice containing the C3H/HeN nuclear genome when fed the atherogenic diet. These changes were associated with parallel alterations in inflammation and fibrosis gene expression in wild-type mice, with intermediate responses in MNX mice. Mice with the C57BL/6J nuclear genome had increased State 4 respiration, whereas MNX mice had decreased State 3 respiration and RCR (respiratory control ratio) when fed the atherogenic diet. Complex IV activity and most mitochondrial biogenesis genes were increased in mice with the C57BL/6J nuclear or mitochondrial genome, or both fed the atherogenic diet. These results reveal new interactions between mitochondrial and nuclear genomes and support the concept that mtDNA influences mitochondrial function and metabolic pathways implicated in NAFLD.
Collapse
|
31
|
Functional roles of protein nitration in acute and chronic liver diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:149627. [PMID: 24876909 PMCID: PMC4021747 DOI: 10.1155/2014/149627] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/01/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Abstract
Nitric oxide, when combined with superoxide, produces peroxynitrite, which is known to be an important mediator for a number of diseases including various liver diseases. Peroxynitrite can modify tyrosine residue(s) of many proteins resulting in protein nitration, which may alter structure and function of each target protein. Various proteomics and immunological methods including mass spectrometry combined with both high pressure liquid chromatography and 2D PAGE have been employed to identify and characterize nitrated proteins from pathological tissue samples to determine their roles. However, these methods contain a few technical problems such as low efficiencies with the detection of a limited number of nitrated proteins and labor intensiveness. Therefore, a systematic approach to efficiently identify nitrated proteins and characterize their functional roles is likely to shed new insights into understanding of the mechanisms of hepatic disease pathophysiology and subsequent development of new therapeutics. The aims of this review are to briefly describe the mechanisms of hepatic diseases. In addition, we specifically describe a systematic approach to efficiently identify nitrated proteins to study their causal roles or functional consequences in promoting acute and chronic liver diseases including alcoholic and nonalcoholic fatty liver diseases. We finally discuss translational research applications by analyzing nitrated proteins in evaluating the efficacies of potentially beneficial agents to prevent or treat various diseases in the liver and other tissues.
Collapse
|
32
|
Nascimento AF, Ip BC, Luvizotto RAM, Seitz HK, Wang XD. Aggravation of nonalcoholic steatohepatitis by moderate alcohol consumption is associated with decreased SIRT1 activity in rats. Hepatobiliary Surg Nutr 2014; 2:252-9. [PMID: 24570955 DOI: 10.3978/j.issn.2304-3881.2013.07.05] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 07/25/2013] [Indexed: 01/22/2023]
Abstract
Chronic alcohol intake decreases adiponectin and sirtuin 1 (SIRT1) expressions, both of which have been implicated in various biological processes including inflammation, apoptosis and metabolism. We have previously shown that moderate consumption of alcohol aggravates liver inflammation and apoptosis in rats with pre-existing nonalcoholic steatohepatitis (NASH). This study investigated whether moderate alcohol intake alters SIRT1 activity, adiponectin/Adiponectin receptor (AdipoR)-related signaling and lipid metabolism in a pre-existing NASH status. Sprague-Dawley rats were fed with a high-fat diet (71% energy from fat) for 6 weeks to induce NASH then subsequently divided into 2 sub-groups: fed either a modified high-fat diet (HFD, 55% energy from fat) or a modified high-fat alcoholic diet (HFA, 55% energy from fat and 16% energy from ethanol) for an additional 4 weeks. We observed in comparison to HFD group, HFA increased hepatic nuclear SIRT1 protein but decreased its deacetylase activity. SREBP-1c protein expression and FAS mRNA levels were significantly upregulated, while DGAT1/2 and CPT-I mRNA levels were downregulated in the livers of HFA compared to HFD. Although hepatic AdipoR1 decreased, HFA did not alter AdipoR2 and their downstream signaling. There were no significant changes in plasma adiponectin and free fatty acids (FFA), as well as adiponectin expression in adipose tissue between the two groups. The present study indicates that suppression in SIRT1 deacetylase activity contributes to alcohol-exacerbated hepatic inflammation and apoptosis in rats with pre-existing NASH. In addition, moderate alcohol intake did not modulate adiponectin/AdipoR signaling axis in this model.
Collapse
Affiliation(s)
- André F Nascimento
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; ; Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Blanche C Ip
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Renata A M Luvizotto
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA; ; Department of Internal Medicine, Botucatu School of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Helmut K Seitz
- Alcohol Research Centre, University of Heidelberg and Department of Medicine, Salem Medical Centre, Heidelberg, Germany
| | - Xiang-Dong Wang
- Nutrition and Cancer Biology Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
33
|
Liang Y, Harris FL, Jones DP, Brown LAS. Alcohol induces mitochondrial redox imbalance in alveolar macrophages. Free Radic Biol Med 2013; 65:1427-1434. [PMID: 24140864 PMCID: PMC3870467 DOI: 10.1016/j.freeradbiomed.2013.10.010] [Citation(s) in RCA: 30] [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] [Received: 06/21/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
Alcohol abuse suppresses the immune responses of alveolar macrophages (AMs) and increases the risk of a respiratory infection via chronic oxidative stress and depletion of critical antioxidants within alveolar cells and the alveolar lining fluid. Although alcohol-induced mitochondrial oxidative stress has been demonstrated, the oxidation of the mitochondrial thioredoxin redox circuit in response to alcohol has not been examined. In vitro ethanol exposure of a mouse AM cell line and AMs from ethanol-fed mice demonstrated NADPH depletion concomitant with oxidation of mitochondrial glutathione and oxidation of the thioredoxin redox circuit system including thioredoxin 2 (Trx2) and thioredoxin 2 reductase (Trx2R). Mitochondrial peroxiredoxins (Prdx's), which are critical for the reduction of the thioredoxin circuit, were irreversibly hyperoxidized to an inactive form. Ethanol also decreased the mRNAs for Trx2, Trx2R, Prdx3, and Prdx5 plus the mitochondrial thiol-disulfide proteins glutaredoxin 2, glutathione reductase, and glutathione peroxidase 2. Thus, the mitochondrial thioredoxin circuit was highly oxidized by ethanol, thereby compromising the mitochondrial antioxidant capacity and ability to detoxify mitochondrial reactive oxygen species. Oxidation of the mitochondrial thioredoxin redox circuit would further compromise the transient oxidation of thiol groups within specific proteins, the basis of redox signaling, and the processes by which cells respond to oxidants. Impaired mitochondria can then jeopardize cellular function of AMs, such as phagocytosis, which may explain the increased risk of respiratory infection in subjects with an alcohol use disorder.
Collapse
Affiliation(s)
- Yan Liang
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Frank L Harris
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Lou Ann S Brown
- Department of Pediatrics, Allergy & Critical Care Medicine, School of Medicine, Emory University, Atlanta, GA 30322, USA; Center for Developmental Lung Biology, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| |
Collapse
|
34
|
Serviddio G, Bellanti F, Vendemiale G. Free radical biology for medicine: learning from nonalcoholic fatty liver disease. Free Radic Biol Med 2013; 65:952-968. [PMID: 23994574 DOI: 10.1016/j.freeradbiomed.2013.08.174] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 08/20/2013] [Accepted: 08/20/2013] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species, when released under controlled conditions and limited amounts, contribute to cellular proliferation, senescence, and survival by acting as signaling intermediates. In past decades there has been an epidemic diffusion of nonalcoholic fatty liver disease (NAFLD) that represents the result of the impairment of lipid metabolism, redox imbalance, and insulin resistance in the liver. To date, most studies and reviews have been focused on the molecular mechanisms by which fatty liver progresses to steatohepatitis, but the processes leading toward the development of hepatic steatosis in NAFLD are not fully understood yet. Several nuclear receptors, such as peroxisome proliferator-activated receptors (PPARs) α/γ/δ, PPARγ coactivators 1α and 1β, sterol-regulatory element-binding proteins, AMP-activated protein kinase, liver-X-receptors, and farnesoid-X-receptor, play key roles in the regulation of lipid homeostasis during the pathogenesis of NAFLD. These nuclear receptors may act as redox sensors and may modulate various metabolic pathways in response to specific molecules that act as ligands. It is conceivable that a redox-dependent modulation of lipid metabolism, nuclear receptor-mediated, could cause the development of hepatic steatosis and insulin resistance. Thus, this network may represent a potential therapeutic target for the treatment and prevention of hepatic steatosis and its progression to steatohepatitis. This review summarizes the redox-dependent factors that contribute to metabolism alterations in fatty liver with a focus on the redox control of nuclear receptors in normal liver as well as in NAFLD.
Collapse
Affiliation(s)
- Gaetano Serviddio
- C.U.R.E. Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy.
| | - Francesco Bellanti
- C.U.R.E. Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| | - Gianluigi Vendemiale
- C.U.R.E. Centre for Liver Disease Research and Treatment, Institute of Internal Medicine, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy
| |
Collapse
|
35
|
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: 2.6] [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
|
36
|
Aliche-Djoudi F, Podechard N, Collin A, Chevanne M, Provost E, Poul M, Le Hégarat L, Catheline D, Legrand P, Dimanche-Boitrel MT, Lagadic-Gossmann D, Sergent O. A role for lipid rafts in the protection afforded by docosahexaenoic acid against ethanol toxicity in primary rat hepatocytes. Food Chem Toxicol 2013; 60:286-96. [PMID: 23907024 DOI: 10.1016/j.fct.2013.07.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/24/2013] [Accepted: 07/22/2013] [Indexed: 12/17/2022]
Abstract
Previously, we demonstrated that eicosapentaenoic acid enhanced ethanol-induced oxidative stress and cell death in primary rat hepatocytes via an increase in membrane fluidity and lipid raft clustering. In this context, another n-3 polyunsaturated fatty acid, docosahexaenoic acid (DHA), was tested with a special emphasis on physical and chemical alteration of lipid rafts. Pretreatment of hepatocytes with DHA reduced significantly ethanol-induced oxidative stress and cell death. DHA protection could be related to an alteration of lipid rafts. Indeed, rafts exhibited a marked increase in membrane fluidity and packing defects leading to the exclusion of a raft protein marker, flotillin. Furthermore, DHA strongly inhibited disulfide bridge formation, even in control cells, thus suggesting a disruption of protein-protein interactions inside lipid rafts. This particular spatial organization of lipid rafts due to DHA subsequently prevented the ethanol-induced lipid raft clustering. Such a prevention was then responsible for the inhibition of phospholipase C-γ translocation into rafts, and consequently of both lysosome accumulation and elevation in cellular low-molecular-weight iron content, a prooxidant factor. In total, the present study suggests that DHA supplementation could represent a new preventive approach for patients with alcoholic liver disease based upon modulation of the membrane structures.
Collapse
Affiliation(s)
- Fatiha Aliche-Djoudi
- UMR Inserm 1085, IRSET, UFR des Sciences Pharmaceutiques et Biologiques, 2, av Pr Léon Bernard, 35043 Rennes Cédex, France; Université de Rennes 1, Biosit UMS3080, 2, av Pr Léon Bernard, 35043 Rennes Cédex, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Eid N, Ito Y, Maemura K, Otsuki Y. Elevated autophagic sequestration of mitochondria and lipid droplets in steatotic hepatocytes of chronic ethanol-treated rats: an immunohistochemical and electron microscopic study. J Mol Histol 2013; 44:311-326. [PMID: 23371376 DOI: 10.1007/s10735-013-9483-x] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/02/2013] [Indexed: 02/07/2023]
Abstract
Ethanol-induced hepatic steatosis may induce the progression of alcoholic liver disease. The involvement of autophagic clearance of damaged mitochondria (mitophagy) and lipid droplets (LDs) (lipophagy) in chronic ethanol-induced hepatic steatosis is not clearly understood. Adult Wistar rats were fed either 5 % ethanol in Lieber-DeCarli liquid diet or an isocaloric control diet for 10 weeks. Light microscopy showed marked steatosis in hepatocytes of ethanol-treated rats (ETRs), which was further revealed by transmission electron microscopy (TEM), where significant numbers of large LDs and damaged mitochondria were detected in steatotic hepatocytes. Moreover, TEM demonstrated that hepatocyte steatosis was associated with greatly enhanced autophagic vacuole (AV) formation compared to control hepatocytes. Mitochondria and LDs were the predominant contents of AVs in steatotic hepatocytes. Immunohistochemistry of LC3, a specific marker of early AVs (autophagosomes), demonstrated an extensive punctate pattern in hepatocytes of ETRs, while LC3 puncta were much less frequent in control hepatocytes. This was confirmed by immunoelectron microscopy (IEM), which showed localization of LC3 to autophagosomes sequestering damaged mitochondria and LDs. In addition, IEM revealed that PINK1 (a sensor of mitochondrial damage and marker of mitophagy) was overexpressed in mitochondria of ETRs. Enhanced autophagic lysosomal activity was evidenced by increased immunolabeling of LAMP-2, a marker of late AVs (autolysosomes) in hepatocytes of ETRs and colocalization of LC3 and lysosomal cathepsins using double immunofluorescence labeling. Increased AVs in hepatocytes of ETRs reflect ethanol toxicity and could represent a possible protective mechanism via stimulation of mitophagy and lipophagy.
Collapse
Affiliation(s)
- Nabil Eid
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki 569-8686, Osaka, Japan
| | | | | | | |
Collapse
|
38
|
Smathers RL, Galligan JJ, Shearn CT, Fritz KS, Mercer K, Ronis M, Orlicky DJ, Davidson NO, Petersen DR. Susceptibility of L-FABP-/- mice to oxidative stress in early-stage alcoholic liver. J Lipid Res 2013; 54:1335-45. [PMID: 23359610 DOI: 10.1194/jlr.m034892] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronic ethanol consumption is a prominent cause of liver disease worldwide. Dysregulation of an important lipid uptake and trafficking gene, liver-fatty acid binding protein (L-FABP), may contribute to alterations in lipid homeostasis during early-stage alcoholic liver. We have reported the detrimental effects of ethanol on the expression of L-FABP and hypothesize this may deleteriously impact metabolic networks regulating fatty acids. Male wild-type (WT) and L-FABP(-/-) mice were fed a modified Lieber-DeCarli liquid diet for six weeks. To assess the response to chronic ethanol ingestion, standard biochemical indicators for alcoholic liver disease (ALD) and oxidative stress were measured. Ethanol ingestion resulted in attenuation of hepatic triglyceride accumulation and elevation of cholesterol in L-FABP(-/-) mice. Lipidomics analysis validated multiple alterations in hepatic lipids resulting from ethanol treatment. Increased immunohistochemical staining for the reactive aldehydes 4-hydroxynonenal and malondialdehyde were observed in WT mice ingesting ethanol; however, L-FABP(-/-) mice displayed prominent protein adducts in liver sections evaluated from pair-fed and ethanol-fed mice. Likewise, alterations in glutathione, thiobarbituric acid reactive substances (TBARS), 8-isoprostanes, and protein carbonyl content all indicated L-FABP(-/-) mice exhibit high sustained oxidative stress in the liver. These data establish that L-FABP is an indirect antioxidant protein essential for sequestering FFA and that its impairment could contribute to in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Rebecca L Smathers
- Skaggs School of Pharmaceutical Sciences and Pharmacy and University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Activation of inflammatory signaling pathways is of central importance in the pathogenesis of alcoholic liver disease (ALD) and nonalcoholic steatohepatitis (NASH). Recent studies demonstrated that Toll-like receptors, the sensors of microbial and endogenous danger signals, are expressed and activated in innate immune cells as well as in parenchymal cells in the liver and thereby contribute to ALD and NASH. In this review, we emphasize the importance of gut-derived endotoxin and its recognition by TLR4 in the liver. The significance of TLR-induced intracellular signaling pathways and cytokine production as well as the contribution of individual cell types to the inflammation is evaluated. The contribution of TLR signaling to the induction of liver fibrosis and to the progression of liver pathology mediated by viral pathogens is reviewed in the context of ALD and NASH.
Collapse
Affiliation(s)
- Jan Petrasek
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | | |
Collapse
|
40
|
Galastri S, Zamara E, Milani S, Novo E, Provenzano A, Delogu W, Vizzutti F, Sutti S, Locatelli I, Navari N, Vivoli E, Caligiuri A, Pinzani M, Albano E, Parola M, Marra F. Lack of CC chemokine ligand 2 differentially affects inflammation and fibrosis according to the genetic background in a murine model of steatohepatitis. Clin Sci (Lond) 2012; 123:459-471. [PMID: 22545719 PMCID: PMC3369401 DOI: 10.1042/cs20110515] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 04/10/2012] [Accepted: 04/30/2012] [Indexed: 01/01/2023]
Abstract
Expression of CCL2 (CC chemokine ligand 2) (or monocyte chemoattractant protein-1) regulates inflammatory cell infiltration in the liver and adipose tissue, favouring steatosis. However, its role in the pathogenesis of steatohepatitis is still uncertain. In the present study, we investigated the development of non-alcoholic steatohepatitis induced by an MCD diet (methionine/choline-deficient diet) in mice lacking the CCL2 gene on two different genetic backgrounds, namely Balb/C and C57/Bl6J. WT (wild-type) and CCL2-KO (knockout) mice were fed on a lipid-enriched MCD diet or a control diet for 8 weeks. In Balb/C mice fed on the MCD diet, a lack of CCL2 was associated with lower ALT (alanine transaminase) levels and reduced infiltration of inflammatory cells, together with a lower generation of oxidative-stress-related products. Sirius Red staining demonstrated pericellular fibrosis in zone 3, and image analysis showed a significantly lower matrix accumulation in CCL2-KO mice. This was associated with reduced hepatic expression of TGF-β (transforming growth factor-β), type I procollagen, TIMP-1 (tissue inhibitor of metalloproteinases-1) and α-smooth muscle actin. In contrast, in mice on a C57Bl/6 background, neither ALT levels nor inflammation or fibrosis were significantly different comparing WT and CCL2-KO animals fed on an MCD diet. In agreement, genes related to fibrogenesis were expressed to comparable levels in the two groups of animals. Comparison of the expression of several genes involved in inflammation and repair demonstrated that IL (interleukin)-4 and the M2 marker MGL-1 (macrophage galactose-type C-type lectin 1) were differentially expressed in Balb/C and C57Bl/6 mice. No significant differences in the degree of steatosis were observed in all groups of mice fed on the MCD diet. We conclude that, in experimental murine steatohepatitis, the effects of CCL2 deficiency are markedly dependent on the genetic background.
Collapse
Key Words
- cc chemokine ligand 2
- chemokine
- cytokine
- liver fibrosis
- non-alcoholic steatohepatitis
- alt, alanine transaminase
- ast, aspartate aminotransferase
- ccl2, cc chemokine ligand 2
- ccr2, cc chemokine receptor 2
- fam, 6-carboxyfluorescein
- gapdh, glyceraldehyde-3-phosphate dehydrogenase
- hne, 4-hydroxynonenal, ifn-γ, interferon-γ
- il, interleukin
- inos, inducible no synthase
- ko, knockout
- mcd diet, methionine/choline-deficient diet
- mgl1, macrophage galactose-type c-type lectin 1
- nafld, non-alcoholic fatty liver disease
- nash, non-alcoholic steatohepatitis
- ros, reactive oxygen species
- rt, real-time
- tgf-β, transforming growth factor-β
- timp-1, tissue inhibitor of metalloproteinases-1
- tnfα, tumour necrosis factor α
- wt, wild-type
Collapse
Affiliation(s)
- Sara Galastri
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Elena Zamara
- †Dipartimento di Medicina e Oncologia Sperimentali, University of Turin, Turin, Italy
| | - Stefano Milani
- ‡Dipartimento di Fisiopatologia Clinica, University of Florence, Florence, Italy
- §Center for Research, Transfer and Higher Education DenoTHE, University of Florence, Florence, Italy
| | - Erica Novo
- †Dipartimento di Medicina e Oncologia Sperimentali, University of Turin, Turin, Italy
| | - Angela Provenzano
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Wanda Delogu
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Francesco Vizzutti
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Salvatore Sutti
- ∥Dipartimento di Scienze Mediche, University of Eastern Piedmont, Novara, Italy
| | - Irene Locatelli
- ∥Dipartimento di Scienze Mediche, University of Eastern Piedmont, Novara, Italy
| | - Nadia Navari
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Elisa Vivoli
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
| | - Alessandra Caligiuri
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
- §Center for Research, Transfer and Higher Education DenoTHE, University of Florence, Florence, Italy
| | - Massimo Pinzani
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
- §Center for Research, Transfer and Higher Education DenoTHE, University of Florence, Florence, Italy
| | - Emanuele Albano
- ∥Dipartimento di Scienze Mediche, University of Eastern Piedmont, Novara, Italy
| | - Maurizio Parola
- †Dipartimento di Medicina e Oncologia Sperimentali, University of Turin, Turin, Italy
| | - Fabio Marra
- *Dipartimento di Medicina Interna, University of Florence, Florence, Italy
- §Center for Research, Transfer and Higher Education DenoTHE, University of Florence, Florence, Italy
| |
Collapse
|
41
|
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: 4.8] [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
|