51
|
Püschel GP, Henkel J. Dietary cholesterol does not break your heart but kills your liver. Porto Biomed J 2019; 3:e12. [PMID: 31595236 PMCID: PMC6726297 DOI: 10.1016/j.pbj.0000000000000012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
It is increasingly accepted that dietary cholesterol has a much lower impact on the progression of cardiovascular disease than previously assumed. However, both animal experiments and human studies seem to support the view that dietary cholesterol may contribute to the transition from benign steatosis to the potentially fatal non-alcoholic steatohepatitis. Cholesterol esters and cholesterol accumulate in the hepatocyte and impair its function. This leads to oxidative stress and endoplasmic reticulum stress triggering the release of pro-inflammatory cytokines and rendering the hepatocyte more susceptible to apoptotic or necrotic cell death. Kupffer cells group around dying hepatocytes and phagocytose the hepatocyte debris and lipids. In addition, they are exposed to lipid peroxidation products released from hepatocytes. Kupffer cells, thus activated, release pro-inflammatory, chemotactic and profibrotic cytokines that promote inflammation and fibrosis. Therefore, dietary cholesterol may be harmful to the liver, in particular when administered in combination with polyunsaturated fatty acids that favor lipid peroxidation.
Collapse
Affiliation(s)
- Gerhard P Püschel
- Department of Nutritional Biochemistry, University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| | - Janin Henkel
- Department of Nutritional Biochemistry, University of Potsdam, Institute of Nutritional Science, Nuthetal, Germany
| |
Collapse
|
52
|
Park JS, Lee YS, Lee DH, Bae SH. Repositioning of niclosamide ethanolamine (NEN), an anthelmintic drug, for the treatment of lipotoxicity. Free Radic Biol Med 2019; 137:143-157. [PMID: 31035006 DOI: 10.1016/j.freeradbiomed.2019.04.030] [Citation(s) in RCA: 18] [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] [Received: 02/19/2019] [Revised: 04/12/2019] [Accepted: 04/25/2019] [Indexed: 12/24/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is a common liver disease associated with metabolic disorders, including obesity and type 2 diabetes (T2D). Despite its worldwide prevalence, there are no effective drugs for the treatment of NASH. The progression of NASH is mainly accelerated by reactive oxygen species (ROS)-induced lipotoxicity. The transcription factor known as nuclear factor erythroid 2-related factor 2 (Nrf2) is pivotal for the elimination of ROS. Accordingly, activators of Nrf2 have been implicated as promising therapeutic targets for the treatment of NASH. Niclosamide (ethanolamine salt; NEN), a drug approved by the US Food and Drug Administration (USFDA), is currently used as an anthelmintic drug for the treatment of parasitic infections. Recently, NEN was shown to improve hepatic steatosis in high-fat diet (HFD)-fed mice. However, the underlying mechanism of its antioxidant function in NASH remains unknown. Here, we demonstrate that NEN induces AMPK-mediated phosphorylation of p62 at S351 that can lead to noncanonical Nrf2 activation. We also demonstrate that NEN protects cells and mouse liver from acute lipotoxic stress through activating p62-dependent Keap1-Nrf2 pathway. Taken together, NEN can be used for clinical applications and has the potential to provide a new therapeutic option for NASH.
Collapse
Affiliation(s)
- Jeong Su Park
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yu Seol Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Republic of Korea
| | - Da Hyun Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Republic of Korea
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
53
|
Li L, Fu J, Sun J, Liu D, Chen C, Wang H, Hou Y, Xu Y, Pi J. Is Nrf2-ARE a potential target in NAFLD mitigation? CURRENT OPINION IN TOXICOLOGY 2019. [DOI: 10.1016/j.cotox.2018.12.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
54
|
Xu D, Xu M, Jeong S, Qian Y, Wu H, Xia Q, Kong X. The Role of Nrf2 in Liver Disease: Novel Molecular Mechanisms and Therapeutic Approaches. Front Pharmacol 2019; 9:1428. [PMID: 30670963 PMCID: PMC6331455 DOI: 10.3389/fphar.2018.01428] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 11/19/2018] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress and inflammation are the most important pathogenic events in the development and progression of liver diseases. Nuclear erythroid 2-related factor 2 (Nrf2) is the master regulator of the cellular protection via induction of anti-inflammatory, antioxidant, and cyto-protective genes expression. Multiple studies have shown that activation or suppression of this transcriptional factor significantly affect progression of liver diseases. Comprehensive understanding the roles of Nrf2 activation/expression and the outcomes of its activators/inhibitors are indispensable for defining the mechanisms and therapeutic strategies against liver diseases. In this current review, we discussed recent advances in the function and principal mechanisms by regulating Nrf2 in liver diseases, including acute liver failure, hepatic ischemia-reperfusion injury (IRI), alcoholic liver disease (ALD), viral hepatitis, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
- Dongwei Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Min Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Seogsong Jeong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yihan Qian
- School of Pharmacy, Fudan University, Shanghai, China
| | - Hailong Wu
- Shanghai Key Laboratory for Molecular Imaging, Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoni Kong
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
55
|
Sánchez-Martín P, Komatsu M. p62/SQSTM1 - steering the cell through health and disease. J Cell Sci 2018; 131:131/21/jcs222836. [PMID: 30397181 DOI: 10.1242/jcs.222836] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
SQSTM1 (also known as p62) is a multifunctional stress-inducible scaffold protein involved in diverse cellular processes. Its functions are tightly regulated through an extensive pattern of post-translational modifications, and include the isolation of cargos degraded by autophagy, induction of the antioxidant response by the Keap1-Nrf2 system, as well as the regulation of endosomal trafficking, apoptosis and inflammation. Accordingly, malfunction of SQSTM1 is associated with a wide range of diseases, including bone and muscle disorders, neurodegenerative and metabolic diseases, and multiple forms of cancer. In this Review, we summarize current knowledge regarding regulation, post-translational modifications and functions of SQSTM1, as well as how they are dysregulated in various pathogenic contexts.
Collapse
Affiliation(s)
- Pablo Sánchez-Martín
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata 951-8510, Japan .,Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
56
|
Matzinger M, Fischhuber K, Heiss EH. Activation of Nrf2 signaling by natural products-can it alleviate diabetes? Biotechnol Adv 2018; 36:1738-1767. [PMID: 29289692 PMCID: PMC5967606 DOI: 10.1016/j.biotechadv.2017.12.015] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/19/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (DM) has reached pandemic proportions and effective prevention strategies are wanted. Its onset is accompanied by cellular distress, the nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor boosting cytoprotective responses, and many phytochemicals activate Nrf2 signaling. Thus, Nrf2 activation by natural products could presumably alleviate DM. We summarize function, regulation and exogenous activation of Nrf2, as well as diabetes-linked and Nrf2-susceptible forms of cellular stress. The reported amelioration of insulin resistance, β-cell dysfunction and diabetic complications by activated Nrf2 as well as the status quo of Nrf2 in precision medicine for DM are reviewed.
Collapse
Affiliation(s)
- Manuel Matzinger
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Katrin Fischhuber
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria
| | - Elke H Heiss
- University of Vienna, Department of Pharmacognosy, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
57
|
Autophagy in Metabolic Age-Related Human Diseases. Cells 2018; 7:cells7100149. [PMID: 30249977 PMCID: PMC6210409 DOI: 10.3390/cells7100149] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 02/08/2023] Open
Abstract
Autophagy is a highly conserved homeostatic cellular mechanism that mediates the degradation of damaged organelles, protein aggregates, and invading pathogens through a lysosome-dependent pathway. Over the last few years, specific functions of autophagy have been discovered in many tissues and organs; however, abnormal upregulation or downregulation of autophagy has been depicted as an attribute of a variety of pathologic conditions. In this review, we will describe the current knowledge on the role of autophagy, from its regulation to its physiological influence, in metabolic age-related disorders. Finally, we propose to discuss the therapeutic potential of pharmacological and nutritional modulators of autophagy to treat metabolic diseases.
Collapse
|
58
|
Yu J, Li X, Matei N, McBride D, Tang J, Yan M, Zhang JH. Ezetimibe, a NPC1L1 inhibitor, attenuates neuronal apoptosis through AMPK dependent autophagy activation after MCAO in rats. Exp Neurol 2018; 307:12-23. [PMID: 29852178 DOI: 10.1016/j.expneurol.2018.05.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/22/2018] [Accepted: 05/27/2018] [Indexed: 12/25/2022]
Abstract
Autophagy activation exerts neuroprotective effects in the ischemic stroke model. Ezetimibe (Eze), a Niemann-Pick disease type C1-Like 1 (NPC1L1) pharmacological inhibitor, has been reported to protect hepatocytes from apoptosis via autophagy activation. In this study, we explored whether Eze could attenuate neuronal apoptosis in the rat model of middle cerebral artery occlusion (MCAO), specifically via activation of the AMPK/ULK1/autophagy pathway. Two hundred and one male Sprague-Dawley rats were subjected to transient MCAO followed by reperfusion. Eze was administered 1 h after MCAO. To elucidate the underlying molecular mechanism, Dorsomorphin, a selective AMPK inhibitor, and 3-methyladenine (3-MA), an autophagy inhibitor, were injected intracerebroventricularly before MCAO. Infarct volume, neurological score, brain cholesterol levels, immunofluorescence staining, Western blot, and Fluoro-Jade C (FJC) staining were used to evaluate the effects of Eze. The endogenous NPC1L1 expression increased and mainly expressed in neurons after MCAO. Intranasal administration of Eze reduced brain infarct volume at 24 and 72 h after MCAO, with improved short and long-term neurological functions after MCAO. Eze reduced brain cholesterol levels (total cholesterol, free cholesterol and cholesteryl esters) and the number of FJC-positive neurons. The expression of phosphorylated AMPK (p-AMPK) and downstream ULK1, Beclin1, LC3BII, Bcl-2, and Bcl-xl increased, while P62 and proapoptotic Bax decreased after treatment with Eze. Pretreatment with Dorsomorphin and 3-MA reversed the beneficial effects of Eze. These findings suggest that intranasal administration of Eze plays neuroprotective role through autophagy activation after MCAO in rats. Lowered cholesterol levels and AMPK activation may act in conjunction to induce autophagy after treatment with Eze. Eze merits further investigation as a potential therapeutic agent in ischemic stroke patients.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Xue Li
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China; Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Nathanael Matei
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Devin McBride
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA; The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jiping Tang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA
| | - Min Yan
- Department of Anesthesiology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, Zhejiang, China.
| | - John H Zhang
- Department of Anesthesiology and Basic Sciences, Loma Linda University, School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
59
|
Yamanashi Y, Takada T, Suzuki H. Associations between Lifestyle-Related Diseases and Transporters Involved in Intestinal Absorption and Biliary Excretion of Cholesterol. Biol Pharm Bull 2018; 41:1-10. [PMID: 29311470 DOI: 10.1248/bpb.b17-00690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Westernization of dietary habits leads to an increase in lipid intake and is thought to be responsible for an increase in patients with dyslipidemia. It is a well-known fact that the impaired cholesterol homeostasis is closely related to the development of various lifestyle-related diseases such as fatty liver, diabetes, and gallstone as well as dyslipidemia leading to atherosclerosis and cardiovascular diseases such as heart attack and stroke. Therefore, appropriate management of cholesterol levels in the body is considered important in prevention and treatments of these lifestyle-related diseases and in addition, molecular mechanisms controlling plasma (and/or hepatic) cholesterol levels have been intensively studied. Due to its hydrophobicity, cholesterol was long believed to pass through cell membranes by passive diffusion. However, recent studies have identified a number of plasma membrane transporters that are responsible for the cellular uptake or efflux of cholesterol and involved in developments of lifestyle-related diseases. In this review, we focus on Niemann-Pick C1 Like 1 (NPC1L1) and a heterodimer of ATP-binding cassette transporter G5 and G8 (ABCG5/G8), both of which are responsible for intestinal cholesterol absorption and biliary cholesterol secretion, and discuss the relationship between these cholesterol transporters and lifestyle-related diseases. In addition, we also discuss the related uncertainties that need to be explored in future studies.
Collapse
Affiliation(s)
- Yoshihide Yamanashi
- Department of Pharmacy, the University of Tokyo Hospital, Faculty of Medicine, the University of Tokyo
| | - Tappei Takada
- Department of Pharmacy, the University of Tokyo Hospital, Faculty of Medicine, the University of Tokyo
| | - Hiroshi Suzuki
- Department of Pharmacy, the University of Tokyo Hospital, Faculty of Medicine, the University of Tokyo
| |
Collapse
|
60
|
Sharma RS, Harrison DJ, Kisielewski D, Cassidy DM, McNeilly AD, Gallagher JR, Walsh SV, Honda T, McCrimmon RJ, Dinkova-Kostova AT, Ashford ML, Dillon JF, Hayes JD. Experimental Nonalcoholic Steatohepatitis and Liver Fibrosis Are Ameliorated by Pharmacologic Activation of Nrf2 (NF-E2 p45-Related Factor 2). Cell Mol Gastroenterol Hepatol 2018; 5:367-398. [PMID: 29552625 PMCID: PMC5852394 DOI: 10.1016/j.jcmgh.2017.11.016] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis (NASH) is associated with oxidative stress. We surmised that pharmacologic activation of NF-E2 p45-related factor 2 (Nrf2) using the acetylenic tricyclic bis(cyano enone) TBE-31 would suppress NASH because Nrf2 is a transcriptional master regulator of intracellular redox homeostasis. METHODS Nrf2+/+ and Nrf2-/- C57BL/6 mice were fed a high-fat plus fructose (HFFr) or regular chow diet for 16 weeks or 30 weeks, and then treated for the final 6 weeks, while still being fed the same HFFr or regular chow diets, with either TBE-31 or dimethyl sulfoxide vehicle control. Measures of whole-body glucose homeostasis, histologic assessment of liver, and biochemical and molecular measurements of steatosis, endoplasmic reticulum (ER) stress, inflammation, apoptosis, fibrosis, and oxidative stress were performed in livers from these animals. RESULTS TBE-31 treatment reversed insulin resistance in HFFr-fed wild-type mice, but not in HFFr-fed Nrf2-null mice. TBE-31 treatment of HFFr-fed wild-type mice substantially decreased liver steatosis and expression of lipid synthesis genes, while increasing hepatic expression of fatty acid oxidation and lipoprotein assembly genes. Also, TBE-31 treatment decreased ER stress, expression of inflammation genes, and markers of apoptosis, fibrosis, and oxidative stress in the livers of HFFr-fed wild-type mice. By comparison, TBE-31 did not decrease steatosis, ER stress, lipogenesis, inflammation, fibrosis, or oxidative stress in livers of HFFr-fed Nrf2-null mice. CONCLUSIONS Pharmacologic activation of Nrf2 in mice that had already been rendered obese and insulin resistant reversed insulin resistance, suppressed hepatic steatosis, and mitigated against NASH and liver fibrosis, effects that we principally attribute to inhibition of ER, inflammatory, and oxidative stress.
Collapse
Key Words
- ACACA, acetyl-CoA carboxylase alpha
- ACLY, ATP citrate lyase
- ACOT7, acetyl-CoA thioesterase 7
- ACOX2, acetyl-CoA oxidase 2
- ADRP, adipose differentiation-related protein
- AP-1, activator protein 1
- ATF4, activating transcription factor-4
- ATF6, activating transcription factor-6
- ApoB, apolipoprotein B
- BCL-2, B-cell lymphoma
- BIP, binding immunoglobulin protein
- C/EBP, CCAAT/enhancer-binding protein
- CAT, catalase
- CD36, cluster of differentiation 36
- CDDO, 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid
- CES1G, carboxylesterase 1g
- CHOP, C/EBP homologous protein
- COL1A1, collagen, type I, alpha-1
- COX2, cyclooxygenase-2
- CPT1A, carnitine palmitoyltransferase 1a
- ChREBP, carbohydrate-responsive element-binding protein
- DGAT2, diacylglycerol acyltransferase-2
- DMSO, dimethyl sulfoxide
- ER, endoplasmic reticulum
- FASN, fatty acid synthase
- FXR, farnesoid X receptor
- GCLC, glutamate-cysteine ligase catalytic
- GCLM, glutamate-cysteine ligase modifier
- GPX2, glutathione peroxidase-2
- GSH, reduced glutathione
- GSSG, oxidized glutathione
- GSTA4, glutathione S-transferase Alpha-4
- GSTM1, glutathione S-transferase Mu-1
- GTT, glucose tolerance test
- H&E, hematoxylin and eosin
- HF, high-fat
- HF30Fr, high-fat diet with 30% fructose in drinking water
- HF55Fr, high-fat diet with 55% fructose in drinking water
- HFFr, high-fat diet with fructose in drinking water
- HMOX1, heme oxygenase-1
- IKK, IκB kinase
- IRE1α, inositol requiring kinase-1α
- ITT, insulin tolerance test
- IκB, inhibitor of NF-κB
- JNK1, c-Jun N-terminal kinase 1
- Keap1, Kelch-like ECH-associated protein-1
- LXRα, liver X receptor α
- MCD, methionine- and choline-deficient
- MCP-1, monocyte chemotactic protein-1
- MGPAT, mitochondrial glycerol-3-phosphate acetyltransferase
- MPO, myeloperoxidase
- MTTP, microsomal triglyceride transfer protein
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NASH
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-κB
- NOS2, nitric oxide synthase-2
- NQO1, NAD(P)H:quinone oxidoreductase 1
- Nrf2
- Nrf2, NF-E2 p45-related factor 2
- PARP, poly ADP ribose polymerase
- PCR, polymerase chain reaction
- PDI, protein disulfide isomerase
- PERK, PRK-like endoplasmic reticulum kinase
- PPARα, peroxisome proliferator-activated receptor α
- PPARγ, peroxisome proliferator-activated receptor γ
- PRDX6, peroxiredoxin 6
- PTGR1, prostaglandin reductase-1
- PTT, pyruvate tolerance test
- RC, regular chow
- SCAD, short-chain acyl-CoA dehydrogenase
- SCD1, stearoyl-CoA desaturase-1
- SFN, sulforaphane
- SHP, small heterodimer partner
- SLC7A11, solute carrier family 7 member 11
- SREBP-1c, sterol regulatory element-binding protein-1c
- TBE-31
- TGFβ, transforming growth factor beta-1
- TNF-α, tumor necrosis factor-α
- TXN1, thioredoxin-1
- TXNRD1, thioredoxin reductase-1
- UPR, unfolded protein response
- XBP1, X-box binding protein-1
- eIf2α, eukaryotic translation initiation factor 2A
- p58IPK, p58 inhibitor of the PKR kinase
- qRT-PCR, quantitative reverse transcriptase PCR
- α-SMA, alpha smooth muscle actin
Collapse
Affiliation(s)
- Ritu S. Sharma
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - David J. Harrison
- School of Medicine, University of St Andrews, St Andrews, Scotland, United Kingdom
| | - Dorothy Kisielewski
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Diane M. Cassidy
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Alison D. McNeilly
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Jennifer R. Gallagher
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Shaun V. Walsh
- Department of Pathology, Ninewells Hospital and Medical School, Tayside NHS Trust, Dundee, Scotland, United Kingdom
| | - Tadashi Honda
- Department of Chemistry and Institute of Chemical Biology & Drug Discovery, Stony Brook University, Stony Brook, New York
| | - Rory J. McCrimmon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Albena T. Dinkova-Kostova
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - Michael L.J. Ashford
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - John F. Dillon
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| | - John D. Hayes
- Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, United Kingdom
| |
Collapse
|
61
|
Chen Y, Xu C, Huang R, Song J, Li D, Xia M. Butyrate from pectin fermentation inhibits intestinal cholesterol absorption and attenuates atherosclerosis in apolipoprotein E-deficient mice. J Nutr Biochem 2018; 56:175-182. [PMID: 29574331 DOI: 10.1016/j.jnutbio.2018.02.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/16/2018] [Accepted: 02/13/2018] [Indexed: 12/13/2022]
Abstract
Short-chain fatty acids (SCFAs), the major products of dietary fiber fermentation by intestinal microflora, exert beneficial effects on pathogenesis of multiple metabolic diseases. The aim of this study was to determine whether SCFAs from fermentation of pectin (PE), a soluble dietary fiber, prevent the development of atherosclerosis in apolipoprotein E-deficient (apoE-/-) mice. Male apoE-/- mice (8-week-old) were fed a high-fat, high-cholesterol diet (HCD; 21% wt/wt fat, 0.15% wt/wt cholesterol) or HCD supplemented with 20% wt/wt PE (HCD+PE) alone or with antibiotics (HCD+PE + A) in drinking water for 12 weeks. Serum lipids and SCFAs concentrations, atherosclerotic lesion area, and intestinal morphology and function were measured. Caco-2 cells were treated with SCFAs to determine whether they affected the expression of genes involved in cholesterol absorption. HCD+PE-treated mice exhibited decreased serum total and low-density lipoprotein cholesterol, and reduced atherosclerotic lesion area compared with HCD mice. These beneficial effects of PE were not observed in the HCD+PE+A group. Incubation of Caco-2 cells with butyrate, but not acetate and propionate, down-regulated the expression of Niemann-Pick C1-Like 1 but up-regulated the ATP-binding cassette transporters G5 and G8 (ABCG5 and G8) at the mRNA level. Butyrate treatment also increased transcriptional activity of liver X receptor in Caco-2 cells. Our data suggest that butyrate from PE intestinal fermentation protects mice from the progression of diet-induced atherosclerosis in apoE-/- mice. These findings suggest a novel mechanism by which dietary fiber may prevent the development of atherosclerosis.
Collapse
Affiliation(s)
- Ying Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, PR China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province 510080, PR China
| | - Chengfang Xu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province 510080, PR China
| | - Rong Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, PR China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province 510080, PR China
| | - Jiayi Song
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, PR China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province 510080, PR China
| | - Di Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, PR China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province 510080, PR China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, Guangdong Province 510080, PR China; Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province 510080, PR China.
| |
Collapse
|
62
|
Long M, Li X, Li L, Dodson M, Zhang DD, Zheng H. Multifunctional p62 Effects Underlie Diverse Metabolic Diseases. Trends Endocrinol Metab 2017; 28:818-830. [PMID: 28966079 DOI: 10.1016/j.tem.2017.09.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/03/2017] [Accepted: 09/05/2017] [Indexed: 12/17/2022]
Abstract
p62, a protein capable of binding both ubiquitin and autophagy substrates, is well established as a key regulator in cancer and neurodegenerative diseases. Recently, there has been accumulating evidence that p62 is also a pivotal regulator in metabolic diseases, such as obesity, T2DM, NAFLD, metabolic bone disease, gout and thyroid disease. This review summarizes the emerging role of p62 on these diseases by considering its functional domains, phenotypes in genetically modified animals, clinically observed alterations, and its effects on downstream metabolic signaling pathways. At the same time, we highlight the need to explore the roles played by p62 in the gastrointestinal environment and immune system, and the extent to which its elevated expression may confer protection against metabolic disorders.
Collapse
Affiliation(s)
- Min Long
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; These authors contributed equally to this work
| | - Xing Li
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; These authors contributed equally to this work
| | - Li Li
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China; These authors contributed equally to this work
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Hongting Zheng
- Department of Endocrinology, Translational Research Key Laboratory for Diabetes, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
63
|
Kim SH, Kim G, Han DH, Lee M, Kim I, Kim B, Kim KH, Song YM, Yoo JE, Wang HJ, Bae SH, Lee YH, Lee BW, Kang ES, Cha BS, Lee MS. Ezetimibe ameliorates steatohepatitis via AMP activated protein kinase-TFEB-mediated activation of autophagy and NLRP3 inflammasome inhibition. Autophagy 2017; 13:1767-1781. [PMID: 28933629 DOI: 10.1080/15548627.2017.1356977] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Impairment in macroautophagy/autophagy flux and inflammasome activation are common characteristics of nonalcoholic steatohepatitis (NASH). Considering the lack of approved agents for treating NASH, drugs that can enhance autophagy and modulate inflammasome pathways may be beneficial. Here, we investigated the novel mechanism of ezetimibe, a widely prescribed drug for hypercholesterolemia, as a therapeutic option for ameliorating NASH. Human liver samples with steatosis and NASH were analyzed. For in vitro studies of autophagy and inflammasomes, primary mouse hepatocytes, human hepatoma cells, mouse embryonic fibroblasts with Ampk or Tsc2 knockout, and human or primary mouse macrophages were treated with ezetimibe and palmitate. Steatohepatitis and fibrosis were induced by feeding Atg7 wild-type, haploinsufficient, and knockout mice a methionine- and choline-deficient diet with ezetimibe (10 mg/kg) for 4 wk. Human livers with steatosis or NASH presented impaired autophagy with decreased nuclear TFEB and increased SQSTM1, MAP1LC3-II, and NLRP3 expression. Ezetimibe increased autophagy flux and concomitantly ameliorated lipid accumulation and apoptosis in palmitate-exposed hepatocytes. Ezetimibe induced AMPK phosphorylation and subsequent TFEB nuclear translocation, related to MAPK/ERK. In macrophages, ezetimibe blocked the NLRP3 inflammasome-IL1B pathway in an autophagy-dependent manner and modulated hepatocyte-macrophage interaction via extracellular vesicles. Ezetimibe attenuated lipid accumulation, inflammation, and fibrosis in liver-specific Atg7 wild-type and haploinsufficient mice, but not in knockout mice. Ezetimibe ameliorates steatohepatitis by autophagy induction through AMPK activation and TFEB nuclear translocation, related to an independent MTOR ameliorative effect and the MAPK/ERK pathway. Ezetimibe dampens NLRP3 inflammasome activation in macrophages by modulating autophagy and a hepatocyte-driven exosome pathway.
Collapse
Affiliation(s)
- Soo Hyun Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Gyuri Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,b Department of Medicine, Samsung Medical Center , Sungkyunkwan University School of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea
| | - Dai Hoon Han
- d Department of Surgery , Yonsei University College of Medicine , Seoul , Korea
| | - Milim Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Irene Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Bohkyung Kim
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea
| | - Kook Hwan Kim
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Young-Mi Song
- f Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital , University of Toronto , Toronto , Canada
| | - Jeong Eun Yoo
- g Department of Pathology , Yonsei University College of Medicine , Seoul , Korea
| | - Hye Jin Wang
- h Department of Pharmacology , Yonsei University College of Medicine , Seoul , Korea
| | - Soo Han Bae
- e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| | - Yong-Ho Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Byung-Wan Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Eun Seok Kang
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Bong-Soo Cha
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,c Graduate School , Yonsei University College of Medicine , Seoul , Korea.,i Institute of Endocrine Research , Yonsei University College of Medicine , Seoul , Korea
| | - Myung-Shik Lee
- a Department of Internal Medicine , Yonsei University College of Medicine , Seoul , Korea.,e Severance Biomedical Science Institute, Yonsei Biomedical Research Institute , Yonsei University College of Medicine , Seoul , Korea
| |
Collapse
|
64
|
Yinchen Linggui Zhugan Decoction Ameliorates Nonalcoholic Fatty Liver Disease in Rats by Regulating the Nrf2/ARE Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:6178358. [PMID: 28932253 PMCID: PMC5592414 DOI: 10.1155/2017/6178358] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/11/2017] [Accepted: 07/30/2017] [Indexed: 12/13/2022]
Abstract
Yinchen Linggui Zhugan Decoction (YCLGZGD) is the combination of Linggui Zhugan (LGZGD) and Yinchenhao (YCHD) decoctions, two famous traditional Chinese medicine prescriptions. In previous studies, we found that Yinchen Linggui Zhugan Decoction (YCLGZGD) could regulate lipid metabolism disorder and attenuate inflammation in pathological process of nonalcoholic fatty liver disease (NAFLD). However, the exact underlying mechanism remains unknown. The aim of this study was to explore the effect of Yinchen Linggui Zhugan Decoction on experimental NAFLD and its mechanism in rats with high-fat diet (HFD) which was established by 8-week administration of HFD. YCLGZGD, LGZGD, and YCHD were administered daily for 4 weeks, after which the rats were euthanized. The level of blood lipid, liver enzymes, H&E, and Oil Red O staining were determined to evaluate NAFLD severity. Western blotting and real-time polymerase chain reaction were, respectively, used to determine hepatic protein and gene expression of Keap1, Nrf2, NQO1, and HO-1. Oral YCLGZGD ameliorated HFD-induced NAFLD. Furthermore, YCLGZGD increased the protein and gene expression of Nrf2, NQO1, and HO-1 without changing Keap1. Overall, these results suggest that YCLGZGD ameliorates HFD-induced NAFLD in rats by upregulating the Nrf2/ARE signaling pathway.
Collapse
|
65
|
Athyros VG, Alexandrides TK, Bilianou H, Cholongitas E, Doumas M, Ganotakis ES, Goudevenos J, Elisaf MS, Germanidis G, Giouleme O, Karagiannis A, Karvounis C, Katsiki N, Kotsis V, Kountouras J, Liberopoulos E, Pitsavos C, Polyzos S, Rallidis LS, Richter D, Tsapas AG, Tselepis AD, Tsioufis K, Tziomalos K, Tzotzas T, Vasiliadis TG, Vlachopoulos C, Mikhailidis DP, Mantzoros C. The use of statins alone, or in combination with pioglitazone and other drugs, for the treatment of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis and related cardiovascular risk. An Expert Panel Statement. Metabolism 2017; 71:17-32. [PMID: 28521870 DOI: 10.1016/j.metabol.2017.02.014] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/23/2017] [Accepted: 02/25/2017] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD), the most common liver disease, is characterized by accumulation of fat (>5% of the liver tissue), in the absence of alcohol abuse or other chronic liver diseases. It is closely related to the epidemic of obesity, metabolic syndrome or type 2 diabetes mellitus (T2DM). NAFLD can cause liver inflammation and progress to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis or hepatocellular cancer (HCC). Nevertheless, cardiovascular disease (CVD) is the most common cause of death in NAFLD/NASH patients. Current guidelines suggest the use of pioglitazone both in patients with T2DM and in those without. The use of statins, though considered safe by the guidelines, have very limited use; only 10% in high CVD risk patients are on statins by tertiary centers in the US. There are data from several animal studies, 5 post hoc analyses of prospective long-term survival studies, and 5 rather small biopsy proven NASH studies, one at baseline and on at the end of the study. All these studies provide data for biochemical and histological improvement of NAFLD/NASH with statins and in the clinical studies large reductions in CVD events in comparison with those also on statins and normal liver. Ezetimibe was also reported to improve NAFLD. Drugs currently in clinical trials seem to have potential for slowing down the evolution of NAFLD and for reducing liver- and CVD-related morbidity and mortality, but it will take time before they are ready to be used in everyday clinical practice. The suggestion of this Expert Panel is that, pending forthcoming randomized clinical trials, physicians should consider using a PPARgamma agonist, such as pioglitazone, or, statin use in those with NAFLD/NASH at high CVD or HCC risk, alone and/or preferably in combination with each other or with ezetimibe, for the primary or secondary prevention of CVD, and the avoidance of cirrhosis, liver transplantation or HCC, bearing in mind that CVD is the main cause of death in NAFLD/NASH patients.
Collapse
Affiliation(s)
- Vasilios G Athyros
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece.
| | - Theodore K Alexandrides
- Department of Internal Medicine, Division of Endocrinology, University of Patras Medical School, Patras, Greece
| | - Helen Bilianou
- Lipid Clinic, Cardiology Department, Tzaneio Hospital, Piraeus, Greece
| | - Evangelos Cholongitas
- 4th Prop. Department of Internal Medicine, Hippocration Hospital, Division of Gastroenterology and Hepatology, Medical School of Aristotle University Thessaloniki, Greece
| | - Michael Doumas
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece
| | - Emmanuel S Ganotakis
- Department of Internal Medicine University Hospital of Crete, University of Crete Medical School, Heraklion, Greece
| | - John Goudevenos
- Department of Cardiology Medical School, University Hospital of Ioannina, Ioannina, Greece
| | - Moses S Elisaf
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Georgios Germanidis
- 1st Department of Internal Medicine, Gastroenterology and Hepatology Section, AHEPA Hospital, Aristotle University Medical School, Thessaloniki, Greece
| | - Olga Giouleme
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece
| | - Asterios Karagiannis
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece
| | - Charalambos Karvounis
- First Cardiology Department, AHEPA Hospital, Medical School, Aristotle University Thessaloniki, Greece
| | - Niki Katsiki
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece
| | - Vasilios Kotsis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University Thessaloniki, Greece
| | - Jannis Kountouras
- 2nd Prop. Department of Internal Medicine, Hippocration Hospital, Medical School of Aristotle University Thessaloniki, Greece
| | - Evangelos Liberopoulos
- Department of Internal Medicine, School of Health Sciences, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Christos Pitsavos
- 1st Cardiology Clinic, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stergios Polyzos
- 2nd Prop. Propedeutic Department of Internal Medicine, Hippocration Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Loukianos S Rallidis
- 2nd Department of Cardiology, University General Hospital Attikon, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Apostolos G Tsapas
- 2nd Department of Internal Medicine-Diabetology, Hippocration Hospital, Aristotle University Thessaloniki, Medical School, Thessaloniki, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Clinic, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Tziomalos
- 1st Prop. Department of Internal Medicine, AHEPA Hospital, Aristotle University Medical School, Thessaloniki, Greece
| | | | - Themistoklis G Vasiliadis
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University Thessaloniki, Greece
| | - Charalambos Vlachopoulos
- 1st Cardiology Clinic, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital Campus, University College London Medical School, University College London (UCL), London, UK
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
66
|
Qiu P, Dong Y, Li B, Kang XJ, Gu C, Zhu T, Luo YY, Pang MX, Du WF, Ge WH. Dihydromyricetin modulates p62 and autophagy crosstalk with the Keap-1/Nrf2 pathway to alleviate ethanol-induced hepatic injury. Toxicol Lett 2017; 274:31-41. [PMID: 28419832 DOI: 10.1016/j.toxlet.2017.04.009] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 03/31/2017] [Accepted: 04/13/2017] [Indexed: 02/07/2023]
Abstract
Increasing evidence has demonstrated that dihydromyricetin (DMY) contains highly effective antioxidative, anti-inflammatory, anti-microbial and anti-diabetic properties. Nevertheless, the underlying hepatoprotective mechanisms of DMY have infrequently been reported thus far. In the present study, C57BL/6 mice were fed with the Lieber-DeCarli diet containing alcohol or isocaloric maltose dextrin as a control diet with or without DMY (75 and 150mg/kg/d bw) for 6 weeks. DMY significantly attenuated hepatic enzyme release, hepatic lipid peroxidation and triglyceride deposition induced by chronic alcohol exposure. In addition, DMY dramatically attenuated the alcohol-triggered elevation of the level of inflammatory cytokines and partially recovered hepatic pathological changes. Notably, DMY remarkably modified aberrant expression of CYP2E1, Keap-1 and HO-1 in the liver and simultaneously ameliorated disordered nuclear localization of NF-κB and Nrf2 to exert its hepatoprotective effects. Further mechanistic exploration suggested that DMY activated Nrf2, possibly mediated through the autophagy pathway. Analysis of the crosstalk among p62, Keap-1 and Nrf2 demonstrated that the p62 upregulation caused by DMY contributes to a positive feedback loop in Nrf2 activation. In summary, DMY likely modulates p62 and autophagy crosstalk with the Keap-1/Nrf2 pathway to alleviate liver steatosis and the inflammatory response in the pathological progression of ALD.
Collapse
Affiliation(s)
- Ping Qiu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Yu Dong
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, Zhejiang Province 310007, China
| | - Bo Li
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Xian-Jie Kang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Chao Gu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Tao Zhu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Yun-Yun Luo
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Min-Xia Pang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Wei-Feng Du
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China
| | - Wei-Hong Ge
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province 310053, China.
| |
Collapse
|
67
|
Lee DH, Park JS, Lee YS, Sung SH, Lee YH, Bae SH. The hypertension drug, verapamil, activates Nrf2 by promoting p62-dependent autophagic Keap1 degradation and prevents acetaminophen-induced cytotoxicity. BMB Rep 2017; 50:91-96. [PMID: 27998394 PMCID: PMC5342872 DOI: 10.5483/bmbrep.2017.50.2.188] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Indexed: 12/19/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) provides a cellular defense against oxidative stress by inducing the expression of antioxidant and detoxification enzymes. The calcium antagonist, verapamil, is an FDA-approved drug prescribed for the treatment of hypertension. Here, we show that verapamil acts as a potent Nrf2 activator without causing cytotoxicity, through degradation of Kelch-like ECH-associated protein 1 (Keap1), a Nrf2 repressor. Furthermore, verapamil-induced Keap1 degradation is prominently mediated by a p62-dependent autophagic pathway. Correspondingly, verapamil protects cells from acetaminophen-induced oxidative damage through Nrf2 activation. These results demonstrated the underlying mechanisms for the protective role of verapamil against acetaminophen-induced cytotoxicity.
Collapse
Affiliation(s)
- Da Hyun Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine; Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul 03722, Korea
| | - Jeong Su Park
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yu Seol Lee
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Su Haeng Sung
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Yong-Ho Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Soo Han Bae
- Severance Biomedical Science Institute, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|