101
|
Oppedisano F, Muscoli C, Musolino V, Carresi C, Macrì R, Giancotta C, Bosco F, Maiuolo J, Scarano F, Paone S, Nucera S, Zito MC, Scicchitano M, Ruga S, Ragusa M, Palma E, Tavernese A, Mollace R, Bombardelli E, Mollace V. The Protective Effect of Cynara Cardunculus Extract in Diet-Induced NAFLD: Involvement of OCTN1 and OCTN2 Transporter Subfamily. Nutrients 2020; 12:E1435. [PMID: 32429274 PMCID: PMC7284543 DOI: 10.3390/nu12051435] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hyperlipidemia and insulin-resistance are often associated with Non-Alcoholic Fatty Liver Disease (NAFLD) thereby representing a true issue worldwide due to increased risk of developing cardiovascular and systemic disorders. Although clear evidence suggests that circulating fatty acids contribute to pathophysiological mechanisms underlying NAFLD and hyperlipidemia, further studies are required to better identify potential beneficial approaches for counteracting such a disease. Recently, several artichoke extracts have been used for both reducing hyperlipidemia, insulin-resistance and NAFLD, though the mechanism is unclear. Here we used a wild type of Cynara Cardunculus extract (CyC), rich in sesquiterpens and antioxidant active ingredients, in rats fed a High Fat Diet (HFD) compared to a Normal Fat Diet (NFD). In particular, in rats fed HFD for four consecutive weeks, we found a significant increase of serum cholesterol, triglyceride and serum glucose. This effect was accompanied by increased body weight and by histopathological features of liver steatosis. The alterations of metabolic parameters found in HFDs were antagonised dose-dependently by daily oral supplementation of rats with CyC 10 and 20 mg/kg over four weeks, an effect associated to significant improvement of liver steatosis. The effect of CyC (20 mg/kg) was also associated to enhanced expression of both OCTN1 and OCTN2 carnitine-linked transporters. Thus, present data suggest a contribution of carnitine system in the protective effect of CyC in diet-induced hyperlipidemia, insulin-resistance and NAFLD.
Collapse
Affiliation(s)
- Francesca Oppedisano
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Carolina Muscoli
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
- San Raffaele IRCCS, 00199 Rome, Italy
| | - Vincenzo Musolino
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Cristina Carresi
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Roberta Macrì
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Caterina Giancotta
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Francesca Bosco
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Jessica Maiuolo
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Federica Scarano
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Sara Paone
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Saverio Nucera
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Maria Caterina Zito
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Stefano Ruga
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Monica Ragusa
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Ernesto Palma
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Annamaria Tavernese
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Rocco Mollace
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Ezio Bombardelli
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health, Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (F.O.); (C.M.); (V.M.); (C.C.); (R.M.); (C.G.); (F.B.); (J.M.); (F.S.); (S.P.); (S.N.); (M.C.Z.); (M.S.); (S.R.); (M.R.); (E.P.); (A.T.); (R.M.); (E.B.)
- San Raffaele IRCCS, 00199 Rome, Italy
| |
Collapse
|
102
|
Raigani S, Carroll C, Griffith S, Pendexter C, Rosales I, Deirawan H, Beydoun R, Yarmush M, Uygun K, Yeh H. Improvement of steatotic rat liver function with a defatting cocktail during ex situ normothermic machine perfusion is not directly related to liver fat content. PLoS One 2020; 15:e0232886. [PMID: 32396553 PMCID: PMC7217452 DOI: 10.1371/journal.pone.0232886] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
There is a significant organ shortage in the field of liver transplantation, partly due to a high discard rate of steatotic livers from donors. These organs are known to function poorly if transplanted but make up a significant portion of the available pool of donated livers. This study demonstrates the ability to improve the function of steatotic rat livers using a combination of ex situ machine perfusion and a "defatting" drug cocktail. After 6 hours of perfusion, defatted livers demonstrated lower perfusate lactate levels and improved bile quality as demonstrated by higher bile bicarbonate and lower bile lactate. Furthermore, defatting was associated with decreased gene expression of pro-inflammatory cytokines and increased expression of enzymes involved in mitochondrial fatty acid oxidation. Rehabilitation of marginal or discarded steatotic livers using machine perfusion and tailored drug therapy can significantly increase the supply of donor livers for transplantation.
Collapse
Affiliation(s)
- Siavash Raigani
- Division of Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Cailah Carroll
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Stephanie Griffith
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Casie Pendexter
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Ivy Rosales
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Hany Deirawan
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Rafic Beydoun
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Martin Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, United States of America
| | - Korkut Uygun
- Division of Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospital for Children, Boston, Massachusetts, United States of America
| | - Heidi Yeh
- Division of Transplant Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
103
|
Hu X, Wang X, Jia F, Tanaka N, Kimura T, Nakajima T, Sato Y, Moriya K, Koike K, Gonzalez FJ, Nakayama J, Aoyama T. A trans-fatty acid-rich diet promotes liver tumorigenesis in HCV core gene transgenic mice. Carcinogenesis 2020; 41:159-170. [PMID: 31300810 PMCID: PMC8456504 DOI: 10.1093/carcin/bgz132] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 06/08/2019] [Accepted: 07/11/2019] [Indexed: 12/19/2022] Open
Abstract
Excess consumption of trans-fatty acid (TFA), an unsaturated fatty acid containing trans double bonds, is a major risk factor for cardiovascular disease and metabolic syndrome. However, little is known about the link between TFA and hepatocellular carcinoma (HCC) despite it being a frequent form of cancer in humans. In this study, the impact of excessive dietary TFA on hepatic tumorigenesis was assessed using hepatitis C virus (HCV) core gene transgenic mice that spontaneously developed HCC. Male transgenic mice were treated for 5 months with either a control diet or an isocaloric TFA-rich diet that replaced the majority of soybean oil with shortening. The prevalence of liver tumors was significantly higher in TFA-rich diet-fed transgenic mice compared with control diet-fed transgenic mice. The TFA-rich diet significantly increased the expression of pro-inflammatory cytokines, as well as oxidative and endoplasmic reticulum stress, and activated nuclear factor-kappa B (NF-κB) and nuclear factor erythroid 2-related factor 2 (NRF2), leading to high p62/sequestosome 1 (SQSTM1) expression. Furthermore, the TFA diet activated extracellular signal-regulated kinase (ERK) and stimulated the Wnt/β-catenin signaling pathway, synergistically upregulating cyclin D1 and c-Myc, driving cell proliferation. Excess TFA intake also promoted fibrogenesis and ductular reaction, presumably contributing to accelerated liver tumorigenesis. In conclusion, these results demonstrate that a TFA-rich diet promotes hepatic tumorigenesis, mainly due to persistent activation of NF-κB and NRF2-p62/SQSTM1 signaling, ERK and Wnt/β-catenin pathways and fibrogenesis. Therefore, HCV-infected patients should avoid a TFA-rich diet to prevent liver tumor development.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xiaojing Wang
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
- Department of Gastroenterology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, People’s Republic of China
| | - Fangping Jia
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
- Research Center for Social Systems, Shinshu University, Matsumoto, Japan
| | - Takefumi Kimura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoshiko Sato
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kyoji Moriya
- Department of Infection Control and Prevention, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
104
|
Wikan N, Tocharus J, Sivasinprasasn S, Kongkaew A, Chaichompoo W, Suksamrarn A, Tocharus C. Capsaicinoid nonivamide improves nonalcoholic fatty liver disease in rats fed a high-fat diet. J Pharmacol Sci 2020; 143:188-198. [PMID: 32414691 DOI: 10.1016/j.jphs.2020.03.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/09/2020] [Accepted: 03/30/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic disease that causes morbidity associated with metabolic syndrome. NAFLD is a worldwide problem and represents a major cause of liver injury, which can lead to liver cell death. We investigated the effects of nonivamide (pelargonic acid vanillylamide, PAVA; 1 mg/kg) and rosuvastatin (RSV; 10 mg/kg) on hepatic steatosis induced by a high-fat diet (HFD). Male Sprague-Dawley rats were fed a HFD for 16 weeks then received PAVA or RSV for 4 additional weeks. We examined the metabolic parameters, function, fat content, histological alterations, reactive oxygen species production, and apoptotic cell death of the liver, in addition to the expression of the following important molecules: transient receptor potential cation channel subfamily V member 1 (TRPV1) phosphorylation of sterol regulatory element binding protein (pSREBP-1c/SREBP-1c), total and membrane glucose transporter 2 (GLUT2), 4-hydroxynonenal (4-HNE), and cleaved caspase-3. HFD-induced hepatic steatosis was associated with significantly increased morphological disorganization, injury markers, oxidative stress, lipid peroxidation, and apoptosis. However, metabolic dysfunction and hepatic injury were reduced by RSV and PAVA treatment. PAVA regulated lipid deposition, improved insulin resistance, and decreased oxidative stress and apoptotic cell death. Therefore, PAVA represents a promising therapeutic approach for treating metabolic disorders in patients with NAFLD.
Collapse
Affiliation(s)
- Naruemon Wikan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Aphisek Kongkaew
- Research Administration Section, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Waraluck Chaichompoo
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
105
|
Kim K, Kim KH. Targeting of Secretory Proteins as a Therapeutic Strategy for Treatment of Nonalcoholic Steatohepatitis (NASH). Int J Mol Sci 2020; 21:ijms21072296. [PMID: 32225108 PMCID: PMC7177791 DOI: 10.3390/ijms21072296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is defined as a progressive form of nonalcoholic fatty liver disease (NAFLD) and is a common chronic liver disease that causes significant worldwide morbidity and mortality, and has no approved pharmacotherapy. Nevertheless, growing understanding of the molecular mechanisms underlying the development and progression of NASH has suggested multiple potential therapeutic targets and strategies to treat this disease. Here, we review this progress, with emphasis on the functional role of secretory proteins in the development and progression of NASH, in addition to the change of expression of various secretory proteins in mouse NASH models and human NASH subjects. We also highlight secretory protein-based therapeutic approaches that influence obesity-associated insulin resistance, liver steatosis, inflammation, and fibrosis, as well as the gut–liver and adipose–liver axes in the treatment of NASH.
Collapse
Affiliation(s)
- Kyeongjin Kim
- Department of Biomedical Sciences, College of Medicine, Inha University, Inha-ro 100, Michuhol-gu, Incheon 22212, Korea
- Correspondence: (K.K.); (K.H.K.)
| | - Kook Hwan Kim
- Metabolic Diseases Research Center, GI Cell, Inc., B-1014, Tera Tower, Songpa-daero 167, Songpa-gu, Seoul 05855, Korea
- Correspondence: (K.K.); (K.H.K.)
| |
Collapse
|
106
|
Wang Y, Nakajima T, Gonzalez FJ, Tanaka N. PPARs as Metabolic Regulators in the Liver: Lessons from Liver-Specific PPAR-Null Mice. Int J Mol Sci 2020; 21:2061. [PMID: 32192216 PMCID: PMC7139552 DOI: 10.3390/ijms21062061] [Citation(s) in RCA: 318] [Impact Index Per Article: 63.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 12/12/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) α, β/δ, and γ modulate lipid homeostasis. PPARα regulates lipid metabolism in the liver, the organ that largely controls whole-body nutrient/energy homeostasis, and its abnormalities may lead to hepatic steatosis, steatohepatitis, steatofibrosis, and liver cancer. PPARβ/δ promotes fatty acid β-oxidation largely in extrahepatic organs, and PPARγ stores triacylglycerol in adipocytes. Investigations using liver-specific PPAR-disrupted mice have revealed major but distinct contributions of the three PPARs in the liver. This review summarizes the findings of liver-specific PPAR-null mice and discusses the role of PPARs in the liver.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan; (Y.W.); (T.N.)
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan; (Y.W.); (T.N.)
| | - Frank J. Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Nagano 390-8621, Japan; (Y.W.); (T.N.)
- Research Center for Social Systems, Shinshu University, Matsumoto, Nagano 390-8621, Japan
| |
Collapse
|
107
|
Ocker M. Challenges and opportunities in drug development for nonalcoholic steatohepatitis. Eur J Pharmacol 2020; 870:172913. [PMID: 31926994 DOI: 10.1016/j.ejphar.2020.172913] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/04/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) are considered major global medical burdens with high prevalence and steeply rising incidence. Despite the characterization of numerous pathophysiologic pathways leading to metabolic disorder, lipid accumulation, inflammation, fibrosis, and ultimately end-stage liver disease or liver cancer formation, so far no causal pharmacological therapy is available. Drug development for NAFLD and NASH is limited by long disease duration and slow progression and the need for sequential biopsies to monitor the disease stage. Additional non-invasive biomarkers could therefore improve design and feasibility of such. Here, the current concepts on preclinical models, biomarkers and clinical endpoints and trial designs are briefly reviewed.
Collapse
|
108
|
Toporova L, Balaguer P. Nuclear receptors are the major targets of endocrine disrupting chemicals. Mol Cell Endocrinol 2020; 502:110665. [PMID: 31760044 DOI: 10.1016/j.mce.2019.110665] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
Abstract
Endocrine disrupting chemicals (EDCs) are exogenous substances that are suspected to cause adverse effects in the endocrine system mainly by acting through their interaction with nuclear receptors such as the estrogen receptors α and β (ERα and ERβ), the androgen receptor (AR), the pregnan X receptor (PXR), the peroxisome proliferator activated receptors α and γ (PPARα, PPARγ) and the thyroid receptors α and β (TRα and TRβ). More recently, the retinoid X receptors (RXRα, RXRβ and RXRγ), the constitutive androstane receptor (CAR) and the estrogen related receptor γ (ERRγ) have also been identified as targets of EDCs. Finally, nuclear receptors still poorly studied for their interaction with environmental ligands such as the progesterone receptor (PR), the mineralocorticoid receptor (MR), the glucocorticoid receptor (GR), the retinoic acid receptors (RAR α, RARβ and RARγ), the farnesoid X receptor (FXR) and the liver X receptors α and β (LXRα and LXβ) as well are suspected targets of EDCs. Humans are generally exposed to low doses of pollutants, therefore the aim of current research is to identify the targets of EDCs at environmental concentrations. In this review, we analyze recent works referring that nuclear receptors are targets of EDCs and we highlight which EDCs are able to act at low concentrations.
Collapse
Affiliation(s)
- Lucia Toporova
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| | - Patrick Balaguer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, ICM, Univ Montpellier, 34090, Montpellier, France.
| |
Collapse
|
109
|
Liu CG, Han YH, Kankala RK, Wang SB, Chen AZ. Subcellular Performance of Nanoparticles in Cancer Therapy. Int J Nanomedicine 2020; 15:675-704. [PMID: 32103936 PMCID: PMC7008395 DOI: 10.2147/ijn.s226186] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
With the advent of nanotechnology, various modes of traditional treatment strategies have been transformed extensively owing to the advantageous morphological, physiochemical, and functional attributes of nano-sized materials, which are of particular interest in diverse biomedical applications, such as diagnostics, sensing, imaging, and drug delivery. Despite their success in delivering therapeutic agents, several traditional nanocarriers often end up with deprived selectivity and undesired therapeutic outcome, which significantly limit their clinical applicability. Further advancements in terms of improved selectivity to exhibit desired therapeutic outcome toward ablating cancer cells have been predominantly made focusing on the precise entry of nanoparticles into tumor cells via targeting ligands, and subsequent delivery of therapeutic cargo in response to specific biological or external stimuli. However, there is enough room intracellularly, where diverse small-sized nanomaterials can accumulate and significantly exert potentially specific mechanisms of antitumor effects toward activation of precise cancer cell death pathways that can be explored. In this review, we aim to summarize the intracellular pathways of nanoparticles, highlighting the principles and state of their destructive effects in the subcellular structures as well as the current limitations of conventional therapeutic approaches. Next, we give an overview of subcellular performances and the fate of internalized nanoparticles under various organelle circumstances, particularly endosome or lysosome, mitochondria, nucleus, endoplasmic reticulum, and Golgi apparatus, by comprehensively emphasizing the unique mechanisms with a series of interesting reports. Moreover, intracellular transformation of the internalized nanoparticles, prominent outcome and potential affluence of these interdependent subcellular components in cancer therapy are emphasized. Finally, we conclude with perspectives with a focus on the contemporary challenges in their clinical applicability.
Collapse
Affiliation(s)
- Chen-Guang Liu
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
| | - Ya-Hui Han
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
| | - Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian361021, People’s Republic of China
| | - Shi-Bin Wang
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian361021, People’s Republic of China
| | - Ai-Zheng Chen
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- College of Chemical Engineering, Huaqiao University, Xiamen, Fujian361021, People’s Republic of China
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen, Fujian361021, People’s Republic of China
| |
Collapse
|
110
|
Fujimori N, Tanaka N, Kimura T, Sano K, Horiuchi A, Kato N, Takahashi Y, Kuribayashi N, Sugiura A, Yamazaki T, Joshita S, Umemura T, Matsumoto A, Tanaka E. Long-term luseogliflozin therapy improves histological activity of non-alcoholic steatohepatitis accompanied by type 2 diabetes mellitus. Clin J Gastroenterol 2020; 13:83-89. [PMID: 31292843 DOI: 10.1007/s12328-019-01018-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
A 60-year-old Japanese woman was referred to our hospital for further examination of persistent liver dysfunction. She had been suffering from type 2 diabetes mellitus since the age of 50 years. Her hemoglobin A1c (HbA1c) value was as high as 7.8% despite treatment with dipeptidyl peptidase-4 inhibitor, metformin, and sulfonylurea. After excluding viral hepatitis, alcohol or drug-induced liver injury, and autoimmune liver diseases, liver histology evidence of macrovesicular steatosis, hepatocyte ballooning, and pericellular fibrosis confirmed a diagnosis of non-alcoholic steatohepatitis (NASH). Luseogliflozin (2.5 mg/day), a sodium-glucose cotransporter 2 inhibitor (SGLT2I), was co-administered to strengthen glycemic control. Liver enzymes and HbA1c gradually improved without any adverse events. A second liver biopsy at 15 months after luseogliflozin commencement revealed improvements in steatosis, fibrosis, and overall histological activity score. This case demonstrates that long-term luseogliflozin may be a good therapeutic option for diabetic NAFLD/NASH patients. The merits of persistent SGLT2I administration for NAFLD/NASH patients warrant validation in future studies.
Collapse
Affiliation(s)
- Naoyuki Fujimori
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Asahi 3-1-1, Matsumoto, Nagano, 390-8621, Japan.
- Research Center for Social Systems, Shinshu University, Matsumoto, Japan.
| | - Takefumi Kimura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kenji Sano
- Department of Pathology, Iida Municipal Hospital, Iida, Japan
| | - Akira Horiuchi
- Digestive Disease Center, Showa Inan General Hospital, Komagane, Japan
| | | | - Yoshiyuki Takahashi
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoya Kuribayashi
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Ayumi Sugiura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tomoo Yamazaki
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Satoru Joshita
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeji Umemura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akihiro Matsumoto
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Eiji Tanaka
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
111
|
Wang P, Shao X, Bao Y, Zhu J, Chen L, Zhang L, Ma X, Zhong XB. Impact of obese levels on the hepatic expression of nuclear receptors and drug-metabolizing enzymes in adult and offspring mice. Acta Pharm Sin B 2020; 10:171-185. [PMID: 31993314 PMCID: PMC6976990 DOI: 10.1016/j.apsb.2019.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/30/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
The prevalence of obesity-associated conditions raises new challenges in clinical medication. Although altered expression of drug-metabolizing enzymes (DMEs) has been shown in obesity, the impacts of obese levels (overweight, obesity, and severe obesity) on the expression of DMEs have not been elucidated. Especially, limited information is available on whether parental obese levels affect ontogenic expression of DMEs in children. Here, a high-fat diet (HFD) and three feeding durations were used to mimic different obese levels in C57BL/6 mice. The hepatic expression of five nuclear receptors (NRs) and nine DMEs was examined. In general, a trend of induced expression of NRs and DMEs (except for Cyp2c29 and 3a11) was observed in HFD groups compared to low-fat diet (LFD) groups. Differential effects of HFD on the hepatic expression of DMEs were found in adult mice at different obese levels. Family-based dietary style of an HFD altered the ontogenic expression of DMEs in the offspring older than 15 days. Furthermore, obese levels of parental mice affected the hepatic expression of DMEs in offspring. Overall, the results indicate that obese levels affected expression of the DMEs in adult individuals and that of their children. Drug dosage might need to be optimized based on the obese levels.
Collapse
Key Words
- 18-HA, adult mice fed with 18 weeks HFD
- 18-LA, adult mice fed with 18 weeks LFD
- 4-HA, adult mice fed with 4 weeks HFD
- 4-LA, adult mice fed with 4 weeks LFD
- 7-ER, 7-ethoxyresorufin
- 8-HA, adult mice fed with 8 weeks HFD
- 8-LA, adult mice fed with 8 weeks LFD
- AhR, aryl hydrocarbon receptor
- BMI, body mass index
- CAR, constitutive androstane receptor
- CHZ, chlorzoxazone
- CYP2E1, cytochrome P450 2E1
- DIO, diet-induced obesity
- DMEs, drug-metabolizing enzymes
- Diet-induced obesity
- Drug-metabolizing enzymes
- EFV, efavirenz
- Gapdh, glyceraldehyde-3-phosphate dehydrogenase
- HFD, high-fat diet
- HNF4α, hepatocyte nuclear factor 4 alpha
- High-fat diet
- LFD, low-fat diet
- MDZ, midazolam
- MPA, mobile phase A
- MPB, mobile phase B
- NADPH, nicotinamide adenine dinucleotide phosphate
- NAFLD, non-alcoholic fatty liver disease
- NRs, nuclear receptors
- Nuclear receptors
- O-18-HA, offspring from parental mice fed with 18 weeks HFD
- O-18-LA, offspring from parental mice fed with 18 weeks LFD
- O-4-HA, offspring from parental mice fed with 4 weeks HFD
- O-4-LA, offspring from parental mice fed with 4 weeks LFD
- O-8-HA, offspring from parental mice fed with 8 weeks HFD
- O-8-LA, offspring from parental mice fed with 8 weeks LFD
- Ontogenic expression
- Overweight
- PBS, phosphate-buffered saline
- PPARα, peroxisome proliferator-activated receptor alpha
- PXR, pregnane X receptor
- RSF, resorufin
- RT-qPCR, real-time quantitative PCR
- SD, standard deviation
- SULT1A1, sulfotransferase 1A1
- UGT1A1, uridine diphosphate glucuronosyltransferase 1A1
Collapse
Affiliation(s)
- Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Xueyan Shao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Yifan Bao
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Junjie Zhu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Liming Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaochao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiao-bo Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
112
|
Baandrup Kristiansen MN, Veidal SS, Christoffersen C, Feigh M, Vrang N, Roth JD, Erickson M, Adorini L, Jelsing J. Validity of biopsy-based drug effects in a diet-induced obese mouse model of biopsy-confirmed NASH. BMC Gastroenterol 2019; 19:228. [PMID: 31883514 PMCID: PMC6935483 DOI: 10.1186/s12876-019-1149-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background Compounds in clinical development for nonalcoholic steatohepatitis (NASH) improve liver histopathology in diet-induced obese mouse models of biopsy-confirmed NASH. Since the biopsy section used for histopathological evaluation represents only < 1% of the whole mouse liver, we evaluated how well biopsy-based quantitative image analyses correlate to stereology-based whole-liver quantitative changes upon drug treatment. Methods Male leptin-deficient Lepob/Lepob mice were fed the Amylin liver NASH (AMLN) diet for 16 weeks before stratification into treatment groups using a biopsy-based evaluation of type I collagen αI (col1a1) levels. Mice were treated for 8 weeks with either vehicle (PO, QD), liraglutide (0.4 mg/kg, SC, QD), elafibranor (30 mg/kg, PO, QD) or INT-767 (10 mg/kg, PO, QD). Terminal quantitative histological assessment of liver lipid (hematoxylin-eosin staining), inflammation (galectin-3 immunohistochemistry (IHC); gal-3), and fibrosis (col1a1 IHC) was performed on terminal liver biopsies and compared with stereologically sampled serial sections spanning the medial, left and right lateral lobe of the liver. Results The distribution of liver lipid and fibrosis was markedly consistent across lobes, whereas inflammation showed some variability. While INT-767 and liraglutide significantly reduced total liver weight by 20 and 48%, respectively, elafibranor tended to exacerbate hepatomegaly in Lepob/Lepob-NASH mice. All three compounds markedly reduced biopsy-based relative liver lipid content. Elafibranor and INT-767 significantly reduced biopsy-based relative gal-3 levels (P < 0.001), whereas INT-767 and liraglutide tended to reduce relative col1a1 levels. When changes in liver weight was accounted for, both INT-767 and liraglutide significantly reduced biopsy-based total col1a1 content. Although minor differences in absolute and relative liver lipid, inflammation and fibrosis levels were observed across lobes, the interpretation of drug-induced effects were consistent with biopsy-based conclusions. Notably, the incorporation of changes in total liver mass revealed that liraglutide’s efficacy reached statistical significances for all analyzed parameters. Conclusions In conclusion, in-depth analyses of liver homogeneity demonstrated that drug-induced improvement in liver biopsy-assessed histopathology is representative for overall liver effects assessed using stereology. Importantly, these findings reveal how changes in whole-liver mass should be considered to provide a deeper understanding of apparent drug treatment efficacy in preclinical NASH studies.
Collapse
Affiliation(s)
- Maria Nicoline Baandrup Kristiansen
- Gubra Aps, Hoersholm, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Christina Christoffersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Bispebjerg Hospital and Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Mary Erickson
- Intercept Pharmaceuticals, San Diego, California, USA
| | | | | |
Collapse
|
113
|
Boeckmans J, Natale A, Rombaut M, Buyl K, Rogiers V, De Kock J, Vanhaecke T, Rodrigues RM. Anti-NASH Drug Development Hitches a Lift on PPAR Agonism. Cells 2019; 9:E37. [PMID: 31877771 PMCID: PMC7016963 DOI: 10.3390/cells9010037] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/15/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects one-third of the population worldwide, of which a substantial number of patients suffer from non-alcoholic steatohepatitis (NASH). NASH is a severe condition characterized by steatosis and concomitant liver inflammation and fibrosis, for which no drug is yet available. NAFLD is also generally conceived as the hepatic manifestation of the metabolic syndrome. Consequently, well-established drugs that are indicated for the treatment of type 2 diabetes and hyperlipidemia are thought to exert effects that alleviate the pathological features of NASH. One class of these drugs targets peroxisome proliferator-activated receptors (PPARs), which are nuclear receptors that play a regulatory role in lipid metabolism and inflammation. Therefore, PPARs are now also being investigated as potential anti-NASH druggable targets. In this paper, we review the mechanisms of action and physiological functions of PPARs and discuss the position of the different PPAR agonists in the therapeutic landscape of NASH. We particularly focus on the PPAR agonists currently under evaluation in clinical phase II and III trials. Preclinical strategies and how refinement and optimization may improve PPAR-targeted anti-NASH drug testing are also discussed. Finally, potential caveats related to PPAR agonism in anti-NASH therapy are stipulated.
Collapse
|
114
|
Ishikawa H, Ino S, Nakashima T, Matsuo H, Takahashi Y, Kohda C, Ōmura S, Tanaka K. Improvement Effects of Trehangelin A on High-Fat Diet Causing Metabolic Clinical Conditions. Biol Pharm Bull 2019; 42:2095-2101. [PMID: 31787724 DOI: 10.1248/bpb.b19-00668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The purpose of this study is to determine whether or not trehangelin A (THG-A) is effective in treating the metabolic clinical condition caused by a high-fat diet. The body weight, epididymal adipose volume, alanine transaminase (ALT), total-cholesterol (T-CHO), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C) and glucose concentrations in serum increased in mice fed a high-fat diet compared to mice fed a control diet. On the other hand, adiponectin level in serum of mice fed a high-fat diet decreased compared to that of control mice. When mice fed a high-fat diet were intraperitoneally administered THG-A of 20 mg/kg three times per week, the levels of TG and glucose in serum were significantly reduced compared to those fed high-fat without THG-A. Interestingly, the levels of high-density lipoprotein cholesterol (HDL-C) in serum were increased by THG-A administration in both mice fed a control diet and those fed high-fat diet. The decreased level of adiponectin by a high-fat diet was also recovered by THG-A treatment. The liver expression of mRNA from pro-inflammatory cytokines, including interleukin (IL)-6 and tumor necrosis factor (TNF)-α, were significantly increased in mice fed a high-fat diet compared to those fed a control diet. However, the increased IL-6 levels in mice fed a high-fat diet were significantly suppressed by THG-A treatment. Furthermore, the increased expression of TNF-α mRNA or COL1A2 mRNA by a high-fat diets tended to be decreased in mice treated with THG-A. These results show that THG-A treatment attenuates the progression of metabolic clinical conditions, suggesting its potential efficacy against obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine
| | - Satoshi Ino
- Department of Microbiology and Immunology, Showa University School of Medicine
| | - Takuji Nakashima
- Kitasato Institute for Life Sciences, Kitasato University.,Graduate School of Pharmaceutical Sciences, Kitasato University
| | - Hirotaka Matsuo
- Kitasato Institute for Life Sciences, Kitasato University.,Graduate School of Pharmaceutical Sciences, Kitasato University
| | - Yōko Takahashi
- Kitasato Institute for Life Sciences, Kitasato University
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine
| | - Satoshi Ōmura
- Kitasato Institute for Life Sciences, Kitasato University
| | - Kazuo Tanaka
- Department of Microbiology and Immunology, Showa University School of Medicine
| |
Collapse
|
115
|
Ahmad TR, Haeusler RA. Bile acids in glucose metabolism and insulin signalling - mechanisms and research needs. Nat Rev Endocrinol 2019; 15:701-712. [PMID: 31616073 PMCID: PMC6918475 DOI: 10.1038/s41574-019-0266-7] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Of all the novel glucoregulatory molecules discovered in the past 20 years, bile acids (BAs) are notable for the fact that they were hiding in plain sight. BAs were well known for their requirement in dietary lipid absorption and biliary cholesterol secretion, due to their micelle-forming properties. However, it was not until 1999 that BAs were discovered to be endogenous ligands for the nuclear receptor FXR. Since that time, BAs have been shown to act through multiple receptors (PXR, VDR, TGR5 and S1PR2), as well as to have receptor-independent mechanisms (membrane dynamics, allosteric modulation of N-acyl phosphatidylethanolamine phospholipase D). We now also have an appreciation of the range of physiological, pathophysiological and therapeutic conditions in which endogenous BAs are altered, raising the possibility that BAs contribute to the effects of these conditions on glycaemia. In this Review, we highlight the mechanisms by which BAs regulate glucose homeostasis and the settings in which endogenous BAs are altered, and provide suggestions for future research.
Collapse
Affiliation(s)
- Tiara R Ahmad
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
116
|
Mouries J, Brescia P, Silvestri A, Spadoni I, Sorribas M, Wiest R, Mileti E, Galbiati M, Invernizzi P, Adorini L, Penna G, Rescigno M. Microbiota-driven gut vascular barrier disruption is a prerequisite for non-alcoholic steatohepatitis development. J Hepatol 2019; 71:1216-1228. [PMID: 31419514 PMCID: PMC6880766 DOI: 10.1016/j.jhep.2019.08.005] [Citation(s) in RCA: 431] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 07/29/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Fatty liver disease, including non-alcoholic fatty liver (NAFLD) and steatohepatitis (NASH), has been associated with increased intestinal barrier permeability and translocation of bacteria or bacterial products into the blood circulation. In this study, we aimed to unravel the role of both intestinal barrier integrity and microbiota in NAFLD/NASH development. METHODS C57BL/6J mice were fed with high-fat diet (HFD) or methionine-choline-deficient diet for 1 week or longer to recapitulate aspects of NASH (steatosis, inflammation, insulin resistance). Genetic and pharmacological strategies were then used to modulate intestinal barrier integrity. RESULTS We show that disruption of the intestinal epithelial barrier and gut vascular barrier (GVB) are early events in NASH pathogenesis. Mice fed HFD for only 1 week undergo a diet-induced dysbiosis that drives GVB damage and bacterial translocation into the liver. Fecal microbiota transplantation from HFD-fed mice into specific pathogen-free recipients induces GVB damage and epididymal adipose tissue enlargement. GVB disruption depends on interference with the WNT/β-catenin signaling pathway, as shown by genetic intervention driving β-catenin activation only in endothelial cells, preventing GVB disruption and NASH development. The bile acid analogue and farnesoid X receptor agonist obeticholic acid (OCA) drives β-catenin activation in endothelial cells. Accordingly, pharmacologic intervention with OCA protects against GVB disruption, both as a preventive and therapeutic agent. Importantly, we found upregulation of the GVB leakage marker in the colon of patients with NASH. CONCLUSIONS We have identified a new player in NASH development, the GVB, whose damage leads to bacteria or bacterial product translocation into the blood circulation. Treatment aimed at restoring β-catenin activation in endothelial cells, such as administration of OCA, protects against GVB damage and NASH development. LAY SUMMARY The incidence of fatty liver disease is reaching epidemic levels in the USA, with more than 30% of adults having NAFLD (non-alcoholic fatty liver disease), which can progress to more severe non-alcoholic steatohepatitis (NASH). Herein, we show that disruption of the intestinal epithelial barrier and gut vascular barrier are early events in the development of NASH. We show that the drug obeticholic acid protects against barrier disruption and thereby prevents the development of NASH, providing further evidence for its use in the prevention or treatment of NASH.
Collapse
Affiliation(s)
- Juliette Mouries
- Humanitas Clinical and Research Center – IRCCS –, via Manzoni 56, 20089 Rozzano, MI, Italy
| | - Paola Brescia
- Humanitas Clinical and Research Center – IRCCS –, via Manzoni 56, 20089 Rozzano, MI, Italy
| | - Alessandra Silvestri
- Humanitas Clinical and Research Center – IRCCS –, via Manzoni 56, 20089 Rozzano, MI, Italy
| | - Ilaria Spadoni
- Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090 Pieve Emanuele, MI, Italy
| | - Marcel Sorribas
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Reiner Wiest
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland,Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Erika Mileti
- European Institute of Oncology, Department of Experimental Oncology, 20139 Milan, MI, Italy
| | - Marianna Galbiati
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milan-Bicocca, Milan, MI, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology and Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milan-Bicocca, Milan, MI, Italy
| | | | - Giuseppe Penna
- Humanitas Clinical and Research Center – IRCCS –, via Manzoni 56, 20089 Rozzano, MI, Italy,Postbiotica srl, Via Rita Levi Montalcini, 20090 Pieve Emanuele, MI, Italy
| | - Maria Rescigno
- Humanitas Clinical and Research Center - IRCCS -, via Manzoni 56, 20089 Rozzano, MI, Italy; Humanitas University, Department of Biomedical Sciences, Via Rita Levi Montalcini, 20090 Pieve Emanuele, MI, Italy.
| |
Collapse
|
117
|
Annunziata C, Lama A, Pirozzi C, Cavaliere G, Trinchese G, Di Guida F, Nitrato Izzo A, Cimmino F, Paciello O, De Biase D, Murru E, Banni S, Calignano A, Mollica MP, Mattace Raso G, Meli R. Palmitoylethanolamide counteracts hepatic metabolic inflexibility modulating mitochondrial function and efficiency in diet-induced obese mice. FASEB J 2019; 34:350-364. [PMID: 31914699 DOI: 10.1096/fj.201901510rr] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α activation controls hepatic lipid homeostasis, stimulating fatty acid oxidation, and adapting the metabolic response to lipid overload and storage. Here, we investigate the effect of palmitoylethanolamide (PEA), an endogenous PPAR-α ligand, in counteracting hepatic metabolic inflexibility and mitochondrial dysfunction induced by high-fat diet (HFD) in mice. Long-term PEA administration (30 mg/kg/die per os) in HFD mice limited hepatic lipid accumulation, increased energy expenditure, and markedly reduced insulin resistance. In isolated liver mitochondria, we have demonstrated PEA capability to modulate mitochondrial oxidative capacity and energy efficiency, leading to the reduction of intracellular lipid accumulation and oxidative stress. Moreover, we have evaluated the effect of PEA on mitochondrial bioenergetics of palmitate-challenged HepG2 cells, using Seahorse analyzer. In vitro data showed that PEA recovered mitochondrial dysfunction and reduced lipid accumulation in insulin-resistant HepG2 cells, increasing fatty acid oxidation. Mechanistic studies showed that PEA effect on lipid metabolism was limited by AMP-activated protein kinase (AMPK) inhibition, providing evidence for a pivotal role of AMPK in PEA-induced adaptive metabolic setting. All these findings identify PEA as a modulator of hepatic lipid and glucose homeostasis, limiting metabolic inflexibility induced by nutrient overload.
Collapse
Affiliation(s)
- Chiara Annunziata
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Adriano Lama
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Claudio Pirozzi
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gina Cavaliere
- Department of Biology, University of Naples Federico II, Naples, Italy
| | | | | | | | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, Naples, Italy
| | - Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato, Cagliari, Italy
| | - Antonio Calignano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | | | - Rosaria Meli
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
118
|
Wang KN, Cao Q, Liu LY, Zhao ZJ, Liu W, Zhou DJ, Tan CP, Xia W, Ji LN, Mao ZW. Charge-driven tripod somersault on DNA for ratiometric fluorescence imaging of small molecules in the nucleus. Chem Sci 2019; 10:10053-10064. [PMID: 32055359 PMCID: PMC6991190 DOI: 10.1039/c9sc03594j] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 09/02/2019] [Indexed: 12/22/2022] Open
Abstract
Although fluorescence tracing of small bioactive molecules in living cells has been extensively studied, it is still a challenging task to detect their variations in the nucleus mainly due to the impermeable nuclear membrane and nucleic acid interference. Herein, we take advantage of the nucleic acid enriched environment in the nucleus to establish a strategy, named "charge-driven tripod somersault on DNA", for ratiometric fluorescence imaging of small bioactive molecules in the nucleus. Taking SO2 derivatives as a typical target analyte, a tripodal probe has been constructed by conjugating two DNA binding groups containing a SO2 derivative reaction site. Mechanism studies demonstrate that upon encountering and reacting with SO3 2-/HSO3 -, a charge variation occurs at the responsive arm of the tripodal probe, triggering a tripod somersault on DNA, resulting in the conformational rearrangement of the DNA binding modes with DNA-modulated fluorescence change, which allows the second emission feature to emerge. In this strategy, probe-DNA binding is not influenced by RNA or non-specific protein association, thus making it ideal for tracing nucleus-localized analytes. The application of this strategy has realized both in vitro and in vivo ratiometric fluorescence imaging of the variations of endogenous SO2 derivatives in the nucleus for the first time, with high specificity and selectivity. Also, in theory, this strategy opens up a new avenue for the design of fluorescence probes for the nucleus-localized biological analytes.
Collapse
Affiliation(s)
- Kang-Nan Wang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Qian Cao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Liu-Yi Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Zi-Jian Zhao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Wenting Liu
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Dan-Jie Zhou
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Wei Xia
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Liang-Nian Ji
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| | - Zong-Wan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry , School of Chemistry , Sun Yat-sen University , Guangzhou , 510275 , P. R. China . ;
| |
Collapse
|
119
|
Duan J, Wu Y, Liu J, Zhang J, Fu Z, Feng T, Liu M, Han J, Li Z, Chen S. Genetic Biomarkers For Hepatocellular Carcinoma In The Era Of Precision Medicine. J Hepatocell Carcinoma 2019; 6:151-166. [PMID: 31696097 PMCID: PMC6805787 DOI: 10.2147/jhc.s224849] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022] Open
Abstract
Being one of the most lethal cancers that exhibit high levels of heterogeneity, hepatocellular carcinoma (HCC) is associated with diverse oncogenic pathways underpinned by varied driver genes. HCC can be induced by different etiological factors including virus infection, toxin exposure or metabolic disorders. Consequently, patients may display varied genetic profiles, and may respond differently to the treatments involving inhibition of target pathways. These DNA/RNA mutations, copy number variations, chromatin structural changes, aberrant expression of non-coding RNAs and epigenetic modifications were considered as biomarkers in the application of precision medication. To explore how genetic testing could contribute to early diagnosis, prognosis, treatment and postoperative monitoring of HCC, we conducted a systematic review of genetic markers associated with different pathologies. Moreover, we summarized on-going clinical trials for HCC treatment, including the trials for multiple kinase inhibitors and immune checkpoint blockade (ICB). The efficacy of ICB treatment in HCC is not as good as what was observed in lung cancer and melanoma, which might be due to the heterogeneity of the microenvironment of the liver.
Collapse
Affiliation(s)
- Jingxian Duan
- Department of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
| | - Yuling Wu
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Jikui Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University Shenzhen Hospital, Shenzhen518036, People’s Republic of China
| | - Jiajia Zhang
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Zhichao Fu
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Tieshan Feng
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Ming Liu
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Jie Han
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| | - Zhicheng Li
- Department of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
| | - Shifu Chen
- Department of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People’s Republic of China
- Department of Oncology, HaploX Biotechnology Co. Ltd, Shenzhen518000, People’s Republic of China
| |
Collapse
|
120
|
Han X, Cui ZY, Song J, Piao HQ, Lian LH, Hou LS, Wang G, Zheng S, Dong XX, Nan JX, Wu YL. Acanthoic acid modulates lipogenesis in nonalcoholic fatty liver disease via FXR/LXRs-dependent manner. Chem Biol Interact 2019; 311:108794. [DOI: 10.1016/j.cbi.2019.108794] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023]
|
121
|
Ishtiaq SM, Rashid H, Hussain Z, Arshad MI, Khan JA. Adiponectin and PPAR: a setup for intricate crosstalk between obesity and non-alcoholic fatty liver disease. Rev Endocr Metab Disord 2019; 20:253-261. [PMID: 31656991 DOI: 10.1007/s11154-019-09510-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adiponectin, a soluble adipocytokine, plays an important role in the functioning of adipose tissue and in the regulation of inflammation, particularly hepatic inflammation. The adiponectin subsequently imparts a crucial role in metabolic and hepato-inflammatory diseases. The most recent evidences indicate that lipotoxicity-induced inflammation in the liver is associated with obesity-derived alterations and remolding in adipose tissue that culminates in most prevalent liver pathology named as non-alcoholic fatty liver disease (NAFLD). A comprehensive crosstalk of adiponectin and its cognate receptors, specifically adiponectin receptor-2 in the liver mediates ameliorative effects in obesity-induced NAFLD by interaction with hepatic peroxisome proliferator-activated receptors (PPARs). Recent studies highlight the implication of molecular mediators mainly involved in the pathogenesis of obesity and obesity-driven NAFLD, however, the plausible mechanisms remain elusive. The present review aimed at collating the data regarding mechanistic approaches of adiponectin and adiponectin-activated PPARs as well as PPAR-induced adiponectin levels in attenuation of hepatic lipoinflammation. Understanding the rapidly occurring adiponectin-mediated pathophysiological outcomes might be of importance in the development of new therapies that can potentially resolve obesity and obesity-associated NAFLD.
Collapse
Affiliation(s)
- Syeda Momna Ishtiaq
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Haroon Rashid
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan
| | - Zulfia Hussain
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan
| | | | - Junaid Ali Khan
- Institute of Pharmacy, Physiology and Pharmacology, University of Agriculture, Faisalabad, 38040, Pakistan.
| |
Collapse
|
122
|
Ding P, Chen Z, Chen H, Zhang Z, Liu Z, Yan X, Zhou H, Gu Q, Li C, Xu J. Structurally Selective Mechanism of Liver X Receptor Ligand: In Silico and In Vitro Studies. J Chem Inf Model 2019; 59:3277-3290. [DOI: 10.1021/acs.jcim.9b00292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Peng Ding
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Ziyang Chen
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Hao Chen
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Zizhen Zhang
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Zhihong Liu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Xin Yan
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Huihao Zhou
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Qiong Gu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Chanjuan Li
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, 132 East Circle at University City, Guangzhou 510006, China
| |
Collapse
|
123
|
Wang X, Tanaka N, Hu X, Kimura T, Lu Y, Jia F, Sato Y, Nakayama J, Moriya K, Koike K, Aoyama T. A high-cholesterol diet promotes steatohepatitis and liver tumorigenesis in HCV core gene transgenic mice. Arch Toxicol 2019; 93:1713-1725. [PMID: 31004178 DOI: 10.1007/s00204-019-02440-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/09/2019] [Indexed: 01/08/2023]
Abstract
Previous epidemiological studies have suggested a link between high-cholesterol intake and liver disease progression, including hepatocellular carcinoma (HCC). However, the precise mechanism of hepatotoxicity and hepatocarcinogenesis caused by excessive cholesterol consumption remains unclear. We aimed to investigate the impact of dietary cholesterol using hepatitis C virus core gene transgenic (HCVcpTg) mice, which spontaneously developed HCC with age. Male HCVcpTg mice were treated for 15 months with either a control diet or an isocaloric diet containing 1.5% cholesterol, and liver phenotypes and tumor-associated signaling pathways were evaluated. The high-cholesterol diet-fed HCVcpTg mice exhibited a significantly higher incidence of liver tumors compared with the control diet mice (100% vs. 41%, P < 0.001). The diet induced steatohepatitis with pericellular fibrosis and evoked higher mRNA expression of pro-inflammatory and pro-fibrotic mediators along with enhanced hepatocyte proliferation and greater oxidative and endoplasmic reticulum stress in the liver. Moreover, long-term consumption of cholesterol-rich diet activated nuclear factor-kappa B (NF-κB) and p62/sequestosome 1 (Sqstm1)-nuclear factor erythroid 2 (NRF2) axis, enhanced fibrogenesis, and consequently accelerated hepatic tumorigenesis. In conclusion, these results demonstrate that a high-cholesterol diet facilitates liver tumorigenesis by inducing steatohepatitis, promoting hepatocyte division, and up-regulating cellular stress and pro-inflammatory NF-κB and detoxifying p62/Sqstm1-NRF2 signals. Therefore, high dietary cholesterol should be avoided in HCV-infected patients to prevent development of steatohepatitis, liver fibrosis, and HCC.
Collapse
Affiliation(s)
- Xiaojing Wang
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
- Department of Gastroenterology, Lishui Hospital, Zhejiang University School of Medicine, Lishui, Zhejiang, People's Republic of China
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan.
- Research Center for Social Systems, Shinshu University, Matsumoto, Japan.
| | - Xiao Hu
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Takefumi Kimura
- Department of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yu Lu
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Fangping Jia
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| | - Yoshiko Sato
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kyoji Moriya
- Department of Infection Control and Prevention, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, 390-8621, Japan
| |
Collapse
|
124
|
Manka P, Zeller A, Syn WK. Fibrosis in Chronic Liver Disease: An Update on Diagnostic and Treatment Modalities. Drugs 2019; 79:903-927. [DOI: 10.1007/s40265-019-01126-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
125
|
Ni M, Zhang B, Zhao J, Feng Q, Peng J, Hu Y, Zhao Y. Biological mechanisms and related natural modulators of liver X receptor in nonalcoholic fatty liver disease. Biomed Pharmacother 2019; 113:108778. [PMID: 30897538 DOI: 10.1016/j.biopha.2019.108778] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/11/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is becoming a worldwide health problem, but no approved medical treatment exists so far. Nuclear receptors are one of the drug targets for nonalcoholic steatohepatitis (NASH). Among them, liver X receptor (LXR) has been studied in recent years in tumors, metabolic diseases and inflammatory diseases, but its physiological and pharmacological effects in the treatment of NASH are controversial. Activation of LXR has the potential to modulate cholesterol homeostasis, induce anti-inflammatory effects and increase insulin sensitivity, but liver lipid deposition and hypertriglyceridemia are also increased. Inhibition of liver LXR transcriptional activity in the context of NAFLD can effectively alleviate hepatic steatosis, inflammation, and fibrosis but elevates the risk of potential cardiovascular disease. The contradictory pharmacodynamic effects of LXR in the treatment of NASH increase the difficulty of developing targeted drugs. Moreover, natural compounds play an important part in drug development, and in recent years, some natural compounds have been reported to treat NAFLD by acting on LXR or LXR pathways with fewer adverse reactions, presenting a promising therapeutic prospect. In this review, we discuss the mechanisms of LXR in NASH and summarize the natural products reported to modulate NAFLD via LXR or the LXR pathway, offering an alternative approach for LXR-related drug development in NAFLD.
Collapse
Affiliation(s)
- Mingzhu Ni
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Binbin Zhang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianan Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qin Feng
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinghua Peng
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key laboratory of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiyang Hu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key laboratory of Traditional Chinese Medicine, Shanghai 201203, China; E-Institute of Shanghai Municipal Education Commission, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yu Zhao
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key laboratory of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
126
|
Esler WP, Bence KK. Metabolic Targets in Nonalcoholic Fatty Liver Disease. Cell Mol Gastroenterol Hepatol 2019; 8:247-267. [PMID: 31004828 PMCID: PMC6698700 DOI: 10.1016/j.jcmgh.2019.04.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
The prevalence and diagnosis of nonalcoholic fatty liver disease (NAFLD) is on the rise worldwide and currently has no FDA-approved pharmacotherapy. The increase in disease burden of NAFLD and a more severe form of this progressive liver disease, nonalcoholic steatohepatitis (NASH), largely mirrors the increase in obesity and type 2 diabetes (T2D) and reflects the hepatic manifestation of an altered metabolic state. Indeed, metabolic syndrome, defined as a constellation of obesity, insulin resistance, hyperglycemia, dyslipidemia and hypertension, is the major risk factor predisposing the NAFLD and NASH. There are multiple potential pharmacologic strategies to rebalance aspects of disordered metabolism in NAFLD. These include therapies aimed at reducing hepatic steatosis by directly modulating lipid metabolism within the liver, inhibiting fructose metabolism, altering delivery of free fatty acids from the adipose to the liver by targeting insulin resistance and/or adipose metabolism, modulating glycemia, and altering pleiotropic metabolic pathways simultaneously. Emerging data from human genetics also supports a role for metabolic drivers in NAFLD and risk for progression to NASH. In this review, we highlight the prominent metabolic drivers of NAFLD pathogenesis and discuss the major metabolic targets of NASH pharmacotherapy.
Collapse
Key Words
- acc, acetyl-coa carboxylase
- alt, alanine aminotransferase
- aso, anti-sense oligonucleotide
- ast, aspartate aminotransferase
- chrebp, carbohydrate response element binding protein
- ci, confidence interval
- dgat, diacylglycerol o-acyltransferase
- dnl, de novo lipogenesis
- fas, fatty acid synthase
- ffa, free fatty acid
- fgf, fibroblast growth factor
- fxr, farnesoid x receptor
- glp-1, glucagon-like peptide-1
- hdl, high-density lipoprotein
- homa-ir, homeostatic model assessment of insulin resistance
- ldl, low-density lipoprotein
- nafld, nonalcoholic fatty liver disease
- nas, nonalcoholic fatty liver disease activity score
- nash, nonalcoholic steatohepatitis
- or, odds ratio
- pdff, proton density fat fraction
- ppar, peroxisome proliferator-activated receptor
- sglt2, sodium glucose co-transporter 2
- srebp-1c, sterol regulatory element binding protein-1c
- t2d, type 2 diabetes
- t2dm, type 2 diabetes mellitus
- tg, triglyceride
- th, thyroid hormone
- thr, thyroid hormone receptor
- treg, regulatory t cells
- tzd, thiazolidinedione
- vldl, very low-density lipoprotein
Collapse
Affiliation(s)
- William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts
| | - Kendra K Bence
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development, and Medical, Cambridge, Massachusetts.
| |
Collapse
|
127
|
Liang N, Damdimopoulos A, Goñi S, Huang Z, Vedin LL, Jakobsson T, Giudici M, Ahmed O, Pedrelli M, Barilla S, Alzaid F, Mendoza A, Schröder T, Kuiper R, Parini P, Hollenberg A, Lefebvre P, Francque S, Van Gaal L, Staels B, Venteclef N, Treuter E, Fan R. Hepatocyte-specific loss of GPS2 in mice reduces non-alcoholic steatohepatitis via activation of PPARα. Nat Commun 2019; 10:1684. [PMID: 30975991 PMCID: PMC6459876 DOI: 10.1038/s41467-019-09524-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 03/12/2019] [Indexed: 02/06/2023] Open
Abstract
Obesity triggers the development of non-alcoholic fatty liver disease (NAFLD), which involves alterations of regulatory transcription networks and epigenomes in hepatocytes. Here we demonstrate that G protein pathway suppressor 2 (GPS2), a subunit of the nuclear receptor corepressor (NCOR) and histone deacetylase 3 (HDAC3) complex, has a central role in these alterations and accelerates the progression of NAFLD towards non-alcoholic steatohepatitis (NASH). Hepatocyte-specific Gps2 knockout in mice alleviates the development of diet-induced steatosis and fibrosis and causes activation of lipid catabolic genes. Integrative cistrome, epigenome and transcriptome analysis identifies the lipid-sensing peroxisome proliferator-activated receptor α (PPARα, NR1C1) as a direct GPS2 target. Liver gene expression data from human patients reveal that Gps2 expression positively correlates with a NASH/fibrosis gene signature. Collectively, our data suggest that the GPS2-PPARα partnership in hepatocytes coordinates the progression of NAFLD in mice and in humans and thus might be of therapeutic interest.
Collapse
Affiliation(s)
- Ning Liang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | | | - Saioa Goñi
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Zhiqiang Huang
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Lise-Lotte Vedin
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Tomas Jakobsson
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Marco Giudici
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Osman Ahmed
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Matteo Pedrelli
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Serena Barilla
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Fawaz Alzaid
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, 75013, France
| | - Arturo Mendoza
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, 10021, USA
| | - Tarja Schröder
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Raoul Kuiper
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
| | - Paolo Parini
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14157, Sweden
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, 14157, Sweden
- Inflammation and Infection Theme, Karolinska University Hospital, Huddinge, 14157, Sweden
| | - Anthony Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, 10021, USA
| | - Philippe Lefebvre
- University Lille, INSERM, CHU Lillie, Institut Pasteur de Lille, U1011-EGID, Lille, F-59000, France
| | - Sven Francque
- Department of Gastroenterology and Hepatology, University of Antwerp, Antwerp, 2610, Belgium
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, 2610, Belgium
| | - Luc Van Gaal
- Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, 2610, Belgium
- Department of Endocrinology, Diabetology and Metabolism, University of Antwerp, Antwerp, 2610, Belgium
| | - Bart Staels
- University Lille, INSERM, CHU Lillie, Institut Pasteur de Lille, U1011-EGID, Lille, F-59000, France
| | - Nicolas Venteclef
- INSERM, Cordeliers Research Centre, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Paris, 75013, France
| | - Eckardt Treuter
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden.
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14157, Sweden.
| |
Collapse
|
128
|
Yao L, Cao B, Cheng Q, Cai W, Ye C, Liang J, Liu W, Tan L, Yan M, Li B, He J, Hwang SH, Zhang X, Wang C, Ai D, Hammock BD, Zhu Y. Inhibition of soluble epoxide hydrolase ameliorates hyperhomocysteinemia-induced hepatic steatosis by enhancing β-oxidation of fatty acid in mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G527-G538. [PMID: 30789748 PMCID: PMC6483021 DOI: 10.1152/ajpgi.00148.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hepatic steatosis is the beginning phase of nonalcoholic fatty liver disease, and hyperhomocysteinemia (HHcy) is a significant risk factor. Soluble epoxide hydrolase (sEH) hydrolyzes epoxyeicosatrienoic acids (EETs) and other epoxy fatty acids, attenuating their cardiovascular protective effects. However, the involvement of sEH in HHcy-induced hepatic steatosis is unknown. The current study aimed to explore the role of sEH in HHcy-induced lipid disorder. We fed 6-wk-old male mice a chow diet or 2% (wt/wt) high-metnionine diet for 8 wk to establish the HHcy model. A high level of homocysteine induced lipid accumulation in vivo and in vitro, which was concomitant with the increased activity and expression of sEH. Treatment with a highly selective specific sEH inhibitor (0.8 mg·kg-1·day-1 for the animal model and 1 μM for cells) prevented HHcy-induced lipid accumulation in vivo and in vitro. Inhibition of sEH activated the peroxisome proliferator-activated receptor-α (PPAR-α), as evidenced by elevated β-oxidation of fatty acids and the expression of PPAR-α target genes in HHcy-induced hepatic steatosis. In primary cultured hepatocytes, the effect of sEH inhibition on PPAR-α activation was further confirmed by a marked increase in PPAR-response element luciferase activity, which was reversed by knock down of PPAR-α. Of note, 11,12-EET ligand dependently activated PPAR-α. Thus increased sEH activity is a key determinant in the pathogenesis of HHcy-induced hepatic steatosis, and sEH inhibition could be an effective treatment for HHcy-induced hepatic steatosis. NEW & NOTEWORTHY In the current study, we demonstrated that upregulation of soluble epoxide hydrolase (sEH) is involved in the hyperhomocysteinemia (HHcy)-caused hepatic steatosis in an HHcy mouse model and in murine primary hepatocytes. Improving hepatic steatosis in HHcy mice by pharmacological inhibition of sEH to activate peroxisome proliferator-activated receptor-α was ligand dependent, and sEH could be a potential therapeutic target for the treatment of nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Liu Yao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Boyang Cao
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Qian Cheng
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenbin Cai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chenji Ye
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jing Liang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Wenli Liu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Lu Tan
- 2Department of Laboratory Animal Science and Technology, Tianjin Medical University, Tianjin, China
| | - Meng Yan
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bochuan Li
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Jinlong He
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Sung Hee Hwang
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Xu Zhang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunjiong Wang
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Ding Ai
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bruce D. Hammock
- 3Department of Entomology and Nematology and University of California, Davis Comprehensive Cancer Center, Davis, California
| | - Yi Zhu
- 1Tianjin Key Laboratory of Metabolic Diseases; Key Laboratory of Immune Microenvironment and Disease (Ministry of Education); Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
129
|
Bueverov AO. Clinical and Pathogenetic Parallels of Nonalcoholic Fatty Liver Disease and Gallstone Disease. ACTA ACUST UNITED AC 2019. [DOI: 10.22416/1382-4376-2019-29-1-17-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aim:to analyze the data that has so far been accumulated on the pathogenetic association of gallstone disease (GD) and non-alcoholic fatty liver disease (NAFLD), as well as to assess the effect of cholecystectomy on the NAFLD course.Key findings.The relationship between GD and NAFLD is very complex and seems to be mutually aggravating. There is no doubt that there is an increased risk of GB in NAFLD patients, which is primarily associated with common pathogenetic mechanisms. These include central and peripheral insulin resistance, changes in the expression of transcription factors (liver X-receptor and farnesoid X-receptor) and the bile acid membrane receptors (TGR5). Conversely, the effect of GD on the NAFLD course is assumed, although the pathogenetic factors of this association are still unknown. In recent years, convincing data has emerged concerning the role of cholecystectomy in the NAFLD progression, which may be connected with the development of small intestinal bacterial overgrowth, as well as with the disruption of the endocrine balance and the signal function of bile acids.Conclusion.The connection between NAFLD, GD and cholecystectomy is complex and multifaceted. The study of this connection will allow new methods of treatment to be developed.
Collapse
Affiliation(s)
- Aleksey O. Bueverov
- I.M. Sechenov First Moscow State Medical University (Sechenov University); M.F. Vladimirsky Moscow Regional Research and Clinical Institute (MONIKI)
| |
Collapse
|
130
|
Molinaro A, Caesar R, L'homme L, Koh A, Ståhlman M, Staels B, Bäckhed F. Liver-specific RORα deletion does not affect the metabolic susceptibility to western style diet feeding. Mol Metab 2019; 23:82-87. [PMID: 30904385 PMCID: PMC6479759 DOI: 10.1016/j.molmet.2019.02.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
Objectives The nuclear receptor superfamily is a potential target for the development of new treatments for obesity and metabolic diseases. Increasing evidence has pointed towards the retinoic acid-related orphan receptor-alpha (RORα) as an important nuclear receptor involved in several biological processes. RORα full body knockout mice display improved metabolic phenotypes on both chow and high fat (60% fat, 20% carbohydrate) diets, but also have severe behavioral abnormalities. Here we investigated the effect of hepatic RORα by generating mice with liver-specific RORα deletion to elucidate the role of this nuclear receptor on host metabolism. Methods 8 week-old mice with liver-specific RORα deletion and littermate controls were fed either chow or western-style diets (40% fat, 40% carbohydrate) for 12 weeks. Metabolic phenotyping was performed at the end of the dietary intervention. Results Here, we show that hepatic RORα deletion does not affect the metabolic susceptibility to either chow or western-style diet in terms of glucose metabolism and adiposity. Conclusions Our data indicate that liver deletion of RORα does not have a pivotal role in the regulation of hepatic glucose and lipid metabolism on chow or western-style diet. Hepatic deletion of RORα does not affect host metabolism on chow diet. Hepatic deletion of RORα does not affect host metabolism on western-style diet. Similar phenotypes between male and female mice.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Robert Caesar
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Laurent L'homme
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000, Lille, France
| | - Ara Koh
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Marcus Ståhlman
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000, Lille, France
| | - Fredrik Bäckhed
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, 413 45, Gothenburg, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research and Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
131
|
Gao J, Song J, Du M, Mao X. Bovine α-lactalbumin hydrolysates (α-LAH) attenuate high-fat diet induced nonalcoholic fatty liver disease by modulating hepatic lipid metabolism in C57BL/6J mice. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.01.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
132
|
Ohtani N, Kawada N. Role of the Gut-Liver Axis in Liver Inflammation, Fibrosis, and Cancer: A Special Focus on the Gut Microbiota Relationship. Hepatol Commun 2019; 3:456-470. [PMID: 30976737 PMCID: PMC6442695 DOI: 10.1002/hep4.1331] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022] Open
Abstract
The gut and the liver are anatomically and physiologically connected, and this “gut–liver axis” exerts various influences on liver pathology. The gut microbiota consists of various microorganisms that normally coexist in the human gut and have a role of maintaining the homeostasis of the host. However, once homeostasis is disturbed, metabolites and components derived from the gut microbiota translocate to the liver and induce pathologic effects in the liver. In this review, we introduce and discuss the mechanisms of liver inflammation, fibrosis, and cancer that are influenced by gut microbial components and metabolites; we include recent advances in molecular‐based therapeutics and novel mechanistic findings associated with the gut–liver axis and gut microbiota.
Collapse
Affiliation(s)
- Naoko Ohtani
- Department of Pathophysiology Osaka City University, Graduate School of Medicine Osaka Japan
| | - Norifumi Kawada
- Department of Hepatology Osaka City University, Graduate School of Medicine Osaka Japan
| |
Collapse
|
133
|
Saderi N, Báez-Ruiz A, Azuara-Álvarez LE, Escobar C, Salgado-Delgado RC. Differential Recovery Speed of Activity and Metabolic Rhythms in Rats After an Experimental Protocol of Shift-Work. J Biol Rhythms 2019; 34:154-166. [PMID: 30764694 DOI: 10.1177/0748730419828534] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The circadian system drives the temporal organization of body physiology in relation to the changing daily environment. Shift-work (SW) disrupts this temporal order and is associated with the loss of homeostasis and metabolic syndrome. In a rodent model of SW based on forced activity in the rest phase for 4 weeks, we describe the occurrence of circadian desynchrony, as well as metabolic and liver dysfunction. To provide better evidence for the impact of altered timing of activity, this study explored how long it takes to recover metabolic rhythms and behavior. Rats were submitted to experimental SW for 4 weeks and then were left to recover for one week. Daily locomotor activity, food intake patterns, serum glucose and triglycerides, and the expression levels of hepatic Pparα, Srebp-1c, Pepck, Bmal1 and Per2 were assessed during the recovery period and were compared with expected data according to a control condition. SW triggered the circadian desynchronization of all of the analyzed parameters. A difference in the time required for realignment was observed among parameters. Locomotor activity achieved the expected phase on day 2, whereas the nocturnal feeding pattern was restored on the sixth recovery day. Daily rhythms of plasma glucose and triglycerides and of Pparα, Pepck and Bmal1 expression in the liver resynchronized on the seventh day, whereas Srebp-1c and Per2 persisted arrhythmic for the entire recovery week. SW does not equally affect behavior and metabolic rhythms, leading to internal desynchrony during the recovery phase.
Collapse
Affiliation(s)
- Nadia Saderi
- Departamento de Fisiología, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Adrián Báez-Ruiz
- Departamento de Fisiología, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Lucia E Azuara-Álvarez
- Departamento de Fisiología, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| | - Carolina Escobar
- Departamento de Anatomia, Facultad de Medicina, Universidad Autónoma de Mexico, Distrito Federal, Mexico
| | - Roberto C Salgado-Delgado
- Departamento de Fisiología, Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, San Luis Potosí, México
| |
Collapse
|
134
|
Tanaka N, Kimura T, Fujimori N, Nagaya T, Komatsu M, Tanaka E. Current status, problems, and perspectives of non-alcoholic fatty liver disease research. World J Gastroenterol 2019; 25:163-177. [PMID: 30670907 PMCID: PMC6337019 DOI: 10.3748/wjg.v25.i2.163] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 12/24/2018] [Accepted: 12/27/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major chronic liver disease that can lead to liver cirrhosis, liver cancer, and ultimately death. NAFLD is pathologically classified as non-alcoholic fatty liver (NAFL) or non-alcoholic steatohepatitis (NASH) based on the existence of ballooned hepatocytes, although the states have been known to transform into each other. Moreover, since the detection of ballooned hepatocytes may be difficult with limited biopsied specimens, its clinical significance needs reconsideration. Repeated liver biopsy to assess histological NAFLD activity for therapeutic response is also impractical, creating the need for body fluid biomarkers and less invasive imaging modalities. Recent longitudinal observational studies have emphasized the importance of advanced fibrosis as a determinant of NAFLD outcome. Thus, identifying predictors of fibrosis progression and developing better screening methods will enable clinicians to isolate high-risk NAFLD patients requiring early intensive intervention. Despite the considerable heterogeneity of NAFLD with regard to underlying disease, patient age, and fibrosis stage, several clinical trials are underway to develop a first-in-class drug. In this review, we summarize the present status and future direction of NAFLD/NASH research towards solving unmet medical needs.
Collapse
Affiliation(s)
- Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
- International Research Center for Agricultural Food Industry, Shinshu University, Matsumoto 390-8621, Japan
| | - Takefumi Kimura
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Naoyuki Fujimori
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Tadanobu Nagaya
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Michiharu Komatsu
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Eiji Tanaka
- Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| |
Collapse
|
135
|
Concurrent exercise improves insulin resistance and nonalcoholic fatty liver disease by upregulating PPAR-γ and genes involved in the beta-oxidation of fatty acids in ApoE-KO mice fed a high-fat diet. Lipids Health Dis 2019; 18:6. [PMID: 30611282 PMCID: PMC6320624 DOI: 10.1186/s12944-018-0933-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 11/27/2018] [Indexed: 12/16/2022] Open
Abstract
Objective To emphasize the mechanism of concurrent exercise effect on lipid disorders in insulin resistance (IR) and nonalcoholic fatty liver disease (NAFLD). Materials and methods Twenty male ApoE knockout mice were randomly divided into two groups: HFD group (n = 10) fed a high fat diet, and HFDE group (n = 10) with high-fat diet intervention for 12 weeks and swimming exercise. Other ten healthy male C57BL/6 J mice were fed a normal diet, and included as control group. Retro-orbital blood samples were collected for biochemical analysis. Oil red O staining of liver tissues was performed to confirm the exercise effect. Western blotting was performed to evaluate the expressions of PPAR-γ, CPT-1, MCAD. Results The levels of TG, TC, LDL, FFA, FIN, FPG and Homa-IRI in the HFD group were significantly higher than ND group, while these were markedly decreased in the HFDE group compared with HFD group. The Oil Red O staining of liver samples further confirmed the exercise effect on the change of lipid deposition in the liver. Western blotting showed increased expressions of PPAR-γ, CPT-1, MCAD induced by high fat diet were significantly downregulated by exercise. Conclusion A concurrent 12-week exercise protocol alleviated the lipid metabolism disorders of IR and NAFLD, probably via PPAR-γ/CPT-1/MCAD signaling.
Collapse
|
136
|
Disease Progression and Pharmacological Intervention in a Nutrient-Deficient Rat Model of Nonalcoholic Steatohepatitis. Dig Dis Sci 2019; 64:1238-1256. [PMID: 30511198 PMCID: PMC6548202 DOI: 10.1007/s10620-018-5395-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 11/22/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND There is a marked need for improved animal models of nonalcoholic steatohepatitis (NASH) to facilitate the development of more efficacious drug therapies for the disease. METHODS Here, we investigated the development of fibrotic NASH in male Wistar rats fed a choline-deficient L-amino acid-defined (CDAA) diet with or without cholesterol supplementation for subsequent assessment of drug treatment efficacy in NASH biopsy-confirmed rats. The metabolic profile and liver histopathology were evaluated after 4, 8, and 12 weeks of dieting. Subsequently, rats with biopsy-confirmed NASH were selected for pharmacological intervention with vehicle, elafibranor (30 mg/kg/day) or obeticholic acid (OCA, 30 mg/kg/day) for 5 weeks. RESULTS The CDAA diet led to marked hepatomegaly and fibrosis already after 4 weeks of feeding, with further progression of collagen deposition and fibrogenesis-associated gene expression during the 12-week feeding period. Cholesterol supplementation enhanced the stimulatory effect of CDAA on gene transcripts associated with fibrogenesis without significantly increasing collagen deposition. Pharmacological intervention with elafibranor, but not OCA, significantly reduced steatohepatitis scores, and fibrosis-associated gene expression, however, was unable to prevent progression in fibrosis scores. CONCLUSION CDAA-fed rats develop early-onset progressive NASH, which offers the opportunity to probe anti-NASH compounds with potential disease-modifying properties.
Collapse
|
137
|
Lu Y, Harada M, Kamijo Y, Nakajima T, Tanaka N, Sugiyama E, Kyogashima M, Gonzalez FJ, Aoyama T. Peroxisome proliferator-activated receptor α attenuates high-cholesterol diet-induced toxicity and pro-thrombotic effects in mice. Arch Toxicol 2019; 93:149-161. [PMID: 30341732 PMCID: PMC6661123 DOI: 10.1007/s00204-018-2335-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is involved in the regulation of fatty acid and cholesterol metabolism. A high-cholesterol (HC) diet increases the risk of developing cardiovascular diseases (CVD); however, it is unclear whether the toxic effects of cholesterol involve changes in thrombotic factor expression, and whether PPARα is necessary for such effects. To investigate this possibility, we fed a HC diet to wild-type (WT) and Ppara-null mice and measured cholesterol and triglyceride contents, liver histology, serum/plasma levels of coagulation factors, hepatic expression of the coagulation factors, liver/serum sulfatide levels, hepatic sulfatide metabolism, hepatic expression of lipid transporters, and hepatic oxidative stress and its relating enzymes. In Ppara-null mice, the HC diet caused triglyceride accumulation and exacerbated inflammation and oxidative stress in liver, increased levels of coagulation factors, including tissue factor, plasminogen activator inhibitor-1 and carboxypeptidase B2 in blood and liver, and decreased levels of anti-thrombotic sulfatides in serum and liver. These changes were much less marked in WT mice. These findings imply that cholesterol overload exerts its toxic effects at least in part by enhancing thrombosis, secondary to abnormal hepatic lipid metabolism, inflammation, and oxidative stress. Moreover, we reveal for the first time that PPARα can attenuate these toxic effects by transcriptional regulation of coagulation factors and sulfatides, in addition to its known effects of controlling lipid homeostasis and suppressing inflammation and oxidative stress. Therapies aimed at activating PPARα might prevent HC diet-induced CVD through modulating various pro- and anti-thrombotic factors.
Collapse
Affiliation(s)
- Yu Lu
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Makoto Harada
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Department of Nephrology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Yuji Kamijo
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
- Department of Nephrology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan.
| | - Takero Nakajima
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Research Center for the Agricultural Food Industry, Shinshu University, Nagano, Nagano, 380-8553, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
- Research Center for the Agricultural Food Industry, Shinshu University, Nagano, Nagano, 380-8553, Japan
| | - Eiko Sugiyama
- Department of Food and Health Sciences, The University of Nagano, 8-49-7 Miwa, Nagano, Nagano, 380-8525, Japan
| | - Mamoru Kyogashima
- Division of Microbiology and Molecular Cell Biology, Nihon Pharmaceutical University, 10281 Komuro, Inamachi, Kita-Adachigun, Saitama, 362-0806, Japan
| | - Frank J Gonzalez
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Toshifumi Aoyama
- Department of Metabolic Regulation, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| |
Collapse
|
138
|
Concomitant PPARα and FXR Activation as a Putative Mechanism of NASH Improvement after Gastric Bypass Surgery: a GEO Datasets Analysis. J Gastrointest Surg 2019; 23:51-57. [PMID: 30206765 DOI: 10.1007/s11605-018-3938-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/16/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Compared to non-surgical weight loss (Diet), weight loss after Roux-en-Y gastric bypass (RYGB) results in greater rates of non-alcoholic steatohepatitis (NASH) resolution. Changes in bile acid physiology and farnesoid X receptor (FXR) signaling are suspected mediators of postoperative NASH improvement. Recent experimental evidence suggests that upregulation of hepatic peroxisome proliferator-activated receptor α (PPARα) activity might also impact NASH improvement. As FXR partly regulates PPARα, we compared resolution of NASH and changes in hepatic PPARα and FXR gene expression following Diet and RYGB. METHODS We searched the Gene Expression Omnibus database to identify human studies with liver biopsies containing genomic data and histologic NASH features, at baseline and after Diet or RYGB. Microarray data were extracted for PPARα and FXR gene expression analyses using GEOquery R package v.2.42.0. RESULTS We identified one study (GSE83452) where patients underwent either Diet (n = 29) or RYGB (n = 25). NASH prevalence was similar at baseline (Diet 76% versus RYGB 60%, P = ns). After 1 year, NASH resolved in 93.3% of RYGB but only in 27.3% of Diet (P < 0.001). Hepatic PPARα and FXR gene expression increased only after RYGB (P < 0.001). These changes were also found when analyzing only patients that resolved NASH (P < 0.01), and patients without NASH at baseline and follow-up (P < 0.05). CONCLUSIONS Compared to Diet, RYGB results in greater NASH resolution with concurrent upregulation of hepatic PPARα and FXR. Our findings point to concurrent PPARα and FXR activation, triggered by RYGB, as a potential mechanism to improve NASH.
Collapse
|
139
|
Guo S, Lu J, Zhuo Y, Xiao M, Xue X, Zhong S, Shen X, Yin C, Li L, Chen Q, Zhu M, Chen B, Zhao M, Zheng L, Tao Y, Yin H. Endogenous cholesterol ester hydroperoxides modulate cholesterol levels and inhibit cholesterol uptake in hepatocytes and macrophages. Redox Biol 2018; 21:101069. [PMID: 30576926 PMCID: PMC6302155 DOI: 10.1016/j.redox.2018.101069] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Dysregulation of cholesterol metabolism represents one of the major risk factors for atherosclerotic cardiovascular disease (CVD). Oxidized cholesterol esters (oxCE) in low-density lipoprotein (LDL) have been implicated in CVD but the underlying mechanisms remain poorly defined. We use a targeted lipidomic approach to demonstrate that levels of oxCEs in human plasma are associated with different types of CVD and significantly elevated in patients with myocardial infarction. We synthesized a major endogenous cholesterol ester hydroperoxide (CEOOH), cholesteryl-13(cis, trans)-hydroperoxy-octadecadienoate (ch-13(c,t)-HpODE) and show that this endogenous compound significantly increases plasma cholesterol level in mice while decrease cholesterol levels in mouse liver and peritoneal macrophages, which is primarily due to the inhibition of cholesterol uptake in macrophages and liver. Further studies indicate that inhibition of cholesterol uptake by ch-13(c,t)-HpODE in macrophages is dependent on LXRα-IDOL-LDLR pathway, whereas inhibition on cholesterol levels in hepatocytes is dependent on LXRα and LDLR. Consistently, these effects on cholesterol levels by ch-13(c,t)-HpODE are diminished in LDLR or LXRα knockout mice. Together, our study provides evidence that elevated plasma cholesterol levels by CEOOHs are primarily due to the inhibition of cholesterol uptake in the liver and macrophages, which may play an important role in the pathogenesis of CVD.
Collapse
Affiliation(s)
- Shuyuan Guo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Jianhong Lu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China
| | - Yujuan Zhuo
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Mengqing Xiao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Xinli Xue
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China
| | - Shanshan Zhong
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China
| | - Xia Shen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Chunzhao Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Luxiao Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Qun Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China
| | - Mingjiang Zhu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Buxing Chen
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing, China
| | - Yongzhen Tao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of Chinese Academy of Sciences, CAS, Beijing 100049, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100000, China.
| |
Collapse
|
140
|
Oh YJ, Kim HY, Lee MH, Suh SH, Choi Y, Nam TG, Kwon WY, Lee SY, Yoo YH. Cilostazol Improves HFD-Induced Hepatic Steatosis by Upregulating Hepatic STAMP2 Expression through AMPK. Mol Pharmacol 2018; 94:1401-1411. [PMID: 30366981 DOI: 10.1124/mol.118.113217] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/05/2018] [Indexed: 12/29/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is an increasingly studied condition that can progress to end-stage liver disease. Although NAFLD was first described in 1980, a complete understanding of the mechanism and causes of this disease is still lacking. Six-transmembrane protein of prostate 2 (STAMP2) plays a role in integrating inflammatory and nutritional signals with metabolism. Our previous study suggested that STAMP2 may be a suitable target for treating NAFLD. In the current study, we performed a focused drug-screening and found that cilostazol could be a potential STAMP2 enhancer. Thus, we examined whether cilostazol alleviates NAFLD through STAMP2. The in vivo and in vitro pharmacological efficacies of cilostazol on STAMP2 expression and lipid accumulation were analyzed in NAFLD mice induced by high-fat diet (HFD) and in HepG2 cell lines treated by oleic acid (OA), respectively. Cilostazol increased the expression of STAMP2 through transcriptional regulation in vivo and in vitro. Cilostazol also dampened the STAMP2 downregulation caused by the HFD and by OA in vivo and in vitro, respectively. Cilostazol activated AMP-activated protein kinase (AMPK) in vivo and in vitro, and AMPK functions upstream of STAMP2, and reversed downregulation of STAMP2 expression through AMPK in the NAFLD model. Cilostazol ameliorates hepatic steatosis by enhancing hepatic STAMP2 expression through AMPK. Enhancing STAMP2 expression with cilostazol represents a potential therapeutic avenue for treatment of NAFLD.
Collapse
Affiliation(s)
- Yoo Jin Oh
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Hye Young Kim
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Mi Hwa Lee
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Sung Hwan Suh
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Yongmun Choi
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Tae-Gyu Nam
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Woo Young Kwon
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Sang Yeob Lee
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| | - Young Hyun Yoo
- Departments of Anatomy and Cell Biology (Y.J.O., H.Y.K., M.H.L., W.Y.K., Y.H.Y.), Endocrinology Medicine (S.H.S.), and Rheumatology (S.Y.L.), Dong-A University College of Medicine, Busan, Republic of Korea; Gyeonggi Bio Center, Gyeonggi-do Business and Science Accelerator, Suwon, Republic of Korea (Y.C.); and Department of Pharmacy and Institute of Pharmaceutical Science and Technology, Hanyang University, Ansan, Republic of Korea (T.-G.N.)
| |
Collapse
|
141
|
Komatsu M, Tanaka N, Kimura T, Fujimori N, Sano K, Horiuchi A, Sugiura A, Yamazaki T, Shibata S, Joshita S, Umemura T, Matsumoto A, Tanaka E. Miglitol attenuates non-alcoholic steatohepatitis in diabetic patients. Hepatol Res 2018; 48:1092-1098. [PMID: 29935004 DOI: 10.1111/hepr.13223] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/03/2018] [Accepted: 06/16/2018] [Indexed: 12/13/2022]
Abstract
AIM Postprandial hyperglycemia is frequently accompanied by non-alcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH). Although α-glucosidase inhibitors (αGIs) can slow glucose absorption from the intestine and suppress the surge of circulating glucose concentration after meals, it remains unclear whether αGIs are also beneficial for NASH. The aim of this prospective study was to examine the efficacy and safety of miglitol, a typical αGI, for NASH. METHODS Seventeen patients with histologically confirmed NASH and hemoglobin A1c (HbA1c) >6.5% were treated with miglitol (150 mg/day) for 12 months. The changes in clinical parameters and liver histology were analyzed. RESULTS All patients completed the 12-month miglitol treatment course with no severe adverse events. The treatment significantly decreased body mass index, serum alanine aminotransferase levels, and HbA1c (all P < 0.001). Post-treatment liver biopsy of 11 patients revealed significant improvements in steatosis (from 2.2 ± 0.6 to 1.5 ± 0.7, P = 0.001), lobular inflammation (from 1.8 ± 0.8 to 1.3 ± 0.5, P = 0.014), portal inflammation scores (from 0.6 ± 0.5 to 0.1 ± 0.3, P = 0.025), and NAFLD activity score (from 5.5 ± 1.5 to 3.9 ± 1.4, P = 0.012). Fibrosis and hepatocyte ballooning scores were unchanged. CONCLUSIONS Miglitol appears to safely ameliorate NASH activity by attenuation of steatosis and lobular/portal inflammation. Appropriately powered controlled trials are warranted to validate our results.
Collapse
Affiliation(s)
- Michiharu Komatsu
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoki Tanaka
- Department of Metabolic Regulation, Shinshu University School of Medicine, Matsumoto, Japan
- International Research Center for Agricultural Food Industry, Shinshu University, Matsumoto, Japan
| | - Takefumi Kimura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoyuki Fujimori
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kenji Sano
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Akira Horiuchi
- Digestive Disease Center, Showa Inan General Hospital, Komagane, Japan
| | - Ayumi Sugiura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tomoo Yamazaki
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Soichiro Shibata
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Satoru Joshita
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeji Umemura
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Akihiro Matsumoto
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Eiji Tanaka
- Department of Internal Medicine, Division of Gastroenterology, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
142
|
Takahashi S, Tanaka N, Fukami T, Xie C, Yagai T, Kim D, Velenosi TJ, Yan T, Krausz KW, Levi M, Gonzalez FJ. Role of Farnesoid X Receptor and Bile Acids in Hepatic Tumor Development. Hepatol Commun 2018; 2:1567-1582. [PMID: 30556042 PMCID: PMC6287584 DOI: 10.1002/hep4.1263] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 09/09/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths worldwide, and an association between altered bile acid (BA) metabolism, down-regulation of farnesoid X receptor (FXR), which is a master regulator of BA metabolism, and hepatocarcinogenesis has been documented. While global FXR deficiency in mice results in spontaneous HCC with aging, the contribution of tissue-specific FXR deficiency to hepatocarcinogenesis remains unclear. In this study, the prevalence of hepatic tumors, expression of genes related to tumorigenesis, and serum/liver BA levels were compared among male whole-body Fxr-null, hepatocyte-specific Fxr-null (Fxr ∆Hep), and enterocyte-specific Fxr-null (Fxr ∆IE) mice at the age of 3, 14, and 20 months. More than 90% of 20-month-old whole-body Fxr-null mice had hepatic tumors with enhanced hepatic expression of myelocytomatosis oncogene (Myc) and cyclin-dependent kinase 4 (Cdk4) messenger RNAs (mRNAs) and elevated serum taurocholate (TCA) and tauromuricholate (TMCA) and their respective unconjugated derivatives. The incidence of hepatic tumors was significantly lower in Fxr ∆Hep and Fxr ∆IE mice (20% and 5%, respectively), and the increases in Myc and Cdk4 mRNA or serum BA concentrations were not detected in these mice compared to Fxr floxed [fl]/fl mice; a similar tendency was observed in 14-month-old mice. However, increased hepatic c-Myc protein expression was found only in Fxr-null mice at the age of 3, 14, and 20 months. Treatment with TCA induced Myc expression in Fxr-null cultured primary mouse hepatocytes but not in wild-type (WT) mouse hepatocytes, demonstrating that the combination of hepatocyte FXR disruption with elevated TCA is required for Myc induction and ensuing age-dependent hepatocarcinogenesis in Fxr-null mice. Conclusion: There is a relatively low risk of hepatic tumors by inhibition of FXR in enterocytes, likely due to the lack of increased TCA and Myc induction.
Collapse
Affiliation(s)
- Shogo Takahashi
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
- Department of Biochemistry and Molecular and Cellular BiologyGeorgetown UniversityWashingtonDC
| | - Naoki Tanaka
- Department of Metabolic RegulationShinshu University School of MedicineMatsumotoJapan
- International Research Center for Agricultural Food IndustryShinshu UniversityMatsumotoJapan
| | - Tatsuki Fukami
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
- Present address:
Department of Drug Metabolism and Toxicology, Faculty of Pharmaceutical SciencesKanazawa UniversityKanazawaJapan
| | - Cen Xie
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Tomoki Yagai
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Donghwan Kim
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Thomas J. Velenosi
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Tingting Yan
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Kristopher W. Krausz
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular BiologyGeorgetown UniversityWashingtonDC
| | - Frank J. Gonzalez
- Laboratory of MetabolismNational Cancer Institute, National Institutes of HealthBethesdaMD
| |
Collapse
|
143
|
Dietrich K, Baumgart J, Eshkind L, Reuter L, Gödtel-Armbrust U, Butt E, Musheev M, Marini F, More P, Grosser T, Niehrs C, Wojnowski L, Mathäs M. Health-Relevant Phenotypes in the Offspring of Mice Given CAR Activators Prior to Pregnancy. Drug Metab Dispos 2018; 46:1827-1835. [PMID: 30154105 DOI: 10.1124/dmd.118.082925] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatic induction in response to drugs and environmental chemicals affects drug therapies and energy metabolism. We investigated whether the induction is transmitted to the offspring. We injected 3-day- and 6-week-old F0 female mice with TCPOBOP, an activator of the nuclear receptor constitutive androstane receptor (CAR, NR1I3), and mated them 1-6 weeks afterward. We detected in the offspring long-lasting alterations of CAR-mediated drug disposition, energy metabolism, and lipid profile. The transmission to the first filial generation (F1) was mediated by TCPOBOP transfer from the F0 adipose tissue via milk, as revealed by embryo transfer, crossfostering experiments, and liquid chromatography-mass spectrometry analyses. The important environmental pollutant PCB153 activated CAR in the F1 generation in a manner similar to TCPOBOP. Our findings indicate that chemicals accumulating and persisting in adipose tissue may exert liver-mediated, health-relevant effects on F1 offspring simply via physical transmission in milk. Such effects may occur even if treatment has been terminated far ahead of conception. This should be considered in assessing developmental toxicity and in the long-term follow-up of offspring of mothers exposed to both approved and investigational drugs, and to chemicals with known or suspected accumulation in adipose tissue.
Collapse
Affiliation(s)
- Karin Dietrich
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Jan Baumgart
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Leonid Eshkind
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Lea Reuter
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Ute Gödtel-Armbrust
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Elke Butt
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Michael Musheev
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Federico Marini
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Piyush More
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Tanja Grosser
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Christof Niehrs
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Leszek Wojnowski
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| | - Marianne Mathäs
- Department of Pharmacology (K.D., L.R., U.G.-A., P.M., T.G., L.W., M.Ma.) and Institute of Medical Biostatistics, Epidemiology and Informatics (F.M.), University Medical Center Mainz, Mainz, Germany; Translational Animal Research Center (J.B., L.E.), University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Institute of Experimental Biomedicine II, University Hospital Würzburg, Würzburg, Germany (E.B.); Institute of Molecular Biology, Mainz, Germany (M.Mu., C.N.); and Division of Molecular Embryology, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany (C.N.)
| |
Collapse
|
144
|
Giancristofaro A, Barbosa AJM, Ammazzalorso A, Amoia P, De Filippis B, Fantacuzzi M, Giampietro L, Maccallini C, Amoroso R. Discovery of new FXR agonists based on 6-ECDCA binding properties by virtual screening and molecular docking. MEDCHEMCOMM 2018; 9:1630-1638. [PMID: 30393515 PMCID: PMC6194413 DOI: 10.1039/c8md00272j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 06/28/2018] [Indexed: 01/03/2023]
Abstract
FXR is a member of the nuclear receptor superfamily, which regulates the expression of various genes involved in bile acid, lipid and glucose metabolism. Targeting FXR with small molecules has been exploited to treat lipid-related disorders and diseases such as cholestasis, gallstones and hepatic disorders. In this work, we expand the existing pool of known FXR agonists using a fast hit-to-lead structure-based pharmacophore and docking screening protocol. A set of 25 molecules was selected after screening a large database of commercial chemicals, and experimental tests were carried out to demonstrate their ability to activate FXR. Three novel FXR agonists are reported, namely, one full agonist, more efficient than the endogenous ligand chenodeoxycholic acid, and two partial agonists.
Collapse
Affiliation(s)
- Antonella Giancristofaro
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Arménio J M Barbosa
- Chemistry Department , Faculdade de Ciências e Tecnologia , Universidade Nova de Lisboa , 2829-516 Caparica , Portugal
| | - Alessandra Ammazzalorso
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Pasquale Amoia
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Barbara De Filippis
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Marialuigia Fantacuzzi
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Letizia Giampietro
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Cristina Maccallini
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| | - Rosa Amoroso
- Department of Pharmacy , University of Chieti "G. d'Annunzio" , via dei vestini 31 , 66100 Chieti , Italy .
| |
Collapse
|
145
|
Maldonado EM, Fisher CP, Mazzatti DJ, Barber AL, Tindall MJ, Plant NJ, Kierzek AM, Moore JB. Multi-scale, whole-system models of liver metabolic adaptation to fat and sugar in non-alcoholic fatty liver disease. NPJ Syst Biol Appl 2018; 4:33. [PMID: 30131870 PMCID: PMC6102210 DOI: 10.1038/s41540-018-0070-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 07/30/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a serious public health issue associated with high fat, high sugar diets. However, the molecular mechanisms mediating NAFLD pathogenesis are only partially understood. Here we adopt an iterative multi-scale, systems biology approach coupled to in vitro experimentation to investigate the roles of sugar and fat metabolism in NAFLD pathogenesis. The use of fructose as a sweetening agent is controversial; to explore this, we developed a predictive model of human monosaccharide transport, signalling and metabolism. The resulting quantitative model comprising a kinetic model describing monosaccharide transport and insulin signalling integrated with a hepatocyte-specific genome-scale metabolic network (GSMN). Differential kinetics for the utilisation of glucose and fructose were predicted, but the resultant triacylglycerol production was predicted to be similar for monosaccharides; these predictions were verified by in vitro data. The role of physiological adaptation to lipid overload was explored through the comprehensive reconstruction of the peroxisome proliferator activated receptor alpha (PPARα) regulome integrated with a hepatocyte-specific GSMN. The resulting qualitative model reproduced metabolic responses to increased fatty acid levels and mimicked lipid loading in vitro. The model predicted that activation of PPARα by lipids produces a biphasic response, which initially exacerbates steatosis. Our data support the evidence that it is the quantity of sugar rather than the type that is critical in driving the steatotic response. Furthermore, we predict PPARα-mediated adaptations to hepatic lipid overload, shedding light on potential challenges for the use of PPARα agonists to treat NAFLD.
Collapse
Affiliation(s)
- Elaina M. Maldonado
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Ciarán P. Fisher
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ UK
| | | | - Amy L. Barber
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
| | - Marcus J. Tindall
- Department of Mathematics and Statistics, University of Reading, Berkshire, RG6 6AX UK
- Institute of Cardiovascular and Metabolic Research, University of Reading, Berkshire, RG6 6UR UK
| | - Nicholas J. Plant
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT UK
| | - Andrzej M. Kierzek
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- Certara UK Limited, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, S1 2BJ UK
| | - J. Bernadette Moore
- School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, GU2 7XH UK
- School of Food Science & Nutrition, University of Leeds, Leeds, West Yorkshire, LS2 9JT UK
| |
Collapse
|
146
|
Xiao XH, Wang YD, Qi XY, Wang YY, Li JY, Li H, Zhang PY, Liao HL, Li MH, Liao ZZ, Yang J, Xu CX, Wen GB, Liu JH. Zinc alpha2 glycoprotein protects against obesity-induced hepatic steatosis. Int J Obes (Lond) 2018; 42:1418-1430. [PMID: 30006580 DOI: 10.1038/s41366-018-0151-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/14/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic steatosis, impaired insulin sensitivity, and chronic low-grade inflammation. Our previous studies indicated that zinc alpha2 glycoprotein (ZAG) alleviates palmitate (PA)-induced intracellular lipid accumulation in hepatocytes. This study is to further characterize the roles of ZAG on the development of hepatic steatosis, insulin resistance (IR), and inflammation. METHODS ZAG protein levels in the livers of NAFLD patients, high-fat diet (HFD)-induced or genetically (ob/ob) induced obese mice, and in PA-treated hepatocytes were determined by western blotting. C57BL/6J mice injected with an adenovirus expressing ZAG were fed HFD for indicated time to induce hepatic steatosis, IR, and inflammation, and then biomedical, histological, and metabolic analyses were conducted to identify pathologic alterations in these mice. The molecular mechanisms underlying ZAG-regulated hepatic steatosis were further explored and verified in mice and hepatocytes. RESULTS ZAG expression was decreased in NAFLD patient liver biopsy samples, obese mice livers, and PA-treated hepatocytes. Simultaneously, ZAG overexpression alleviated intracellular lipid accumulation via upregulating adiponectin and lipolytic genes (FXR, PPARα, etc.) while downregulating lipogenic genes (SREBP-1c, LXR, etc.) in obese mice as well as in cultured hepatocytes. ZAG improved insulin sensitivity and glucose tolerance via activation of IRS/AKT signaling. Moreover, ZAG significantly inhibited NF-ĸB/JNK signaling and thus resulting in suppression of obesity-associated inflammatory response in hepatocytes. CONCLUSIONS Our results revealed that ZAG could protect against NAFLD by ameliorating hepatic steatosis, IR, and inflammation.
Collapse
Affiliation(s)
- Xin-Hua Xiao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Ya-Di Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Xiao-Yan Qi
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Yuan-Yuan Wang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jiao-Yang Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Han Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Pei-Ying Zhang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Hai-Lin Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Mei-Hua Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Zhe-Zhen Liao
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jing Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Can-Xin Xu
- Department of Pathology & Immunology, Washington University in, St. Louis, MO, 63110, USA
| | - Ge-Bo Wen
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China
| | - Jiang-Hua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital of University of South China, Hengyang, 421001, China.
| |
Collapse
|
147
|
Hu YB, Liu XY, Zhan W. Farnesoid X receptor agonist INT-767 attenuates liver steatosis and inflammation in rat model of nonalcoholic steatohepatitis. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2213-2221. [PMID: 30038487 PMCID: PMC6052932 DOI: 10.2147/dddt.s170518] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction Nonalcoholic steatohepatitis (NASH) is largely driven by the dysregulation of liver metabolism and inflammation. Bile acids and their receptor Farnesoid X receptor (FXR) play a critical role in the disease development. Here, we investigated whether INT-767, the newly-identified dual FXR/TGR5 agonist, can protect rat from liver injury during NASH. Materials and methods NASH model was established by feeding the male SD rats with high-fat diet for 16 weeks. INT-767 was given by gavage to NASH rats from week 13 to week 16. At the end of 16 weeks, liver and serum were harvested, and bile acids, glucose and lipid metabolism, liver injury and histological features were evaluated. Results INT-767 treatment significantly alleviates high-fat caused liver damage characterized with lipid accumulation and hepatic infiltration of immune cells. INT-767 robustly restores the lipid, glucose metabolism to normal level, attenuates insulin resistance through upregulating FXR level and reverting the dysregulation of its target genes in liver metabolism. Molecularly INT-767 also attenuates the pro-inflammatory response by suppression of TNF-α and NF-κB signaling pathway. Conclusion INT-767 may be an attractive candidate for a potential novel strategy on the treatment of NASH.
Collapse
Affiliation(s)
- Ying-Bin Hu
- Department of Gastroenterology, Puai Hospital (Wuhan Fourth Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,
| | - Xin-Yu Liu
- Cancer Science Institute, National University of Singapore, Singapore
| | - Wei Zhan
- Department of Gastroenterology, Puai Hospital (Wuhan Fourth Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China,
| |
Collapse
|
148
|
Wang X, Wu F, Li G, Zhang N, Song X, Zheng Y, Gong C, Han B, He G. Lipid-modified cell-penetrating peptide-based self-assembly micelles for co-delivery of narciclasine and siULK1 in hepatocellular carcinoma therapy. Acta Biomater 2018; 74:414-429. [PMID: 29787814 DOI: 10.1016/j.actbio.2018.05.030] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 05/05/2018] [Accepted: 05/18/2018] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer, and one therapeutic approach is to target both the AMPK and autophagy pathways in order to synergistically promote programmed cell death. Here, a series of amphiphilic, lipid-modified cell-penetrating peptides were synthesized and allowed to self-assemble into micelles loaded with the AMPK activator narciclasine (Narc) and short interfering RNA targeting the unc-51-like kinase 1 (siULK1). The size of these micelles, their efficiency of transfection into cells, and their ability to release drug or siRNA cargo in vitro were pH-sensitive, such that drug release was facilitated in the acidic microenvironment of the tumor. Transfecting the micelles into HCC cells significantly inhibited protective autophagy within tumor cells, and delivering the micelles into mice carrying HCC xenografts induced apoptosis, slowed tumor growth, and inhibited autophagy. Our results indicate that co-delivering Narc and siULK1 in biocompatible micelles can safely inhibit tumor growth and protective autophagy, justifying further studies into this promising therapeutic approach against HCC. STATEMENT OF SIGNIFICANCE We have focused on the targeted therapy of HCC via synergistically inhibiting the autophagy and inducing apoptosis. The lipid-modified cell-penetrating peptide can not only aggregate into micelles to load natural product narciclasine and ULK1 siRNA simultaneously, but also facilitate uptake and endosome escape with a pH-sensitive manner in HepG2 cells. HepG2 cell treated with siULK1-M-Narc has increased apoptotic levels and declined autophagy via the targeted regulation of AMPK-ULK1 signaling axis. The in vivo studies have confirmed that siULK1-M-Narc efficiently reduce the growth of tumor on HCC xenograft models with good safety. Thus, we suppose the lipid-modified cell-penetrating peptide has good application prospects in the targeted combinational therapy of HCC.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Fengbo Wu
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Guoyou Li
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610065, China.
| | - Nan Zhang
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xiangrong Song
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Yu Zheng
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Changyang Gong
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo Han
- State Key Laboratory Breeding Base of Systematic Research Development and Utilization of Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Gu He
- Department of Pharmacy and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
149
|
Tran M, Liu Y, Huang W, Wang L. Nuclear receptors and liver disease: Summary of the 2017 basic research symposium. Hepatol Commun 2018; 2:765-777. [PMID: 30129636 PMCID: PMC6049066 DOI: 10.1002/hep4.1203] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/03/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022] Open
Abstract
The nuclear receptor superfamily contains important transcriptional regulators that play pleiotropic roles in cell differentiation, development, proliferation, and metabolic processes to govern liver physiology and pathology. Many nuclear receptors are ligand-activated transcription factors that regulate the expression of their target genes by modulating transcriptional activities and epigenetic changes. Additionally, the protein complex associated with nuclear receptors consists of a multitude of coregulators, corepressors, and noncoding RNAs. Therefore, acquiring new information on nuclear receptors may provide invaluable insight into novel therapies and shed light on new interventions to reduce the burden and incidence of liver diseases. (Hepatology Communications 2018;2:765-777).
Collapse
Affiliation(s)
- Melanie Tran
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT
| | - Yanjun Liu
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute City of Hope National Medical Center Duarte CA
| | - Li Wang
- Department of Physiology and Neurobiology and Institute for Systems Genomics, University of Connecticut, Storrs, CT.,Veterans Affairs Connecticut Healthcare System West Haven CT.,Department of Internal Medicine, Section of Digestive Diseases Yale University New Haven CT
| |
Collapse
|
150
|
Noratto GD, Lage NN, Chew BP, Mertens-Talcott SU, Talcott ST, Pedrosa ML. Non-anthocyanin phenolics in cherry (Prunus avium L.) modulate IL-6, liver lipids and expression of PPARδ and LXRs in obese diabetic (db/db) mice. Food Chem 2018; 266:405-414. [PMID: 30381205 DOI: 10.1016/j.foodchem.2018.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/09/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022]
Abstract
Anthocyanin-rich cherries are known for preventing/decreasing risk factors associated with obesity; however, the specific benefits exerted by cherry non-anthocyanin phenolics are not clear. Obese diabetic (db/db) mice fed a diet supplemented with anthocyanin-depleted cherry powder (cherry) were compared to db/db (obese) or lean counterparts (lean) fed a control isocaloric diet for 12 weeks. The reduced plasma interleukin (IL)-6 and improved liver health may be mediated by cherry fibre and non-anthocyanin phenolics. Benefits for liver health included reduction of lipids and protein carbonyls, and modulation of peroxisome proliferator-activated receptor (PPAR)δ mRNA to resemble levels in lean. Lack of plasma antilipidemic, improvement of antioxidant defenses, and PPARα/γ mRNA modulation in liver suggest cherry anthocyanins specific benefits. This is the first study to elucidate in vivo the potential benefits of cherry non-anthocyanin phenolics for diabetes-induced liver disorders and the importance of choosing processing technologies that preserve anthocyanins and health benefits of whole cherries.
Collapse
Affiliation(s)
- Giuliana D Noratto
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, United States.
| | - Nara N Lage
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, United States; Research Center in Biological Sciences, Federal University of Ouro Preto, Minas Gerais, Brazil
| | - Boon P Chew
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, United States
| | | | - Stephen T Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, United States
| | - Maria L Pedrosa
- Research Center in Biological Sciences, Federal University of Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|