1
|
Jung E, Baek EB, Hong EJ, Kang JH, Park S, Park S, Hong EJ, Cho YE, Ko JW, Won YS, Kwon HJ. TXNIP in liver sinusoidal endothelial cells ameliorates alcohol-associated liver disease via nitric oxide production. Int J Biol Sci 2024; 20:606-620. [PMID: 38169654 PMCID: PMC10758096 DOI: 10.7150/ijbs.90781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/02/2023] [Indexed: 01/05/2024] Open
Abstract
Dysregulation of liver sinusoidal endothelial cell (LSEC) differentiation and function has been reported in alcohol-associated liver disease (ALD). Impaired nitric oxide (NO) production stimulates LSEC capillarization and dysfunction; however, the mechanism underlying NO production remains unclear. Here, we investigated the role of thioredoxin-interacting protein (TXNIP), an important regulator of redox homeostasis, in endothelial cell NO production and its subsequent effects on ALD progression. We found that hepatic TXNIP expression was upregulated in patients with ALD and in ethanol diet-fed mice with high expression in LSECs. Endothelial cell-specific Txnip deficiency (TxnipΔEC) in mice exacerbated alcohol-induced liver injury, inflammation, fibrosis, and hepatocellular carcinoma development. Deletion of Txnip in LSECs led to sinusoidal capillarization, downregulation of NO production, and increased release of proinflammatory cytokines and adhesion molecules, whereas TXNIP overexpression had the opposite effects. Mechanistically, TXNIP interacted with transforming growth factor β-activated kinase 1 (TAK1) and subsequently suppressed the TAK1 pathway. Inhibition of TAK1 activation restored NO production and decreased the levels of proinflammatory cytokines, thereby, blocking liver injury and inflammation in TxnipΔEC mice. Our findings indicate that upregulated TXNIP expression in LSECs serves a protective role in ameliorating ALD. Enhancing TXNIP expression could, therefore, be a potential therapeutic approach for ALD.
Collapse
Affiliation(s)
- Eunhye Jung
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eun Bok Baek
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Eun-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jee Hyun Kang
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Suyoung Park
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sehee Park
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk 28116, Republic of Korea
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Eun Cho
- Andong National University, Andong 36729, Republic of Korea
| | - Je-Won Ko
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Suk Won
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Chungbuk 28116, Republic of Korea
| | - Hyo-Jung Kwon
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
2
|
Liu XQ, Wang JJ, Wu X, Liu ZN, Wu BM, Lv XW. Blocking ATP-P1Rs axis attenuate alcohol-related liver fibrosis. Life Sci 2023; 328:121896. [PMID: 37385371 DOI: 10.1016/j.lfs.2023.121896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
AIMS The aim of this study was to explore the fibrogenic effects of ATP-P1Rs axis and ATP-P2Rs axis on alcohol-related liver fibrosis (ALF). MATERIALS AND METHODS C57BL/6J CD73 knock out (KO) mice were used in our study. 8-12 weeks male mice were used as an ALF model in vivo. In conclusion, after one week of adaptive feeding, 5 % alcohol liquid diet was given for 8 weeks. High-concentration alcohol (31.5 %, 5 g/kg) was administered by gavage twice weekly, and 10 % CCl4 intraperitoneal injections (1 ml/kg) were administered twice weekly for the last two weeks. The mice in the control group were injected intraperitoneally with an equivalent volume of normal saline. Fasting for 9 h after the last injection, blood samples were collected, and related indicators were tested. In vitro, rat hepatic stellate cells (HSCs) were treated with 200 μM acetaldehyde to establish an alcoholic liver fibrosis for 48 h, then tested related indicators. KEY FINDINGS We found that both adenosine receptors including adenosine A1, A2A, A2B, A3 receptors (A1R, A2AR, A2BR, A3R) and ATP receptors including P2X7, P2Y2 receptors (P2X7R, P2Y2R) were expressed increased in ALF. After CD73 was knocked out, we found that adenosine receptors expression decreased, ATP expression increased, and fibrosis degree decreased. SIGNIFICANCE Based on the research, we discovered that adenosine plays a more important role in ALF. Therefore, blocking the ATP-P1Rs axis represented a potential treatment for ALF, and CD73 will become a potential therapeutic target.
Collapse
Affiliation(s)
- Xue-Qi Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Jun-Jie Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Zhen-Ni Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Bao-Ming Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiong-Wen Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| |
Collapse
|
3
|
Yan C, Hu W, Tu J, Li J, Liang Q, Han S. Pathogenic mechanisms and regulatory factors involved in alcoholic liver disease. J Transl Med 2023; 21:300. [PMID: 37143126 PMCID: PMC10158301 DOI: 10.1186/s12967-023-04166-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/27/2023] [Indexed: 05/06/2023] Open
Abstract
Alcoholism is a widespread and damaging behaviour of people throughout the world. Long-term alcohol consumption has resulted in alcoholic liver disease (ALD) being the leading cause of chronic liver disease. Many metabolic enzymes, including alcohol dehydrogenases such as ADH, CYP2E1, and CATacetaldehyde dehydrogenases ALDHsand nonoxidative metabolizing enzymes such as SULT, UGT, and FAEES, are involved in the metabolism of ethanol, the main component in alcoholic beverages. Ethanol consumption changes the functional or expression profiles of various regulatory factors, such as kinases, transcription factors, and microRNAs. Therefore, the underlying mechanisms of ALD are complex, involving inflammation, mitochondrial damage, endoplasmic reticulum stress, nitrification, and oxidative stress. Moreover, recent evidence has demonstrated that the gut-liver axis plays a critical role in ALD pathogenesis. For example, ethanol damages the intestinal barrier, resulting in the release of endotoxins and alterations in intestinal flora content and bile acid metabolism. However, ALD therapies show low effectiveness. Therefore, this review summarizes ethanol metabolism pathways and highly influential pathogenic mechanisms and regulatory factors involved in ALD pathology with the aim of new therapeutic insights.
Collapse
Affiliation(s)
- Chuyun Yan
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Key Lab of Microanalytical Methods & Instrumentation, Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Jinqi Tu
- The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College of Wuhu, Wannan Medical College, Wuhu, 241000, Anhui, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Key Lab of Microanalytical Methods & Instrumentation, Department of Chemistry, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China
| | - Shuxin Han
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, 830046, China.
| |
Collapse
|
4
|
Kim YS, Ko B, Kim DJ, Tak J, Han CY, Cho JY, Kim W, Kim SG. Induction of the hepatic aryl hydrocarbon receptor by alcohol dysregulates autophagy and phospholipid metabolism via PPP2R2D. Nat Commun 2022; 13:6080. [PMID: 36241614 PMCID: PMC9568535 DOI: 10.1038/s41467-022-33749-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Disturbed lipid metabolism precedes alcoholic liver injury. Whether and how AhR alters degradation of lipids, particularly phospho-/sphingo-lipids during alcohol exposure, was not explored. Here, we show that alcohol consumption in mice results in induction and activation of aryl hydrocarbon receptor (AhR) in the liver, and changes the hepatic phospho-/sphingo-lipids content. The levels of kynurenine, an endogenous AhR ligand, are elevated with increased hepatic tryptophan metabolic enzymes in alcohol-fed mice. Either alcohol or kynurenine treatment promotes AhR activation with autophagy dysregulation via AMPK. Protein Phosphatase 2 Regulatory Subunit-Bdelta (Ppp2r2d) is identified as a transcriptional target of AhR. Consequently, PPP2R2D-dependent AMPKα dephosphorylation causes autophagy inhibition and mitochondrial dysfunction. Hepatocyte-specific AhR ablation attenuates steatosis, which is associated with recovery of phospho-/sphingo-lipids content. Changes of AhR targets are corroborated using patient specimens. Overall, AhR induction by alcohol inhibits autophagy in hepatocytes through AMPKα, which is mediated by Ppp2r2d gene transactivation, revealing an AhR-dependent metabolism of phospho-/sphingo-lipids.
Collapse
Affiliation(s)
- Yun Seok Kim
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea ,grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Bongsub Ko
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea
| | - Da Jung Kim
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.412484.f0000 0001 0302 820XMetabolomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul, 03082 Korea
| | - Jihoon Tak
- grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea ,grid.255168.d0000 0001 0671 5021College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| | - Chang Yeob Han
- grid.31501.360000 0004 0470 5905College of Pharmacy, Seoul National University, Seoul, Republic of Korea ,grid.411545.00000 0004 0470 4320School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do, 54896 Korea
| | - Joo-Youn Cho
- grid.31501.360000 0004 0470 5905Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, 03080 Korea ,grid.31501.360000 0004 0470 5905Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Won Kim
- grid.31501.360000 0004 0470 5905Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul National University College of Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Sang Geon Kim
- grid.255168.d0000 0001 0671 5021College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Kyeonggi-do 10326 Republic of Korea
| |
Collapse
|
5
|
Arumugam MK, Chava S, Perumal SK, Paal MC, Rasineni K, Ganesan M, Donohue TM, Osna NA, Kharbanda KK. Acute ethanol-induced liver injury is prevented by betaine administration. Front Physiol 2022; 13:940148. [PMID: 36267591 PMCID: PMC9577233 DOI: 10.3389/fphys.2022.940148] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Binge drinking is the most common form of excessive alcohol use. Repeated episodes of binge drinking cause multiple organ injuries, including liver damage. We previously demonstrated that chronic ethanol administration causes a decline in the intrahepatic ratio of S-adenosylmethionine (SAM) to S-adenosylhomocysteine (SAH). This decline causes impairments in essential methylation reactions that result in alcohol-induced fatty liver (steatosis) and other features of alcohol-associated liver disease (ALD). Co-treatment with betaine during chronic ethanol feeding, normalizes hepatocellular SAM:SAH ratio and alleviates many features of liver damage including steatosis. Here, we sought to examine whether betaine treatment similarly protects against liver injury in an alcohol binge-drinking model. We hypothesized that ethanol binge with prior or simultaneous betaine administration would prevent or attenuate acute alcohol-induced liver damage. Male C57Bl/6 mice were gavaged twice, 12 h apart, with either 6 g ethanol/kg BW or with an equal volume/kg BW of 0.9% NaCl. Two separate groups of mice (n = 5/group) were gavaged with 4 g betaine/kg BW, either 2 h before or simultaneously with the ethanol or saline gavages. All mice were sacrificed 8 h after the last gavage and serum and liver parameters were quantified. Ethanol binges caused a 50% decrease in hepatic SAM:SAH ratio and a >3-fold rise in liver triglycerides (p ≤ 0.05). These latter changes were accompanied by elevated serum AST and ALT activities and blood alcohol concentrations (BAC) that were ∼three-times higher than the legal limit of intoxication in humans. Mice that were treated with betaine 2 h before or simultaneously with the ethanol binges exhibited similar BAC as in mice given ethanol-alone. Both betaine treatments significantly elevated hepatic SAM levels thereby normalizing the SAM:SAH ratio and attenuating hepatic steatosis and other injury parameters, compared with mice given ethanol alone. Simultaneous betaine co-administration with ethanol was more effective in preventing or attenuating liver injury than betaine given before ethanol gavage. Our findings confirm the potential therapeutic value of betaine administration in preventing liver injury after binge drinking in an animal model.
Collapse
Affiliation(s)
- Madan Kumar Arumugam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Srinivas Chava
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Sathish Kumar Perumal
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Matthew C. Paal
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Karuna Rasineni
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Terrence M. Donohue
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
6
|
Han M, Böhlke M, Maher T, Kim J. Alcohol exposure increases manganese accumulation in the brain and exacerbates manganese-induced neurotoxicity in mice. Arch Toxicol 2021; 95:3665-3679. [PMID: 34590183 DOI: 10.1007/s00204-021-03166-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
Environmental and occupational exposure to heavy metals remains one of the major concerns in public health. Increased levels of manganese (Mn) pollution are associated with profound neurotoxic effects, including neurobehavioral deficits and disturbances resembling Parkinson's disease. While Mn absorption is in part mediated by iron transporters, recent studies have shown that the levels of iron transporters are modified by alcohol and that chronic alcohol consumption increases body iron stores. However, it is largely unexplored whether alcohol exposure influences the transport and neurotoxicity of Mn. To address this question, we exposed mice to ethanol (10%; v/v) by drinking water for 4 weeks, during which period MnCl2 (5 mg/kg) or saline solutions were administered daily by intranasal instillation. Ethanol consumption in mice increased brain Mn levels in a dose-dependent manner after Mn instillation, determined by inductively-coupled plasma mass spectrometry, which was accompanied by up-regulation of iron transporters, as assessed by western blotting and qPCR. In addition, alcohol drinking increased hypoxic response and decreased hepcidin expression, providing the molecular mechanism of increased iron transporters and Mn uptake upon alcohol consumption. Moreover, brain dopamine levels, analyzed by HPLC, were decreased after intranasal Mn instillation, which was worsened by alcohol. Likewise, alcohol-Mn co-exposure synergistically altered dopaminergic protein expression. Finally, alcohol binge-drinking, which resembles alcohol drinking manner in humans, increased brain Mn content along with upregulation of iron transporters. Our study suggests that individuals who consume alcohol may have a higher risk of Mn neurotoxicity upon Mn exposure.
Collapse
Affiliation(s)
- Murui Han
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Mark Böhlke
- Department of Pharmaceutical Sciences, MCPHS University, Boston, MA, USA
| | - Timothy Maher
- Department of Pharmaceutical Sciences, MCPHS University, Boston, MA, USA
| | - Jonghan Kim
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA. .,Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Suite 4, Lowell, MA, 01854, USA.
| |
Collapse
|
7
|
Lazebnik LB, Golovanova EV, Alekseenko SA, Bueverov AO, Plotnikova EY, Dolgushina AI, Ilchenko LY, Ermolova TV, Tarasova LV, Lee ED, Tsyganova YV, Akhmedov VA, Ageeva EA, Losev VM, Kupriyanova IN, Serikova SN, Korochanskaya NV, Vologzhanina LG, Zimmerman YS, Sas EI, Zhuravel SV, Okovitiy SV, Osipenko MF, Radchenko VG, Soldatova GS, Sitkin SI, Seliverstov PV, Shavkuta GV, Butova EN, Kozhevnikova SA. Russian Consensus “Hyperammonemia in Adults” (Version 2021). EXPERIMENTAL AND CLINICAL GASTROENTEROLOGY 2021:97-118. [DOI: 10.31146/1682-8658-ecg-187-3-97-118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Justification Given the large number of reports on the peculiarities of liver lesions during the Sars-Cov-2 infection [1], a team of experts who participated in the 23rd Congress of the Scientific Society of Gastroenterologists of Russia and 15 National Congress of Therapists of November 19, 2020 decided to make additions to the Russian Consensus of “Hyperammonemia in Adults” published early 2020 [2, 3].
Collapse
Affiliation(s)
- L. B. Lazebnik
- “Moscow State University of Medicine and Density n. a. A. I. Evdokimov”
| | - E. V. Golovanova
- “Moscow State University of Medicine and Density n. a. A. I. Evdokimov”
| | | | - A. O. Bueverov
- I. M. Sechenov first Moscow state medical university (Sechenov university); Moscow regional research and clinical Institute of M. F. Vladimirsky
| | - E. Yu. Plotnikova
- Federal State Budgetary Institution of Higher Education Kemerovo state medical University of the Ministry of health of Russia
| | - A. I. Dolgushina
- “South-Ural State Medical University” of the Ministry of Health of Russia
| | - L. Yu. Ilchenko
- Pirogov Russian National Research Medical University (RNRMU)
| | - T. V. Ermolova
- North- Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - L. V. Tarasova
- BI of HE “The Surgut State University”; “The Chuvashian State University”
| | - E. D. Lee
- Multifocal Medicine Center of The Central Bank of Russian Federation
| | | | - V. A. Akhmedov
- “Omsk State Medical University” of the Ministry of Health
| | | | | | - I. N. Kupriyanova
- “Ural state medical University” of the Ministry of health of the Russian Federation
| | - S. N. Serikova
- State Budgetary Institution of Health Care “Region Clinic Hospital Nr 2” Health Ministry of Krasnodar Region
| | - N. V. Korochanskaya
- State Budgetary Institution of Health Care “Region Clinic Hospital Nr 2” Health Ministry of Krasnodar Region
| | - L. G. Vologzhanina
- “Perm State Medical University named E. A. Wagner” of the Ministry of Health of Russia
| | - Ya. S. Zimmerman
- “Perm State Medical University named E. A. Wagner” of the Ministry of Health of Russia
| | - E. I. Sas
- Military Medical Academy named after S. M. Kirov
| | - S. V. Zhuravel
- “Moscow State University of Medicine and Density n. a. A. I. Evdokimov”; Scientific Research Institute of Emergency Medicine of N. V. Sklifosovskiy of Healthcare Department of Moscow
| | - S. V. Okovitiy
- Saint Petersburg State Chemical Pharmaceutical University (SPCPA)
| | - M. F. Osipenko
- Public budgetary educational institution of higher education “Novosibirsk State Medical University” of the Ministry of Healthcare of the Russia
| | | | | | - S. I. Sitkin
- North- Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation; Federal Medical and Biological Agency “State Research Institute of Highly Pure Biopreparations”
| | - P. V. Seliverstov
- North- Western state medical University named after I. I. Mechnikov, Ministry of health of the Russian Federation
| | - G. V. Shavkuta
- Rostov State Medical University of the Ministry of Health of Russia
| | - E. N. Butova
- Rostov State Medical University of the Ministry of Health of Russia
| | | |
Collapse
|
8
|
Cheng L, Jain D, Kakar S, Torbenson MS, Wu TT, Yeh MM. Hepatocellular neoplasms arising in genetic metabolic disorders: steatosis is common in both the tumor and background liver. Hum Pathol 2020; 108:93-99. [PMID: 33245984 DOI: 10.1016/j.humpath.2020.11.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/31/2020] [Accepted: 11/19/2020] [Indexed: 01/10/2023]
Abstract
Hepatocellular neoplasms can develop in multiple genetic metabolic disorders. While there have been rare case reports, clinical and pathological characterizations have not been systematically performed. We conducted a retrospective study in 9 patients with these rare genetic metabolic disorders, including glycogen storage disease type 1, ornithine carbamyl transferase deficiency, hereditary tyrosinemia type 1, and Navajo neurohepatopathy, who developed hepatocellular neoplasms. Our results show that steatosis is a common finding in both tumor (6/9 cases, 67%) and background liver parenchyma (8/9 cases, 89%), underlying a possible role for steatosis in tumorigenesis in these genetic metabolic disorders. Our findings also raise a consideration of underlying genetic metabolic disorder when young patients with hepatocellular neoplasm show steatosis in both the tumor and background liver.
Collapse
Affiliation(s)
- Lin Cheng
- Rush University Medical Center, Department of Pathology, Chicago, IL 60612, United States
| | - Dhanpat Jain
- Yale University, Department of Pathology, New Haven, CT 06510, United States
| | - Sanjay Kakar
- University of California, Department of Pathology, San Francisco, CA 94143, United States
| | | | - Tsung-Teh Wu
- Mayo Clinic, Department of Pathology, Rochester, MN 55905, United States
| | - Matthew M Yeh
- University of Washington, Department of Laboratory Medicine and Pathology, Seattle, WA 98195, United States; University of Washington, Department of Medicine, Seattle, WA 98195, United States.
| |
Collapse
|
9
|
Kim YS, Kim SG. Endoplasmic reticulum stress and autophagy dysregulation in alcoholic and non-alcoholic liver diseases. Clin Mol Hepatol 2020; 26:715-727. [PMID: 32951410 PMCID: PMC7641579 DOI: 10.3350/cmh.2020.0173] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022] Open
Abstract
Alcoholic and non-alcoholic liver diseases begin from an imbalance in lipid metabolism in hepatocytes as the earliest response. Both liver diseases share common disease features and stages (i.e., steatosis, hepatitis, cirrhosis, and hepatocellular carcinoma). However, the two diseases have differential pathogenesis and clinical symptoms. Studies have elucidated the molecular basis underlying similarities and differences in the pathogenesis of the diseases; the factors contributing to the progression of liver diseases include depletion of sulfhydryl pools, enhanced levels of reactive oxygen and nitrogen intermediates, increased sensitivity of hepatocytes to toxic cytokines, mitochondrial dysfunction, and insulin resistance. Endoplasmic reticulum (ER) stress, which is caused by the accumulation of misfolded proteins and calcium depletion, contributes to the pathogenesis, often causing catastrophic cell death. Several studies have demonstrated a mechanism by which ER stress triggers liver disease progression. Autophagy is an evolutionarily conserved process that regulates organelle turnover and cellular energy balance through decomposing damaged organelles including mitochondria, misfolded proteins, and lipid droplets. Autophagy dysregulation also exacerbates liver diseases. Thus, autophagy-related molecules can be potential therapeutic targets for liver diseases. Since ER stress and autophagy are closely linked to each other, an understanding of the molecules, gene clusters, and networks engaged in these processes would be of help to find new remedies for alcoholic and non-alcoholic liver diseases. In this review, we summarize the recent findings and perspectives in the context of the molecular pathogenesis of the liver diseases.
Collapse
Affiliation(s)
- Yun Seok Kim
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy, Seoul National University, Seoul, Korea.,College of Pharmacy, Dongguk University, Goyang, Korea
| |
Collapse
|
10
|
Arumugam MK, Talawar S, Listenberger L, Donohue TM, Osna NA, Kharbanda KK. Role of Elevated Intracellular S-Adenosylhomocysteine in the Pathogenesis of Alcohol-Related Liver Disease. Cells 2020; 9:1526. [PMID: 32585865 PMCID: PMC7349643 DOI: 10.3390/cells9061526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The earliest manifestation of alcohol-related liver disease (ALD) is steatosis, characterized by the accumulation of lipid droplets (LDs) in hepatocytes. Findings from our laboratory have indicated that many pathological changes, including steatosis, correlate with the alcohol-induced hepatocellular increases in S-adenosylhomocysteine (SAH). Based on these considerations, we hypothesized that an experimental increase in intracellular SAH alone will result in similar steatotic changes to those seen after alcohol exposure. METHODS Freshly isolated rat hepatocytes grown on collagen-coated plates were exposed to serum-free medium containing 50 µmol/L oleic acid and varying concentrations of 3-deazaadenosine (DZA) to experimentally elevate intracellular SAH levels. RESULTS Overnight exposure to DZA treatment dose-dependently increased hepatocellular triglyceride accumulation, which was also evident by morphological visualization of larger-sized LDs. The rise in triglycerides and LDs accompanied increases in mRNA and protein levels of several LD-associated proteins known to regulate LD number and size. Furthermore, DZA treatment caused a decline in the levels of lipases that prevent fat accumulation as well as increased the expression of factors involved in lipogenesis and fatty acid mobilization. Collectively, our results indicate that the elevation of intracellular SAH is sufficient to promote fat accumulation in hepatocytes, which is similar to that seen after alcohol exposure.
Collapse
Affiliation(s)
- Madan Kumar Arumugam
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.K.A.); (S.T.); (T.M.D.J.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sharanappa Talawar
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.K.A.); (S.T.); (T.M.D.J.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Laura Listenberger
- Departments of Biology and Chemistry, St. Olaf College, Northfield, MN 55057, USA;
| | - Terrence M. Donohue
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.K.A.); (S.T.); (T.M.D.J.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.K.A.); (S.T.); (T.M.D.J.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.K.A.); (S.T.); (T.M.D.J.); (N.A.O.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
11
|
Distinct metabolic adaptation of liver circadian pathways to acute and chronic patterns of alcohol intake. Proc Natl Acad Sci U S A 2019; 116:25250-25259. [PMID: 31757851 DOI: 10.1073/pnas.1911189116] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Binge drinking and chronic exposure to ethanol contribute to alcoholic liver diseases (ALDs). A potential link between ALDs and circadian disruption has been observed, though how different patterns of alcohol consumption differentially impact hepatic circadian metabolism remains virtually unexplored. Using acute versus chronic ethanol feeding, we reveal differential reprogramming of the circadian transcriptome in the liver. Specifically, rewiring of diurnal SREBP transcriptional pathway leads to distinct hepatic signatures in acetyl-CoA metabolism that are translated into the subcellular patterns of protein acetylation. Thus, distinct drinking patterns of alcohol dictate differential adaptation of hepatic circadian metabolism.
Collapse
|
12
|
Zhang Y, Xue W, Zhang W, Yuan Y, Zhu X, Wang Q, Wei Y, Yang D, Yang C, Chen Y, Sun Y, Wang S, Huang K, Zheng L. Histone methyltransferase G9a protects against acute liver injury through GSTP1. Cell Death Differ 2019; 27:1243-1258. [PMID: 31515511 PMCID: PMC7206029 DOI: 10.1038/s41418-019-0412-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 11/24/2022] Open
Abstract
Acute liver injury is commonly caused by bacterial endotoxin/lipopolysaccharide (LPS), and by drug overdose such as acetaminophen (APAP). The exact role of epigenetic modification in acute liver injury remains elusive. Here, we investigated the role of histone methyltransferase G9a in LPS- or APAP overdose-induced acute liver injury. Under d-galactosamine sensitization, liver-specific G9a-deficient mice (L-G9a−/−) exhibited 100% mortality after LPS injection, while the control and L-G9a+/− littermates showed very mild mortality. Moreover, abrogation of hepatic G9a or inhibiting the methyltransferase activity of G9a aggravated LPS-induced liver damage. Similarly, under sublethal APAP overdose, L-G9a−/− mice displayed more severe liver injury. Mechanistically, ablation of G9a inhibited H3K9me1 levels at the promoters of Gstp1/2, two liver detoxifying enzymes, and consequently suppressed their transcription. Notably, treating L-G9a−/− mice with recombinant mouse GSTP1 reversed the LPS- or APAP overdose-induced liver damage. Taken together, we identify a novel beneficial role of G9a-GSTP1 axis in protecting against acute liver injury.
Collapse
Affiliation(s)
- Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Weili Xue
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Wenquan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Xiuqin Zhu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Yujuan Wei
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Dong Yang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Chen Yang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Yan Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China
| | - Yu Sun
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China
| | - Shun Wang
- Department of Blood Transfusion, Wuhan Hospital of Traditional and Western Medicine, Wuhan, 430022, PR China
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, Hubei, PR China.
| |
Collapse
|
13
|
Yan T, Huang J, Nisar MF, Wan C, Huang W. The Beneficial Roles of SIRT1 in Drug-Induced Liver Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8506195. [PMID: 31354914 PMCID: PMC6636535 DOI: 10.1155/2019/8506195] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/27/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023]
Abstract
Drug-induced liver injury (DILI) is a major cause of acute liver failure (ALF) as a result of accumulated drugs in the human body metabolized into toxic agents and helps generate heavy oxidative stress, inflammation, and apoptosis, which induces necrosis in hepatocytes and ultimately damages the liver. Sirtuin 1 (SIRT1) is said to have multiple vital roles in cell proliferation, aging, and antistress systems of the human body. The levels of SIRT1 and its activation precisely modulate its critical role in the interaction between multiple step procedures of DILI. The nuclear factor kappa-light-chain-enhancer of activated B cell- (NF-κB-) mediated inflammation signaling pathway, reactive oxygen species (ROS), DNA damage, mitochondrial membrane potential collapse, and endoplasmic reticulum (ER) stress also contribute to aggravate DILI. Apoptosis is regarded as the terminal reaction followed by multiple signaling cascades including caspases, p53, and mitochondrial dysfunction which have been said to contribute in DILI. The SIRT1 activator is regarded as a potential candidate for DILI, because the former could inhibit signaling of p53, NF-κB, and ER stress. On the other hand, overexpression of SIRT1 also enhances the activation of antioxidant responses via Kelch-like ECH-associated protein 1- (Keap1-) nuclear factor- (erythroid-derived 2-) like 2 (Nrf2) signaling. The current manuscript will highlight the mechanism of DILI and the interaction of SIRT1 with various cytoplasmic factors leading to DILI along with the summary of potent SIRT1 agonists.
Collapse
Affiliation(s)
- Tingdong Yan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jinlong Huang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| | - Muhammad Farrukh Nisar
- Department of Physiology and Biochemistry, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Chunpeng Wan
- Jiangxi Key Laboratory for Postharvest Technology and Nondestructive Testing of Fruits & Vegetables, Collaborative Innovation Center of Post-Harvest Key Technology and Quality Safety of Fruits and Vegetables, College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China
| | - Weifeng Huang
- The Institute of Infection and Inflammation, Department of Microbiology and Immunology, Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| |
Collapse
|
14
|
Lamas-Paz A, Hao F, Nelson LJ, Vázquez MT, Canals S, Gómez del Moral M, Martínez-Naves E, Nevzorova YA, Cubero FJ. Alcoholic liver disease: Utility of animal models. World J Gastroenterol 2018; 24:5063-5075. [PMID: 30568384 PMCID: PMC6288648 DOI: 10.3748/wjg.v24.i45.5063] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease (ALD) is a major cause of acute and chronic liver injury. Extensive evidence has been accumulated on the pathological process of ALD during the past decades. However, effective treatment options for ALD are very limited due to the lack of suitable in vivo models that recapitulate the full spectrum of ALD. Experimental animal models of ALD, particularly rodents, have been used extensively to mimic human ALD. An ideal animal model should recapitulate all aspects of the ALD process, including significant steatosis, hepatic neutrophil infiltration, and liver injury. A better strategy against ALD depends on clear diagnostic biomarkers, accurate predictor(s) of its progression and new therapeutic approaches to modulate stop or even reverse the disease. Numerous models employing rodent animals have been established in the last decades to investigate the effects of acute and chronic alcohol exposure on the initiation and progression of ALD. Although significant progress has been made in gaining better knowledge on the mechanisms and pathology of ALD, many features of ALD are unknown, and require further investigation, ideally with improved animal models that more effectively mimic human ALD. Although differences in the degree and stages of alcoholic liver injury inevitably exist between animal models and human ALD, the acquisition and translational relevance will be greatly enhanced with the development of new and improved animal models of ALD.
Collapse
Affiliation(s)
- Arantza Lamas-Paz
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Fengjie Hao
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Leonard J Nelson
- Institute for Bioengineering (IBioE), School of Engineering, Faraday Building, The University of Edinburgh, Edinburgh EH9 3 JL, Scotland, United Kingdom
| | - Maria Teresa Vázquez
- Department of Human Anatomy and Embryology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Santiago Canals
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas, Universidad Miguel Hernández, San Juan de Alicante 03550, Spain
| | - Manuel Gómez del Moral
- Department of Cell Biology, Complutense University School of Medicine, Madrid 28040, Spain
| | - Eduardo Martínez-Naves
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
- Yulia A Nevzovova, Francisco Javier Cubero, 12 de Octubre Health Research Institute (imas12), Madrid 28041, Spain
| | - Yulia A Nevzorova
- Department of Genetics, Physiology and Microbiology, Faculty of Biology, Universidad Complutense, Madrid 28040, Spain
- Department of Internal Medicine III, University Hospital RWTH Aachen, Aachen 52062, Germany
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ORL, Complutense University School of Medicine, Madrid 28040, Spain
| |
Collapse
|
15
|
Wang J, Xu S, Gao J, Zhang L, Zhang Z, Yang W, Li Y, Liao S, Zhou H, Liu P, Liang B. SILAC-based quantitative proteomic analysis of the livers of spontaneous obese and diabetic rhesus monkeys. Am J Physiol Endocrinol Metab 2018; 315:E294-E306. [PMID: 29664677 DOI: 10.1152/ajpendo.00016.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a severe metabolic disorder that affects more than 10% of the population worldwide. Obesity is a major cause of insulin resistance and contributes to the development of T2DM. Liver is an essential metabolic organ that plays crucial roles in the pathogenesis of obesity and diabetes. However, the underlying mechanisms of liver in the transition of obesity to diabetes are not fully understood. The nonhuman primate rhesus monkey is an appropriate animal for research of human diseases. Here, we first screened and selected three individuals of spontaneously diabetic rhesus monkeys. Interestingly, the diabetic monkeys were obese with a high body mass index at the beginning, but gradually lost their body weight during one year of observation. Furthermore, we performed stable isotope labeling with amino acids in cell culture-based quantitative proteomics to identify proteins and signaling pathways with altered expression in the liver of obese and diabetic monkeys. In total, 3,509 proteins were identified and quantified, of which 185 proteins displayed an altered expression level. Gene ontology analysis revealed that the expression of proteins involved in fatty acids β-oxidation and galactose metabolism was increased in obese monkeys; while proteins involved in oxidative phosphorylation and branched chain amino acid (BCAA) degradation were upregulated in diabetic monkeys. In addition, we observed mild apoptosis in the liver of diabetic monkeys, suggesting liver injury at the late onset of diabetes. Taken together, our liver proteomics may reveal a distinct metabolic transition from fatty acids β-oxidation in obese monkey to BCAA degradation in diabetic monkeys.
Collapse
Affiliation(s)
- Junlong Wang
- College of Pharmaceutical Sciences, Soochow University , Suzhou , China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Jing Gao
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai , China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Wenhui Yang
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Department of Geriatrics, Yan'an Affiliated Hospital of Kunming Medical University , Kunming , China
| | - Yunhai Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Shasha Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences , Shanghai , China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences , Beijing , China
| | - Bin Liang
- College of Pharmaceutical Sciences, Soochow University , Suzhou , China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences , Kunming , China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences , Kunming , China
| |
Collapse
|
16
|
You Y, Li WZ, Zhang S, Hu B, Li YX, Li HD, Tang HH, Li QW, Guan YY, Liu LX, Bao WL, Shen X. SNX10 mediates alcohol-induced liver injury and steatosis by regulating the activation of chaperone-mediated autophagy. J Hepatol 2018; 69:129-141. [PMID: 29452206 DOI: 10.1016/j.jhep.2018.01.038] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/03/2018] [Accepted: 01/30/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Alcoholic liver disease (ALD) is a major cause of morbidity and mortality worldwide. However, the cellular defense mechanisms underlying ALD are not well understood. Recent studies highlighted the involvement of chaperone-mediated autophagy (CMA) in regulating hepatic lipid metabolism. Sorting nexin (SNX)-10 has a regulatory function in endolysosomal trafficking and stabilisation. Here, we investigated the roles of SNX10 in CMA activation and in the pathogenesis of alcohol-induced liver injury and steatosis. METHODS Snx10 knockout (Snx10 KO) mice and their wild-type (WT) littermates fed either the Lieber-DeCarli liquid alcohol diet or a control liquid diet, and primary cultured WT and Snx10 KO hepatocytes stimulated with ethanol, were used as in vivo and in vitro ALD models, respectively. Activation of CMA, liver injury parameters, inflammatory cytokines, oxidative stress and lipid metabolism were measured. RESULTS Compared with WT littermates, Snx10 KO mice exhibited a significant amelioration in ethanol-induced liver injury and hepatic steatosis. Both in vivo and in vitro studies showed that SNX10 deficiency upregulated lysosome-associated membrane protein type 2A (LAMP-2A) expression and CMA activation, which could be reversed by SNX10 overexpression in vitro. LAMP-2A interference confirmed that the upregulation of Nrf2 and AMPK signalling pathways induced by SNX10 deficiency relied on CMA activation. Pull-down assays revealed an interaction between SNX10 and cathepsin A (CTSA), a key enzyme involved in LAMP-2A degradation. Deficiency in SNX10 inhibited CTSA maturation and increased the stability of LAMP-2A, resulting in an increase in CMA activity. CONCLUSIONS SNX10 controls CMA activity by mediating CTSA maturation, and, thus, has an essential role in alcohol-induced liver injury and steatosis. Our results provide evidence for SNX10 as a potential promising therapeutic target for preventing or ameliorating liver injury in ALD. LAY SUMMARY Alcoholic liver disease is a major cause of morbidity and mortality worldwide. Recent studies highlight the involvement of chaperone-mediated autophagy (CMA) in regulating hepatic lipid metabolism. Our study reveals that deficiency of sorting nexin (SNX) 10 increases the stability of LAMP-2A by inhibiting cathepsin A maturation, resulting in the increase of CMA activity and, thus, alleviates alcohol-induced liver injury and steatosis.
Collapse
Affiliation(s)
- Yan You
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wan-Zhen Li
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Sulin Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Bin Hu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yue-Xuan Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hai-Dong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Huan-Huan Tang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Qian-Wen Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yun-Yun Guan
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Li-Xin Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei-Lian Bao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Bertola A. WITHDRAWN: Rodent models of fatty liver diseases. LIVER RESEARCH 2018. [DOI: 10.1016/j.livres.2018.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Chen YX, Tao SC, Xu ZL, Yin WJ, Zhang YL, Yin JH, Gao YS, Zhang CQ. Novel Akt activator SC-79 is a potential treatment for alcohol-induced osteonecrosis of the femoral head. Oncotarget 2018; 8:31065-31078. [PMID: 28415692 PMCID: PMC5458189 DOI: 10.18632/oncotarget.16075] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Accepted: 02/28/2017] [Indexed: 01/20/2023] Open
Abstract
Alcohol is a leading risk factor for osteonecrosis of the femoral head (ONFH). We explored the molecular mechanisms underlying alcohol-induced ONFH and investigated the protective effect of the novel Akt activator SC-79 against this disease. We found that ethanol inhibited expression of the osteogenic genes RUNX2 and OCN, downregulated osteogenic differentiation, impaired the recruitment of Akt to the plasma membrane, and suppressed Akt phosphorylation at Ser473, thereby inhibiting the Akt/GSK3β/β-catenin signaling pathway in bone mesenchymal stem cells. To assess SC-79′s ability to counteract the inhibitory effect of ethanol on Akt-Ser73 phosphorylation, we performed micro-computerized tomography and immunofluorescent staining of osteopontin, osteocalcin and collagen type 1 in a rat model of alcohol-induced ONFH. We found that SC-79 injections inhibited alcohol-induced osteonecrosis. These results show that alcohol-induced ONFH is associated with suppression of p-Akt-Ser473 in the Akt/GSK3β/β-catenin signaling pathway in bone mesenchymal stem cells. We propose that SC-79 treatment to rescue Akt activation could be tested in the clinic as a potential therapeutic approach to preventing the development of alcohol-induced ONFH.
Collapse
Affiliation(s)
- Yi-Xuan Chen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Shi-Cong Tao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Zheng-Liang Xu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wen-Jing Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yue-Lei Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jun-Hui Yin
- Institute of Microsurgery on Extremities, Shanghai 200233, China
| | - You-Shui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.,Institute of Microsurgery on Extremities, Shanghai 200233, China
| |
Collapse
|
19
|
|
20
|
Thomsen KL, De Chiara F, Rombouts K, Vilstrup H, Andreola F, Mookerjee RP, Jalan R. Ammonia: A novel target for the treatment of non-alcoholic steatohepatitis. Med Hypotheses 2018. [PMID: 29523305 DOI: 10.1016/j.mehy.2018.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a spectrum of liver diseases ranging from steatosis, through non-alcoholic steatohepatitis (NASH) to cirrhosis. The development of fibrosis is the most important factor contributing to NASH-associated morbidity and mortality. Hepatic stellate cells (HSCs) are responsible for extracellular matrix deposition in conditions of frank hepatocellular injury and are key cells involved in the development of fibrosis. In experimental models and patients with NASH, urea cycle enzyme gene and protein expression is reduced resulting in functional reduction in the in vivo capacity for ureagenesis and subsequent hyperammonemia at a pre-cirrhotic stage. Ammonia has been shown to activate HSCs in vivo and in vitro. Hyperammonemia in the context of NASH may therefore favour the progression of fibrosis and the disease. We therefore hypothesise that ammonia is a potential target for prevention of fibrosis progression of patients with NASH.
Collapse
Affiliation(s)
- Karen Louise Thomsen
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom; Department of Hepatology & Gastroenterology, Aarhus University Hospital, Denmark
| | - Francesco De Chiara
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom
| | - Krista Rombouts
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom
| | - Hendrik Vilstrup
- Department of Hepatology & Gastroenterology, Aarhus University Hospital, Denmark
| | - Fausto Andreola
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom
| | - Rajeshwar P Mookerjee
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom
| | - Rajiv Jalan
- UCL Institute for Liver and Digestive Health, University College London, United Kingdom.
| |
Collapse
|
21
|
NaoXinTong Enhances Atorvastatin-induced Plaque Stability While Ameliorating Atorvastatin-induced Hepatic Inflammation. J Cardiovasc Pharmacol 2017; 69:55-64. [PMID: 27828790 DOI: 10.1097/fjc.0000000000000441] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Buchang NaoXinTong (NXT) is a Chinese medicine that has been used for many years for treatment of patients with coronary heart disease (CHD) in China. Statins substantially reduce hypercholesterolemia and CHD mortality and morbidity. However, there is still a lot of CHD patients who do not respond well to statin therapy. Herein, we report the effects of NXT on atorvastatin-inhibited atherosclerosis and atorvastatin-induced hepatic side effects. After 10 weeks of high-fat diet (HFD) feeding, apoE-deficient mice were randomly divided into 4 groups and received the following treatment for another 8 weeks: group 1, HFD; group 2, HFD containing NXT; group 3, HFD containing atorvastatin; and group 4, HFD containing both NXT and atorvastatin. After treatment, serum lipid profiles, atherosclerotic lesions, and hepatic lipid content and inflammation were determined. NXT moderately increased high-density lipoprotein cholesterol levels, although had little effect on atorvastatin-induced reduction of low-density lipoprotein cholesterol levels. Both NXT and atorvastatin reduced en face lesions and sinus lesions of aortic root. In addition, NXT enhanced atorvastatin-induced lesion plaque stability by increasing smooth muscle cell/collagen content and reducing macrophage accumulation and calcification in lesion areas. The co-treatment of NXT and atorvastatin further reduced hepatic triglyceride levels by downregulating acyl-CoA:diacylglycerol acyltransferase 1 while activating hormone-sensitive lipase, adipose triglyceride lipase, and comparative gene identification-58 expression. The AMPKα pathway was also further activated by the co-treatment. More importantly, the liver injuries caused by atorvastatin, such as hepatic inflammation and elevated serum aminotransferase activities, were substantially attenuated by NXT. Therefore, our study demonstrates that NXT enhances atorvastatin-induced plaque stability and ameliorates atorvastatin-induced hepatic side effects.
Collapse
|
22
|
Avila DV, Barker DF, Zhang J, McClain CJ, Barve S, Gobejishvili L. Dysregulation of hepatic cAMP levels via altered Pde4b expression plays a critical role in alcohol-induced steatosis. J Pathol 2017; 240:96-107. [PMID: 27287961 DOI: 10.1002/path.4760] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 05/06/2016] [Accepted: 05/31/2016] [Indexed: 12/15/2022]
Abstract
Alcohol-induced hepatic steatosis is a significant risk factor for progressive liver disease. Cyclic adenosine monophosphate (cAMP) signalling has been shown to significantly regulate lipid metabolism; however, the role of altered cAMP homeostasis in alcohol-mediated hepatic steatosis has never been studied. Our previous work demonstrated that increased expression of hepatic phosphodiesterase 4 (Pde4), which specifically hydrolyses and decreases cAMP levels, plays a pathogenic role in the development of liver inflammation/injury. The aim of this study was to examine the role of PDE4 in alcohol-induced hepatic steatosis. C57BL/6 wild-type and Pde4b knockout (Pde4b(-/-) ) mice were pair-fed control or ethanol liquid diets. One group of wild-type mice received rolipram, a PDE4-specific inhibitor, during alcohol feeding. We demonstrate for the first time that an early increase in PDE4 enzyme expression and a resultant decrease in hepatic cAMP levels are associated with the significant reduction in carnitine palmitoyltransferase 1A (Cpt1a) expression. Notably, alcohol-fed (AF) Pde4b(-/-) mice and AF wild-type mice treated with rolipram had significantly lower hepatic free fatty acid content compared with AF wild-type mice. Importantly, PDE4 inhibition in alcohol-fed mice prevented the decrease in hepatic Cpt1a expression via the Pparα/Sirt1/Pgc1α pathway. These results demonstrate that the alcohol- induced increase in hepatic Pde4, specifically Pde4b expression, and compromised cAMP signalling predispose the liver to impaired fatty acid oxidation and the development of steatosis. Moreover, these data also suggest that hepatic PDE4 may be a clinically relevant therapeutic target for the treatment of alcohol-induced hepatic steatosis. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Diana V Avila
- Department of Pharmacology and Toxicology, University of Louisville Medical Center, Louisville, Kentucky, USA
| | - David F Barker
- Department of Internal Medicine, University of Louisville Medical Center, Louisville, Kentucky, USA
| | - JingWen Zhang
- Department of Internal Medicine, University of Louisville Medical Center, Louisville, Kentucky, USA
| | - Craig J McClain
- Department of Pharmacology and Toxicology, University of Louisville Medical Center, Louisville, Kentucky, USA.,Department of Internal Medicine, University of Louisville Medical Center, Louisville, Kentucky, USA.,Robley Rex VA Medical Center, Louisville, Kentucky, USA
| | - Shirish Barve
- Department of Pharmacology and Toxicology, University of Louisville Medical Center, Louisville, Kentucky, USA.,Department of Internal Medicine, University of Louisville Medical Center, Louisville, Kentucky, USA
| | - Leila Gobejishvili
- Department of Pharmacology and Toxicology, University of Louisville Medical Center, Louisville, Kentucky, USA.,Department of Internal Medicine, University of Louisville Medical Center, Louisville, Kentucky, USA
| |
Collapse
|
23
|
Cho YE, Im EJ, Moon PG, Mezey E, Song BJ, Baek MC. Increased liver-specific proteins in circulating extracellular vesicles as potential biomarkers for drug- and alcohol-induced liver injury. PLoS One 2017; 12:e0172463. [PMID: 28225807 PMCID: PMC5321292 DOI: 10.1371/journal.pone.0172463] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/06/2017] [Indexed: 12/21/2022] Open
Abstract
Drug- and alcohol-induced liver injury are a leading cause of liver failure and transplantation. Emerging evidence suggests that extracellular vesicles (EVs) are a source of biomarkers because they contain unique proteins reflecting the identity and tissue-specific origin of the EV proteins. This study aimed to determine whether potentially hepatotoxic agents, such as acetaminophen (APAP) and binge alcohol, can increase the amounts of circulating EVs and evaluate liver-specific EV proteins as potential biomarkers for liver injury. The circulating EVs, isolated from plasma of APAP-exposed, ethanol-fed mice, or alcoholic hepatitis patients versus normal control counterparts, were characterized by proteomics and biochemical methods. Liver specific EV proteins were analyzed by immunoblots and ELISA. The amounts of total and liver-specific proteins in circulating EVs from APAP-treated mice significantly increased in a dose- and time-dependent manner. Proteomic analysis of EVs from APAP-exposed mice revealed that the amounts of liver-specific and/or hepatotoxic proteins were increased compared to those of controls. Additionally, the increased protein amounts in EVs following APAP exposure returned to basal levels when mice were treated with N-acetylcysteine or glutathione. Similar results of increased amounts and liver-specific proteins in circulating EVs were also observed in mice exposed to hepatotoxic doses of thioacetamide or d-galactosamine but not by non-hepatotoxic penicillin or myotoxic bupivacaine. Additionally, binge ethanol exposure significantly elevated liver-specific proteins in circulating EVs from mice and alcoholics with alcoholic hepatitis, compared to control counterparts. These results indicate that circulating EVs in drug- and alcohol-mediated hepatic injury contain liver-specific proteins that could serve as specific biomarkers for hepatotoxicity.
Collapse
Affiliation(s)
- Young-Eun Cho
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Eun-Ju Im
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Pyong-Gon Moon
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Esteban Mezey
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry and Biophysics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, Maryland, United States of America
| | - Moon-Chang Baek
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- * E-mail:
| |
Collapse
|
24
|
Gonçalves JL, Lacerda-Queiroz N, Sabino JF, Marques PE, Galvão I, Gamba CO, Cassali GD, de Carvalho LM, da Silva e Silva DA, Versiani A, Teixeira MM, de Faria AMC, Vieira AT, Brunialti-Godard AL. Evaluating the effects of refined carbohydrate and fat diets with acute ethanol consumption using a mouse model of alcoholic liver injury. J Nutr Biochem 2017; 39:93-100. [DOI: 10.1016/j.jnutbio.2016.08.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/13/2016] [Accepted: 08/10/2016] [Indexed: 02/07/2023]
|
25
|
Choi JW, Kim IH, Kim YM, Lee MK, Choi YH, Nam TJ. Protective effect of Pyropia yezoensis glycoprotein on chronic ethanol consumption-induced hepatotoxicity in rats. Mol Med Rep 2016; 14:4881-4886. [PMID: 27748873 DOI: 10.3892/mmr.2016.5820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/20/2016] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the protective effect of Pyropia yezoensis glycoprotein (PYGP) against chronic ethanol consumption‑mediated hepatotoxicity in rats. Male Sprague-Dawley rats (n=20; 6 weeks old) were randomly divided into four groups. The rats in each group were treated for 30 days with the following: i) CON group, distilled water only; ii) EtOH group, 20% ethanol 3.7 g/kg/BW; iii) EtOH+150 group, 20% ethanol 3.7 g/kg/BW+PYGP 150 mg/kg/BW; iv) EtOH+300 group, 20% ethanol 3.7 g/kg/BW+PYGP 300 mg/kg/BW. EtOH, PYGP and water were orally administered. The rats were sacrificed after 30 days, and blood and liver samples were collected for analysis. Treatment with ethanol caused significant elevation of serum levels of glutamic oxaloacetic transaminase (GOT) and glutamic pyruvic transaminase (GPT). Furthermore, inhibition of the antioxidant defense system in the liver, including glutathione (GSH), glutathione peroxidase (GSH‑px) and catalase (CAT) was observed. However, co‑administration with PYGP recovered the antioxidant defense system, and the serum levels of GOT and GPT. PYGP was shown to attenuate ethanol toxicity via the inactivation of mitogen‑activated protein kinases (MAKPs). PYGP suppressed the overexpression of cytochrome P450 2E1 (CYP2E1), inducible nitric oxide synthase and cyclooxygenase‑2. These results suggested that the protective effect of PYGP was associated with antioxidant activities, MAPKs and the CYP2E1 signaling pathway.
Collapse
Affiliation(s)
- Jeong-Wook Choi
- Department of Food and Life Science, Pukyong National University, Busan 608‑737, Republic of Korea
| | - In-Hye Kim
- Institute of Fisheries Science, Pukyong National University, Busan 619‑911, Republic of Korea
| | - Young-Min Kim
- Department of Food and Life Science, Pukyong National University, Busan 608‑737, Republic of Korea
| | - Min-Kyeong Lee
- Department of Food and Life Science, Pukyong National University, Busan 608‑737, Republic of Korea
| | - Youn-Hee Choi
- Institute of Fisheries Science, Pukyong National University, Busan 619‑911, Republic of Korea
| | - Taek-Jeong Nam
- Department of Food and Life Science, Pukyong National University, Busan 608‑737, Republic of Korea
| |
Collapse
|
26
|
Chen LY, Chen Q, Zhu XJ, Kong DS, Wu L, Shao JJ, Zheng SZ. Diallyl trisulfide protects against ethanol-induced oxidative stress and apoptosis via a hydrogen sulfide-mediated mechanism. Int Immunopharmacol 2016; 36:23-30. [DOI: 10.1016/j.intimp.2016.04.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/01/2016] [Accepted: 04/12/2016] [Indexed: 01/25/2023]
|
27
|
Cho I, Kim J, Jung J, Sung S, Kim J, Lee N, Ku S. Hepatoprotective effects of hoveniae semen cum fructus extracts in ethanol intoxicated mice. J Exerc Nutrition Biochem 2016; 20:49-64. [PMID: 27298813 PMCID: PMC4899896 DOI: 10.20463/jenb.2016.03.20.1.4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 12/17/2022] Open
Abstract
[Purpose] The objective of this study was to evaluate the hepatoprotective effects of Hoveniae Semen Cum Fructus extract in ethanol induced hepatic damages. [Methods] Hepatic damages were induced by oral administration of ethanol and then Hoveniae Semen Cum Fructus extract was administered. [Results] Following Hoveniae Semen Cum Fructus extract administration, body and liver weights were increased, while aspartate aminotransferase, alanine aminotransferase, albumin, γ-glutamyl transferase, and triglyceride levels in the serum, triglyceride contents, tumor necrosis factor -α level, cytochrome (CY) P450 2E1 activity in the liver and mRNA expression of hepatic lipogenic genes, and Nitrotyrosine and 4-HNE-immunolabelled hepatocytes were decreased. However, mRNA expression of genes involved in fatty acid oxidation was increased. Also, as a protective mechanism for hepatic antioxidant defense systems, decreased liver MDA contents, increased glutathione contents, increased dismutase and catalase activities were observed when compared to the ethanol control. [Conclusion] Hoveniae Semen Cum Fructus extract favorably protected against liver damages, mediated by its potent anti-inflammatory and anti-steatosis properties through the augmentation of the hepatic antioxidant defense system by NF-E2-related factor-2 activation, and down-regulation of the mRNA expression of hepatic lipogenic genes or up-regulation of the mRNA expression of genes involved in fatty acid oxidation.
Collapse
Affiliation(s)
- Ilje Cho
- Department of Anatomy and Histology, Daegu Haany University, Gyeongsan-si Republic of Korea
| | - Joowan Kim
- Aribio Central Research Institute, Aribio Inc., Sungnam-si Republic of Korea
| | - Jaijun Jung
- Aribio Central Research Institute, Aribio Inc., Sungnam-si Republic of Korea
| | - Soohyun Sung
- Aribio Central Research Institute, Aribio Inc., Sungnam-si Republic of Korea
| | - Jongkyu Kim
- Aribio Central Research Institute, Aribio Inc., Sungnam-si Republic of Korea
| | - Namju Lee
- Department of Sports Medicine, Jungwon University, Goesan-gun Republic of Korea
| | - Saekwang Ku
- Department of Anatomy and Histology, Daegu Haany University, Gyeongsan-si Republic of Korea
| |
Collapse
|
28
|
Lee YJ, Zhao RJ, Kim YW, Kang SJ, Lee EK, Kim NJ, Chang S, Kim JM, Lee JE, Ku SK, Lee BH. Acupuncture inhibits liver injury induced by morphine plus acetaminophen through antioxidant system. Eur J Integr Med 2016. [DOI: 10.1016/j.eujim.2015.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Argininosuccinate synthetase regulates hepatic AMPK linking protein catabolism and ureagenesis to hepatic lipid metabolism. Proc Natl Acad Sci U S A 2016; 113:E3423-30. [PMID: 27247419 DOI: 10.1073/pnas.1606022113] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A key sensor of cellular energy status, AMP-activated protein kinase (AMPK), interacts allosterically with AMP to maintain an active state. When active, AMPK triggers a metabolic switch, decreasing the activity of anabolic pathways and enhancing catabolic processes such as lipid oxidation to restore the energy balance. Unlike oxidative tissues, in which AMP is generated from adenylate kinase during states of high energy demand, the ornithine cycle enzyme argininosuccinate synthetase (ASS) is a principle site of AMP generation in the liver. Here we show that ASS regulates hepatic AMPK, revealing a central role for ureagenesis flux in the regulation of metabolism via AMPK. Treatment of primary rat hepatocytes with amino acids increased gluconeogenesis and ureagenesis and, despite nutrient excess, induced both AMPK and acetyl-CoA carboxylase (ACC) phosphorylation. Antisense oligonucleotide knockdown of hepatic ASS1 expression in vivo decreased liver AMPK activation, phosphorylation of ACC, and plasma β-hydroxybutyrate concentrations. Taken together these studies demonstrate that increased amino acid flux can activate AMPK through increased AMP generated by ASS, thus providing a novel link between protein catabolism, ureagenesis, and hepatic lipid metabolism.
Collapse
|
30
|
Song X, Yin S, Huo Y, Liang M, Fan L, Ye M, Hu H. Glycycoumarin ameliorates alcohol-induced hepatotoxicity via activation of Nrf2 and autophagy. Free Radic Biol Med 2015; 89:135-46. [PMID: 26169726 DOI: 10.1016/j.freeradbiomed.2015.07.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/24/2015] [Accepted: 07/07/2015] [Indexed: 02/06/2023]
Abstract
Licorice, a traditional Chinese medicine, has been used to treat various diseases, including liver disease, for centuries. However, the chemical basis and biological mechanisms underlying the biological functions of licorice remain elusive. The purpose of the current study was to test the hepatoprotective effect of glycycoumarin (GCM), a representative coumarin in licorice, using animal models of both chronic and acute alcoholic liver injury. C57BL/6J mice were used to evaluate the hepatoprotective effect of GCM on liver injury induced by either chronic or acute ethanol exposure. AML-12 and HepG2 cells were utilized to determine the functional role of Nrf2 in the hepatoprotective effect of GCM and to decipher the mechanisms of GCM-induced Nrf2 activation. We found that treatment with GCM leads to a significant reduction in hepatotoxicity in response to either chronic or acute ethanol exposure. Further mechanistic investigations reveal that activation of Nrf2 via the p38 pathway and induction of autophagy by GCM contribute to its hepatoprotective activity. In addition, we demonstrate that p62 upregulation by a transcriptional mechanism also contributes to Nrf2 activation via a positive feedback loop. Our study has identified GCM as a novel active ingredient that contributes to the hepatoprotective activity of licorice.
Collapse
Affiliation(s)
- Xinhua Song
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Shutao Yin
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yazhen Huo
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Min Liang
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Lihong Fan
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Min Ye
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Hongbo Hu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
31
|
Shearn CT, Fritz KS, Shearn AH, Saba LM, Mercer KE, Engi B, Galligan JJ, Zimniak P, Orlicky DJ, Ronis MJ, Petersen DR. Deletion of GSTA4-4 results in increased mitochondrial post-translational modification of proteins by reactive aldehydes following chronic ethanol consumption in mice. Redox Biol 2015; 7:68-77. [PMID: 26654979 PMCID: PMC4683459 DOI: 10.1016/j.redox.2015.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 12/21/2022] Open
Abstract
Chronic alcohol consumption induces hepatic oxidative stress resulting in production of highly reactive electrophilic α/β-unsaturated aldehydes that have the potential to modify proteins. A primary mechanism of reactive aldehyde detoxification by hepatocytes is through GSTA4-driven enzymatic conjugation with GSH. Given reports that oxidative stress initiates GSTA4 translocation to the mitochondria, we hypothesized that increased hepatocellular damage in ethanol (EtOH)-fed GSTA4−/− mice is due to enhanced mitochondrial protein modification by reactive aldehydes. Chronic ingestion of EtOH increased hepatic protein carbonylation in GSTA4−/− mice as evidenced by increased 4-HNE and MDA immunostaining in the hepatic periportal region. Using mass spectrometric analysis of biotin hydrazide conjugated carbonylated proteins, a total of 829 proteins were identified in microsomal, cytosolic and mitochondrial fractions. Of these, 417 were novel to EtOH models. Focusing on mitochondrial fractions, 1.61-fold more carbonylated proteins were identified in EtOH-fed GSTA4−/− mice compared to their respective WT mice ingesting EtOH. Bioinformatic KEGG pathway analysis of carbonylated proteins from the mitochondrial fractions revealed an increased propensity for modification of proteins regulating oxidative phosphorylation, glucose, fatty acid, glutathione and amino acid metabolic processes in GSTA4−/− mice. Additional analysis revealed sites of reactive aldehyde protein modification on 26 novel peptides/proteins isolated from either SV/GSTA4−/− PF or EtOH fed mice. Among the peptides/proteins identified, ACSL, ACOX2, MTP, and THIKB contribute to regulation of fatty acid metabolism and ARG1, ARLY, and OAT, which regulate nitrogen and ammonia metabolism having direct relevance to ethanol-induced liver injury. These data define a role for GSTA4-4 in buffering hepatic oxidative stress associated with chronic alcohol consumption and that this GST isoform plays an important role in protecting against carbonylation of mitochondrial proteins. We demonstrate increased mitochondrial carbonylation in GSTA4-4 KO mice chronically fed EtOH. Using LC-MS we identify 829 total carbonylated proteins (417 novel to murine ALD). Pathway analysis revealed a propensity for adduction of fatty acid metabolic and electron transport proteins. Using MS/MS, 26 novel adducted peptides were identified. Reactive aldehyde modification of proteins contributes to pathogenesis of ALD.
Collapse
Affiliation(s)
- Colin T Shearn
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.
| | - Kristofer S Fritz
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | | | - Laura M Saba
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Kelly E Mercer
- Department of Pediatrics, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - Bridgette Engi
- Department of Pediatrics, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - James J Galligan
- Department of Biochemistry, Vanderbilt, Nashville, TN, United States
| | - Piotr Zimniak
- Department of Pediatrics, Arkansas Children's Nutrition Center, Little Rock, AR, United States
| | - David J Orlicky
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Center, Aurora, CO, United States
| | - Martin J Ronis
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Dennis R Petersen
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
32
|
Momen-Heravi F, Saha B, Kodys K, Catalano D, Satishchandran A, Szabo G. Increased number of circulating exosomes and their microRNA cargos are potential novel biomarkers in alcoholic hepatitis. J Transl Med 2015; 13:261. [PMID: 26264599 PMCID: PMC4533956 DOI: 10.1186/s12967-015-0623-9] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/29/2015] [Indexed: 12/13/2022] Open
Abstract
Background It has been well documented that alcohol and its metabolites induce injury and inflammation in the liver. However, there is no potential biomarker to monitor the extent of liver injury in alcoholic hepatitis patients. MicroRNAs (miRNAs) are a class of non-coding RNAs that are involved in various physiologic and pathologic processes. In the circulation, a great proportion of miRNAs is associated with extracellular vesicles (EVs)/exosomes. Here, we hypothesized that the exosome-associated miRNAs can be used as potential biomarkers in alcoholic hepatitis (AH). Methods Exosomes were isolated from sera of alcohol-fed mice or pair-fed mice, and plasma of alcoholic hepatitis patients or healthy controls by ExoQuick. The exosomes were characterized by transmission electron microscopy and Western blot and enumerated with a Nanoparticle Tracking Analysis system. Firefly™ microRNA Assay was performed on miRNA extracted from mice sera. TaqMan microRNA assay was used to identify differentially expressed miRNAs in plasma of cohort of patients with AH versus controls followed by construction of receiver operating characteristic (ROC) curves to determine the sensitivity and specificity of the candidates. Results The total number of circulating EVs was significantly increased in mice after alcohol feeding. Those EVs mainly consisted of exosomes, the smaller size vesicle subpopulation of EVs. By performing microarray screening on exosomes, we found nine inflammatory miRNAs which were deregulated in sera of chronic alcohol-fed mice compared to controls including upregulated miRNAs: miRNA-192, miRNA-122, miRNA-30a, miRNA-744, miRNA-1246, miRNA 30b and miRNA-130a. The ROC analyses indicated excellent diagnostic value of miRNA-192, miRNA-122, and miRNA-30a to identify alcohol-induced liver injury. We further validated findings from our animal model in human samples. Consistent with the animal model, total number of EVs, mostly exosomes, was significantly increased in human subjects with AH. Both miRNA-192 and miRNA-30a were significantly increased in the circulation of subjects with AH. miRNA-192 showed promising value for the diagnosis of AH. Conclusion Elevated level of EVs/exosomes and exosome-associated miRNA signature could serve as potential diagnostic markers for AH. In addition to the biomarker diagnostic capabilities, these findings may facilitate development of novel strategies for diagnostics, monitoring, and therapeutics of AH. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0623-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fatemeh Momen-Heravi
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Banishree Saha
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Karen Kodys
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Donna Catalano
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Abhishek Satishchandran
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| | - Gyongyi Szabo
- Department of Medicine, University of Massachusetts Medical School, LRB208, 364 Plantation Street, Worcester, MA, 01605, USA.
| |
Collapse
|
33
|
Xing H, Jia K, He J, Shi C, Fang M, Song L, Zhang P, Zhao Y, Fu J, Li S. Establishment of the tree shrew as an alcohol-induced Fatty liver model for the study of alcoholic liver diseases. PLoS One 2015; 10:e0128253. [PMID: 26030870 PMCID: PMC4451149 DOI: 10.1371/journal.pone.0128253] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/23/2015] [Indexed: 12/17/2022] Open
Abstract
Currently, the pathogenesis of alcoholic liver diseases (ALDs) is not clear. As a result, there is no effective treatment for ALDs. One limitation is the lack of a suitable animal model for use in studying ALDs. The tree shrew is a lower primate animal, characterized by a high-alcohol diet. This work aimed to establish a fatty liver model using tree shrews and to assess the animals' suitability for the study of ALDs. Tree shrews were treated with alcohol solutions (10% and 20%) for two weeks. Hemophysiology, blood alcohol concentrations (BACs), oxidative stress factors, alcohol metabolic enzymes and hepatic pathology were checked and assayed with an automatic biochemical analyzer, enzyme-linked immunosorbent assay (ELISA), western blot, hematoxylin-eosin (HE) staining and oil red O staining, and magnetic resonance imaging (MRI). Compared with the normal group, the levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transpeptidase (GGT), total cholesterol (TC), triglyceride (TG), reactive oxygen species (ROS), and malondialdehyde (MDA) were significantly enhanced in alcohol-treated tree shrews. However, the activity of reduced glutathione hormone (GSH) and superoxide dismutase (SOD) declined. Notable changes in alcohol dehydrogenase(ADH1), aldehyde dehydrogenase(ALDH2), CYP2E1, UDP-glucuronosyl transferase 1A1 (UGT1A1) and nuclear factor erythroid-related factor 2 (Nrf2) were observed. HE and oil red O staining showed that hepatocyte swelling, hydropic degeneration, and adipohepatic syndrome occurred in the tree shrews. Alcohol can induce fatty liver-like pathological changes and result in alterations in liver function, oxidative stress factors, alcohol metabolism enzymes and Nrf2. Therefore, the established fatty liver model of tree shrews induced by alcohol should be a promising tool for the study of ALDs.
Collapse
Affiliation(s)
- Huijie Xing
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, PR China
- Guangdong Provincial Key Laboratory for the Prevention and Control of Severe Clinical Animal Diseases, Guangzhou, Guangdong Province, PR China
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Kun Jia
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, PR China
- Guangdong Provincial Key Laboratory for the Prevention and Control of Severe Clinical Animal Diseases, Guangzhou, Guangdong Province, PR China
| | - Jun He
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Changzheng Shi
- Medical Imaging Center, the First Attached Hospital of Jinan University, Guangzhou, Guangdong Province, PR China
| | - Meixia Fang
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Linliang Song
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Pu Zhang
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Yue Zhao
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
| | - Jiangnan Fu
- Institute of Laboratory Animals, Jinan University, Guangzhou, Guangdong Province, PR China
- * E-mail: (SJL); (JNF)
| | - Shoujun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, PR China
- Guangdong Provincial Key Laboratory for the Prevention and Control of Severe Clinical Animal Diseases, Guangzhou, Guangdong Province, PR China
- * E-mail: (SJL); (JNF)
| |
Collapse
|
34
|
You M, Jogasuria A, Taylor C, Wu J. Sirtuin 1 signaling and alcoholic fatty liver disease. Hepatobiliary Surg Nutr 2015; 4:88-100. [PMID: 26005675 DOI: 10.3978/j.issn.2304-3881.2014.12.06] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 10/29/2014] [Indexed: 12/12/2022]
Abstract
Alcoholic fatty liver disease (AFLD) is one of the most prevalent forms of liver disease worldwide and can progress to inflammation (hepatitis), fibrosis/cirrhosis, and ultimately lead to end stage liver injury. The mechanisms, by which ethanol consumption leads to AFLD, are complicated and multiple, and remain incompletely understood. Nevertheless, understanding its pathogenesis will facilitate the development of effective pharmacological or nutritional therapies for treating human AFLD. Chronic ethanol consumption causes steatosis and inflammation in rodents or humans by disturbing several important hepatic transcriptional regulators, including AMP-activated kinase (AMPK), lipin-1, sterol regulatory element binding protein 1 (SREBP-1), PPARγ co-activator-1α (PGC-1α), and nuclear transcription factor-κB (NF-κB). Remarkably, the effects of ethanol on these regulators are mediated in whole or in part by inhibition of a central signaling molecule, sirtuin 1 (SIRT1), which is a nicotinamide adenine dinucleotide (NAD(+), NADH)-dependent class III protein deacetylase. In recent years, SIRT1 has emerged as a pivotal molecule controlling the pathways of hepatic lipid metabolism, inflammatory responses and in the development of AFLD in rodents and in humans. Ethanol-mediated SIRT1 inhibition suppresses or stimulates the activities of above described transcriptional regulators and co-regulators, thereby deregulating diverse lipid metabolism and inflammatory response pathways including lipogenesis, fatty acid β-oxidation, lipoprotein uptake and secretion and expression of pro-inflammatory cytokines in the liver. This review aims to highlight our current understanding of SIRT1 regulatory mechanisms and its response to ethanol-induced toxicity, thus, affirming significant role of SIRT1 signaling in the development of AFLD.
Collapse
Affiliation(s)
- Min You
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Alvin Jogasuria
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Charles Taylor
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Jiashin Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| |
Collapse
|
35
|
Szuba A, Kasprowicz-Maluśki A, Wojtaszek P. Nitration of plant apoplastic proteins from cell suspension cultures. J Proteomics 2015; 120:158-68. [PMID: 25805245 DOI: 10.1016/j.jprot.2015.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 12/27/2022]
Abstract
Nitric oxide causes numerous protein modifications including nitration of tyrosine residues. This modification, though one of the greatest biological importance, is poorly recognized in plants and is usually associated with stress conditions. In this study we analyzed nitrotyrosines from suspension cultures of Arabidopsis thaliana and Nicotiana tabacum, treated with NO modulators and exposed to osmotic stress, as well as of BY2 cells long-term adapted to osmotic stress conditions. Using confocal microscopy, we showed that the cell wall area is one of the compartments most enriched in nitrotyrosines within a plant cell. Subsequently, we analyzed nitration of ionically-bound cell-wall proteins and identified selected proteins with MALDI-TOF spectrometry. Proteomic analysis indicated that there was no significant increase in the amount of nitrated proteins under the influence of NO modulators, among them 3-morpholinosydnonimine (SIN-1), considered a donor of nitrating agent, peroxynitrite. Moreover, osmotic stress conditions did not increase the level of nitration in cell wall proteins isolated from suspension cells, and in cultures long-term adapted to stress conditions; that level was even reduced in comparison with control samples. Among identified nitrotyrosine-containing proteins dominated the ones associated with carbon circulation as well as the numerous proteins responding to stress conditions, mainly peroxidases. BIOLOGICAL SIGNIFICANCE High concentrations of nitric oxide found in the cell wall and the ability to produce large amounts of ROS make the apoplast a site highly enriched in nitrotyrosines, as presented in this paper. Analysis of ionically bound fraction of the cell wall proteins indicating generally unchanged amounts of nitrotyrosines under influence of NO modulators and osmotic stress, is noticeably different from literature data concerning, however, the total plant proteins analysis. This observation is supplemented by further nitroproteome analysis, for cells long-term adapted to stressful conditions, and results showing that such conditions did not always cause an increase in nitrotyrosine content. These findings may be interpreted as characteristic features of apoplastic protein nitration.
Collapse
Affiliation(s)
- Agnieszka Szuba
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznań, Poland; Institute of Dendrology, Polish Academy of Sciences, Parkowa 5, 62-035 Kórnik Poland.
| | - Anna Kasprowicz-Maluśki
- Department of Molecular and Cellular Biology, Adam Mickiewicz University, Umultowska 89, 61-613 Poznań, Poland
| | - Przemysław Wojtaszek
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznań, Poland; Department of Molecular and Cellular Biology, Adam Mickiewicz University, Umultowska 89, 61-613 Poznań, Poland
| |
Collapse
|
36
|
Zielińska M, Obara-Michlewska M, Hilgier W, Albrecht J. Citrulline uptake in rat cerebral cortex slices: modulation by Thioacetamide -Induced hepatic failure. Metab Brain Dis 2014; 29:1053-60. [PMID: 24385142 PMCID: PMC4234898 DOI: 10.1007/s11011-013-9472-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 12/13/2013] [Indexed: 11/01/2022]
Abstract
L-citrulline (Cit) is a co-product of NO synthesis and a direct L-arginine (Arg) precursor for de novo NO synthesis. Acute liver failure (ALF) is associated with increased nitric oxide (NO) and cyclic GMP (cGMP) synthesis in the brain, indirectly implicating a role for active transport of Cit. In the present study we characterized [(3)H]Cit uptake to the cortical brain slices obtained from control rats and rats with thioacetamide (TAA)-induced ALF ("TAA slices"). In both control and TAA slices the uptake was partially Na(+)-dependent and markedly inhibited by substrates of systems L and N, including L-glutamine (Gln), which accumulates in excess in brain during ALF. Cit uptake was not affected by Arg, the y(+)/y(+)L transport system substrate, nor by amino acids taken up by systems A, xc (-)or XAG. The Vmax of the uptake in TAA slices was ~60 % higher than in control slices. Chromatographic (HPLC) analysis revealed a ~30 % increase of Cit concentration in the cerebral cortical homogenates of TAA rats. The activity of argininosuccinate synthase (ASS) and argininosuccinate lyase (ASL), the two enzymes of Cit-NO cycle catalyzing synthesis of Arg, showed an increase in TAA rats, consistent with increased ASS and ASL protein expression, by ~30 and ~20 %, respectively. The increased Cit-NO cycle activity was paralleled by increased expression of mRNA coding for inducible nitric oxide synthase (iNOS). Taken together, the results suggest a role for Cit in the activation of cerebral NO synthesis during ALF.
Collapse
Affiliation(s)
- Magdalena Zielińska
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Marta Obara-Michlewska
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Wojciech Hilgier
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| | - Jan Albrecht
- Department of Neurotoxicology, Medical Research Centre, Polish Academy of Sciences, Pawinskiego 5, 02-106 Warsaw, Poland
| |
Collapse
|
37
|
Ge X, Antoine DJ, Lu Y, Arriazu E, Leung TM, Klepper AL, Branch AD, Fiel MI, Nieto N. High mobility group box-1 (HMGB1) participates in the pathogenesis of alcoholic liver disease (ALD). J Biol Chem 2014; 289:22672-22691. [PMID: 24928512 PMCID: PMC4132775 DOI: 10.1074/jbc.m114.552141] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 06/05/2014] [Indexed: 12/16/2022] Open
Abstract
Growing clinical and experimental evidence suggests that sterile inflammation contributes to alcoholic liver disease (ALD). High mobility group box-1 (HMGB1) is highly induced during liver injury; however, a link between this alarmin and ALD has not been established. Thus, the aim of this work was to determine whether HMGB1 contributes to the pathogenesis of ALD. Liver biopsies from patients with ALD showed a robust increase in HMGB1 expression and translocation, which correlated with disease stage, compared with healthy explants. Similar findings were observed in chronic ethanol-fed wild-type (WT) mice. Using primary cell culture, we validated the ability of hepatocytes from ethanol-fed mice to secrete a large amount of HMGB1. Secretion was time- and dose-dependent and responsive to prooxidants and antioxidants. Selective ablation of Hmgb1 in hepatocytes protected mice from alcohol-induced liver injury due to increased carnitine palmitoyltransferase-1, phosphorylated 5'AMP-activated protein kinase-α, and phosphorylated peroxisome proliferator-activated receptor-α expression along with elevated LDL plus VLDL export. Native and post-translationally modified HMGB1 were detected in humans and mice with ALD. In liver and serum from control mice and in serum from healthy volunteers, the lysine residues within the peptides containing nuclear localization signals (NLSs) 1 and 2 were non-acetylated, and all cysteine residues were reduced. However, in livers from ethanol-fed mice, in addition to all thiol/non-acetylated isoforms of HMGB1, we observed acetylated NLS1 and NLS2, a unique phosphorylation site in serine 35, and an increase in oxidation of HMGB1 to the disulfide isoform. In serum from ethanol-fed mice and from patients with ALD, there was disulfide-bonded hyperacetylated HMGB1, disulfide-bonded non-acetylated HMGB1, and HMGB1 phosphorylated in serine 35. Hepatocytes appeared to be a major source of these HMGB1 isoforms. Thus, hepatocyte HMGB1 participates in the pathogenesis of ALD and undergoes post-translational modifications (PTMs) that could condition its toxic effects.
Collapse
MESH Headings
- Acetylation
- Animals
- Antioxidants/pharmacology
- Cells, Cultured
- Female
- HMGB1 Protein/genetics
- HMGB1 Protein/metabolism
- Hepatocytes/metabolism
- Hepatocytes/pathology
- Humans
- Lipoproteins, LDL/genetics
- Lipoproteins, LDL/metabolism
- Lipoproteins, VLDL/genetics
- Lipoproteins, VLDL/metabolism
- Liver/metabolism
- Liver/pathology
- Liver Diseases, Alcoholic/genetics
- Liver Diseases, Alcoholic/mortality
- Liver Diseases, Alcoholic/pathology
- Male
- Mice
- Mice, Knockout
- Oxidants/pharmacology
- Phosphorylation/genetics
- Primary Cell Culture
- Protein Processing, Post-Translational
Collapse
Affiliation(s)
- Xiaodong Ge
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Daniel J Antoine
- Medical Research Council Centre for Drug Safety Science, Molecular and Clinical Pharmacology, University of Liverpool, Sherrington Buildings, Ashton Street, Liverpool L69 3GE, United Kingdom
| | - Yongke Lu
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Elena Arriazu
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Tung-Ming Leung
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Arielle L Klepper
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Andrea D Branch
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Maria Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York 10029 and
| | - Natalia Nieto
- Division of Liver Diseases, Department of Medicine and Icahn School of Medicine at Mount Sinai, New York, New York 10029.
| |
Collapse
|
38
|
Gao L, Zhou Y, Zhong W, Zhao X, Chen C, Chen X, Gu Y, Chen J, Lv Z, Shen J. Caveolin-1 is essential for protecting against binge drinking-induced liver damage through inhibiting reactive nitrogen species. Hepatology 2014; 60:687-99. [PMID: 24710718 DOI: 10.1002/hep.27162] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 04/02/2014] [Indexed: 12/11/2022]
Abstract
UNLABELLED Caveolin-1 (Cav-1) is known to participate in many diseases, but its roles in alcoholic liver injury remain unknown. In the present study, we aimed to explore the roles of Cav-1 in protecting hepatocytes from ethanol-mediated nitrosative injury. We hypothesized that Cav-1 could attenuate ethanol-mediated nitrosative stress and liver damage through regulating epidermal growth factor receptor/signal transducer and activator of transcription 3/inducible nitric oxide synthase (EGFR/STAT3/iNOS)-signaling cascades. Ethanol-fed mice had time- and dose-dependent increases of Cav-1 in serum and liver with peak increase at 12 hours. Compared to wild-type mice, Cav-1 deficiency mice revealed higher expression of iNOS, higher levels of nitrate/nitrite and peroxynitrite, and had more serious liver damage, accompanied with higher levels of cleaved caspase-3 and apoptotic cell death in liver, and higher levels of alanine aminotransferase and aspartate aminotransferase in serum. Furthermore, the results revealed that the ethanol-mediated Cav-1 increase was in an extracellular signal-regulated kinase-dependent manner, and Cav-1 protected hepatocytes from ethanol-mediated apoptosis by inhibiting iNOS activity and regulating EGFR- and STAT3-signaling cascades. In agreement with these findings, clinical trials in human subjects revealed that serum Cav-1 level was time dependently elevated and peak concentration was observed 12 hours after binge drinking. Alcohol-induced liver lesions were negatively correlated with Cav-1 level, but positively correlated with nitrate/nitrite level, in serum of binge drinkers. CONCLUSIONS Cav-1 could be a cellular defense protein against alcoholic hepatic injury through inhibiting reactive nitrogen species and regulating EGFR/STAT3/iNOS-signaling cascades.
Collapse
Affiliation(s)
- Lei Gao
- School of Chinese Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Shearn CT, Backos DS, Orlicky DJ, Smathers-McCullough RL, Petersen DR. Identification of 5' AMP-activated kinase as a target of reactive aldehydes during chronic ingestion of high concentrations of ethanol. J Biol Chem 2014; 289:15449-62. [PMID: 24722988 DOI: 10.1074/jbc.m113.543942] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The production of reactive aldehydes including 4-hydroxy-2-nonenal (4-HNE) is a key component of the pathogenesis in a spectrum of chronic inflammatory hepatic diseases including alcoholic liver disease (ALD). One consequence of ALD is increased oxidative stress and altered β-oxidation in hepatocytes. A major regulator of β-oxidation is 5' AMP protein kinase (AMPK). In an in vitro cellular model, we identified AMPK as a direct target of 4-HNE adduction resulting in inhibition of both H2O2 and 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR)-induced downstream signaling. By employing biotin hydrazide capture, it was confirmed that 4-HNE treatment of cells resulted in carbonylation of AMPKα/β, which was not observed in untreated cells. Using a murine model of alcoholic liver disease, treatment with high concentrations of ethanol resulted in an increase in phosphorylated as well as carbonylated AMPKα. Despite increased AMPK phosphorylation, there was no significant change in phosphorylation of acetyl CoA carboxylase. Mass spectrometry identified Michael addition adducts of 4-HNE on Cys(130), Cys(174), Cys(227), and Cys(304) on recombinant AMPKα and Cys(225) on recombinant AMPKβ. Molecular modeling analysis of identified 4-HNE adducts on AMPKα suggest that inhibition of AMPK occurs by steric hindrance of the active site pocket and by inhibition of hydrogen peroxide induced oxidation. The observed inhibition of AMPK by 4-HNE provides a novel mechanism for altered β-oxidation in ALD, and these data demonstrate for the first time that AMPK is subject to regulation by reactive aldehydes in vivo.
Collapse
Affiliation(s)
| | | | - David J Orlicky
- Department of Pathology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado 80045 and
| | | | | |
Collapse
|
40
|
Wang Y, Kou Y, Wang X, Cederbaum A, Wang R. Multifactorial comparative proteomic study of cytochrome P450 2E1 function in chronic alcohol administration. PLoS One 2014; 9:e92504. [PMID: 24658151 PMCID: PMC3962406 DOI: 10.1371/journal.pone.0092504] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 02/17/2014] [Indexed: 12/16/2022] Open
Abstract
With the use of iTRAQ technique, a multifactorial comparative proteomic study can be performed. In this study, to obtain an overview of ethanol, CYP2E1 and gender effects on liver injury and gain more insight into the underlying molecular mechanism, mouse liver proteomes were quantitatively analyzed using iTRAQ under eight conditions including mice of different genders, wild type versus CYP2E1 knockout, and normal versus alcohol diet. A series of statistical and bioinformatic analyses were explored to simplify and clarify multifactorial comparative proteomic data. First, with the Principle Component analysis, six proteins, CYP2E1, FAM25, CA3, BHMT, HIBADH and ECHS1, involved in oxidation reduction, energy and lipid metabolism and amino acid metabolism, were identified as the most differentially expressed gene products across all of the experimental conditions of our chronic alcoholism model. Second, hierarchical clustering analysis showed CYP2E1 knockout played a primary role in the overall differential protein expression compared with ethanol and gender factors. Furthermore, pair-wise multiple comparisons have revealed that the only significant expression difference lied in wild-type and CYP2E1 knockout mice both treated with ethanol. Third, K-mean clustering analysis indicated that the CYP2E1 knockout had the reverse effect on ethanol induced oxidative stress and lipid oxidation. More importantly, IPA analysis of proteomic data inferred that the gene expressions of two upstream regulators, NRF2 and PPARα, regulated by chronic alcohol feeding and CYP2E1 knockout, are involved in ethanol induced oxidative stress and lipid oxidation. The present study provides an effectively comprehensive data analysis strategy to compare multiple biological factors, contributing to biochemical effects of alcohol on the liver. The mass spectrometry proteomics data have been deposited to the ProteomeXchange with data set identifier of PXD000635.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Yan Kou
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Xiaodong Wang
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Arthur Cederbaum
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Rong Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
41
|
Acute ethanol causes hepatic mitochondrial depolarization in mice: role of ethanol metabolism. PLoS One 2014; 9:e91308. [PMID: 24618581 PMCID: PMC3950152 DOI: 10.1371/journal.pone.0091308] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 02/12/2014] [Indexed: 12/20/2022] Open
Abstract
Background/Aims An increase of ethanol metabolism and hepatic mitochondrial respiration occurs in vivo after a single binge of alcohol. Here, our aim was to determine how ethanol intake affects hepatic mitochondrial polarization status in vivo in relation to ethanol metabolism and steatosis. Methods Hepatic mitochondrial polarization, permeability transition (MPT), and reduce pyridine nucleotides, and steatosis in mice were monitored by intravital confocal/multiphoton microscopy of the fluorescence of rhodamine 123 (Rh123), calcein, NAD(P)H, and BODIPY493/503, respectively, after gavage with ethanol (1–6 g/kg). Results Mitochondria depolarized in an all-or-nothing fashion in individual hepatocytes as early as 1 h after alcohol. Depolarization was dose- and time-dependent, peaked after 6 to 12 h and maximally affected 94% of hepatocytes. This mitochondrial depolarization was not due to onset of the MPT. After 24 h, mitochondria of most hepatocytes recovered normal polarization and were indistinguishable from untreated after 7 days. Cell death monitored by propidium iodide staining, histology and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) was low throughout. After alcohol, mitochondrial NAD(P)H autofluorescence increased and decreased, respectively, in hepatocytes with polarized and depolarized mitochondria. Ethanol also caused steatosis mainly in hepatocytes with depolarized mitochondria. Depolarization was linked to ethanol metabolism, since deficiency of alcohol dehydrogenase and cytochrome-P450 2E1 (CYP2E1), the major ethanol-metabolizing enzymes, decreased mitochondrial depolarization by ∼70% and ∼20%, respectively. Activation of aldehyde dehydrogenase decreased depolarization, whereas inhibition of aldehyde dehydrogenase enhanced depolarization. Activation of aldehyde dehydrogenase also markedly decreased steatosis. Conclusions Acute ethanol causes reversible hepatic mitochondrial depolarization in vivo that may contribute to steatosis and increased mitochondrial respiration. Onset of this mitochondrial depolarization is linked, at least in part, to metabolism of ethanol to acetaldehyde.
Collapse
|
42
|
Bruinsma BG, Yarmush ML, Uygun K. Organomatics and organometrics: Novel platforms for long-term whole-organ culture. TECHNOLOGY 2014; 2:13. [PMID: 25035864 PMCID: PMC4097862 DOI: 10.1142/s2339547814300029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Organ culture systems are instrumental as experimental whole-organ models of physiology and disease, as well as preservation modalities facilitating organ replacement therapies such as transplantation. Nevertheless, a coordinated system of machine perfusion components and integrated regulatory control has yet to be fully developed to achieve long-term maintenance of organ function ex vivo. Here we outline current strategies for organ culture, or organomatics, and how these systems can be regulated by means of computational algorithms, or organometrics, to achieve the organ culture platforms anticipated in modern-day biomedicine.
Collapse
|
43
|
Shen CH, Tung SY, Huang WS, Lu CC, Lee KC, Hsieh YY, Chang PJ, Liang HF, Chen JH, Lin TH, Hsieh MC, Kuo HC. Exploring the effects of tert-butylhydroperoxide induced liver injury using proteomic approach. Toxicology 2014; 316:61-70. [PMID: 24394546 DOI: 10.1016/j.tox.2013.12.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/04/2013] [Accepted: 12/22/2013] [Indexed: 01/29/2023]
Abstract
Tert-butyl hydroperoxide (t-BHP), an organic lipid hydroperoxide analog, has been demonstrated to exert pro-oxidant effects to evaluate mechanisms involving oxidative stress in hepatocyte cells and rat liver. Herein, we present an investigation of the event of molecular mechanism of t-BHP related acute liver injury. A proteomic approach was used to identify proteins which are differentially expressed in liver cells following t-BHP treatment and the mechanism of its action in apoptotic and endoplasmic reticulum stress pathways. Our results demonstrate that the t-BHP treatment of liver cells increased cell cytoxicity and apoptosis. t-BHP dose-dependent induction of cell apoptosis and stained liver sections relieved the acute rat liver injury were accompanied by sustained phosphorylation of JNK1/2 and p65. In addition, there were 13 differentially displayed proteins between the t-BHP-induced and untreated were assayed and validated in vivo. Furthermore, we demonstrated that t-BHP induced human Chang liver cell viability and apoptosis properties by up-regulating the levels of ETFA (electron transfer flavoprotein subunit alpha). This study demonstrated that there was an increase in the cellular levels of ETFA in the t-BHP induction in viability and apoptosis via the activation of JNK1/2 and NFκB signaling modules. NAC administration and shRNA ETFA conferred resistance to t-BHP-increased ETFA and CHOP expression via IRE1-alpha/TRAF2 complex formation, activation of JNK1/2 and p50. We concluded that the mechanism of t-BHP-induced an apoptosis cascade and endoplasmic reticulum stress in hepatocyte cells by up-regulation of ETFA, providing a new mechanism for liver injury.
Collapse
Affiliation(s)
- Chien-Heng Shen
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Shui-Yi Tung
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan; Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Shih Huang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Chien-Chang Lu
- Department of Colorectal Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ko-Chao Lee
- Department of Colorectal Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan; Department of Surgery, Chang Gung Memorial Hospital - Kaohsiung Medical Center, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yung-Yu Hsieh
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Pey-Jium Chang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Hwey-Fang Liang
- Department of Nursing, Chang Gung University of Science and Technology, Taiwan; Chronic Diseases and Health Promotion Research Center, CGUST, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Jiann-Hwa Chen
- School of Medicine, Fu-Jen Catholic University, Taipei, Taiwan; Institute of Traditional Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Emergency Medicine, Cathay General Hospital, Taipei 10630, Taiwan
| | - Tseng-Hsi Lin
- Division of Hematology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Internal Medicine, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Meng Chiao Hsieh
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taiwan
| | - Hsing-Chun Kuo
- Department of Nursing, Chang Gung University of Science and Technology, Taiwan; Chronic Diseases and Health Promotion Research Center, CGUST, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
44
|
Zhong W, Li Q, Xie G, Sun X, Tan X, Sun X, Jia W, Zhou Z. Dietary fat sources differentially modulate intestinal barrier and hepatic inflammation in alcohol-induced liver injury in rats. Am J Physiol Gastrointest Liver Physiol 2013; 305:G919-G932. [PMID: 24113767 PMCID: PMC3882440 DOI: 10.1152/ajpgi.00226.2013] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/03/2013] [Indexed: 01/31/2023]
Abstract
Endotoxemia is a causal factor in the development of alcoholic liver injury. The present study aimed at determining the interactions of ethanol with different fat sources at the gut-liver axis. Male Sprague-Dawley rats were pair fed control or ethanol liquid diet for 8 wk. The liquid diets were based on a modified Lieber-DeCarli formula, with 30% total calories derived from corn oil (rich in polyunsaturated fatty acids). To test the effects of saturated fats, corn oil in the ethanol diet was replaced by either cocoa butter (CB, rich in long-chain saturated fatty acids) or medium-chain triglycerides (MCT, exclusively medium-chain saturated fatty acids). Ethanol feeding increased hepatic lipid accumulation and inflammatory cell infiltration and perturbed hepatic and serum metabolite profiles. Ethanol feeding with CB or MCT alleviated ethanol-induced liver injury and attenuated ethanol-induced metabolic perturbation. Both CB and MCT also normalized ethanol-induced hepatic macrophage activation, cytokine expression, and neutrophil infiltration. Ethanol feeding elevated serum endotoxin level, which was normalized by MCT but not CB. In accordance, ethanol-induced downregulations of intestinal occludin and zonula occludens-1 were normalized by MCT but not CB. However, CB normalized ethanol-increased hepatic endotoxin level in association with upregulation of an endotoxin detoxifying enzyme, argininosuccinate synthase 1 (ASS1). Knockdown ASS1 in H4IIEC3 cells resulted in impaired endotoxin clearance and upregulated cytokine expression. These data demonstrate that the protection of saturated fats against alcohol-induced liver injury occur via different actions at the gut-liver axis and are chain length dependent.
Collapse
Affiliation(s)
- Wei Zhong
- Center for Translational Biomedical Research and Dept. of Nutrition, Univ. of North Carolina at Greensboro, North Carolina Research Campus, 500 Laureate Way, Suite 4226, Kannapolis, NC 28081.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Fowler S, Akins M, Zhou H, Figeys D, Bennett SA. The liver connexin32 interactome is a novel plasma membrane-mitochondrial signaling nexus. J Proteome Res 2013; 12:2597-610. [PMID: 23590695 PMCID: PMC3714164 DOI: 10.1021/pr301166p] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Indexed: 02/07/2023]
Abstract
Connexins are the structural subunits of gap junctions and act as protein platforms for signaling complexes. Little is known about tissue-specific connexin signaling nexuses, given significant challenges associated with affinity-purifying endogenous channel complexes to the level required for interaction analyses. Here, we used multiple subcellular fractionation techniques to isolate connexin32-enriched membrane microdomains from murine liver. We show, for the first time, that connexin32 localizes to both the plasma membrane and inner mitochondrial membrane of hepatocytes. Using a combination of immunoprecipitation-high throughput mass spectrometry, reciprocal co-IP, and subcellular fractionation methodologies, we report a novel interactome validated using null mutant controls. Eighteen connexin32 interacting proteins were identified. The majority represent resident mitochondrial proteins, a minority represent plasma membrane, endoplasmic reticulum, or cytoplasmic partners. In particular, connexin32 interacts with connexin26 and the mitochondrial protein, sideroflexin-1, at the plasma membrane. Connexin32 interaction enhances connexin26 stability. Converging bioinformatic, biochemical, and confocal analyses support a role for connexin32 in transiently tethering mitochondria to connexin32-enriched plasma membrane microdomains through interaction with proteins in the outer mitochondrial membrane, including sideroflexin-1. Complex formation increases the pool of sideroflexin-1 that is present at the plasma membrane. Together, these data identify a novel plasma membrane/mitochondrial signaling nexus in the connexin32 interactome.
Collapse
Affiliation(s)
- Stephanie
L. Fowler
- Neural
Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of
Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Mark Akins
- Neural
Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of
Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Hu Zhou
- Neural
Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of
Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai,
China
| | - Daniel Figeys
- Neural
Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of
Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Steffany A.L. Bennett
- Neural
Regeneration Laboratory, Ottawa Institute of Systems Biology, Department of
Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
46
|
Ge X, Lu Y, Leung TM, Sørensen ES, Nieto N. Milk osteopontin, a nutritional approach to prevent alcohol-induced liver injury. Am J Physiol Gastrointest Liver Physiol 2013; 304:G929-39. [PMID: 23518682 PMCID: PMC3652071 DOI: 10.1152/ajpgi.00014.2013] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alcohol consumption is a leading cause of liver disease worldwide; thus, there is an urgent need to develop novel therapeutic interventions. Key events for the onset and progression of alcoholic liver disease result in part from the gut-to-liver interaction. Osteopontin is a cytokine present at high concentration in human milk, umbilical cord, and infants' plasma with beneficial potential. We hypothesized that dietary administration of milk osteopontin could prevent alcohol-induced liver injury perhaps by maintaining gut integrity and averting hepatic inflammation and steatosis. Wild-type mice were fed either the control or the ethanol Lieber-DeCarli diets alone or in combination with milk osteopontin for 3 wk, and parameters of gut and liver damage were measured. Milk osteopontin protected the stomach and the gut by increasing gland height, crypt cell plus enterocyte proliferation, and mucin content in addition to lowering macrophages, plasmacytes, lymphocytes, and neutrophils in the mucosa and submucosa in alcohol-fed mice. Milk osteopontin targeted the gut-liver axis, preserving the expression of tight-junction proteins in alcohol-fed mice thus maintaining intestinal integrity and permeability. There was protection from liver injury since transaminases, the activity scores, triglyceride levels, neutrophil infiltration, 3-nitrotyrosine residues, lipid peroxidation end products, translocation of gram-negative bacteria, lipopolysaccharide levels, and tumor necrosis factor-α were lower in cotreated than in ethanol-fed mice. Furthermore, milk osteopontin diminished ethanol-mediated liver injury in OPN knockout mice. Milk osteopontin could be a simple effective nutritional therapeutic strategy to prevent alcohol hepatotoxicity due, among others, to gut protective, anti-inflammatory, and anti-steatotic actions.
Collapse
Affiliation(s)
- Xiaodong Ge
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Yongke Lu
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Tung-Ming Leung
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| | - Esben S. Sørensen
- 2Department of Molecular Biology and Genetics, Aarhus University, Aarhus Science Park, Denmark
| | - Natalia Nieto
- 1Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, New York, New York; and
| |
Collapse
|
47
|
Abstract
Chronic alcohol consumption is a leading cause of chronic liver disease worldwide, leading to cirrhosis and hepatocellular carcinoma. Currently, the most widely used model for alcoholic liver injury is ad libitum feeding with the Lieber-DeCarli liquid diet containing ethanol for 4-6 weeks; however, this model, without the addition of a secondary insult, only induces mild steatosis, slight elevation of serum alanine transaminase (ALT) and little or no inflammation. Here we describe a simple mouse model of alcoholic liver injury by chronic ethanol feeding (10-d ad libitum oral feeding with the Lieber-DeCarli ethanol liquid diet) plus a single binge ethanol feeding. This protocol for chronic-plus-single-binge ethanol feeding synergistically induces liver injury, inflammation and fatty liver, which mimics acute-on-chronic alcoholic liver injury in patients. This feeding protocol can also be extended to chronic feeding for longer periods of time up to 8 weeks plus single or multiple binges. Chronic-binge ethanol feeding leads to high blood alcohol levels; thus, this simple model will be very useful for the study of alcoholic liver disease (ALD) and of other organs damaged by alcohol consumption.
Collapse
|