1
|
Li M, Houben T, Bitorina AV, Meesters DM, Israelsen M, Kjærgaard M, Koek GH, Hendrikx T, Verbeek J, Krag A, Thiele M, Shiri-Sverdlov R. Plasma cathepsin D as an early indicator of alcohol-related liver disease. JHEP Rep 2024; 6:101117. [PMID: 39263329 PMCID: PMC11388167 DOI: 10.1016/j.jhepr.2024.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 09/13/2024] Open
Abstract
Background & Aims People who drink alcohol excessively are at increased risk of developing metabolic dysfunction and alcohol-related liver disease (MetALD) or the more severe form alcohol-related liver disease (ALD). One of the most significant challenges concerns the early detection of MetALD/ALD. Previously, we have demonstrated that the lysosomal enzyme cathepsin D (CTSD) is an early marker for metabolic dysfunction-associated steatohepatitis (MASH). Here, we hypothesized that plasma CTSD can also serve as an early indicator of MetALD/ALD. Methods We included 303 persistent heavy drinkers classified as having MetALD or ALD (n = 152) and abstinent patients with a history of excessive drinking (n = 151). Plasma CTSD levels of patients with MetALD/ALD without decompensation were compared with 40 healthy controls. Subsequently, the relationship between plasma CTSD levels and hepatic histological scores was established. Receiver-operating characteristic curves were generated to assess the precision of plasma CTSD levels in detecting MetALD/ALD. Lastly, plasma CTSD levels were compared between abstainers and drinkers. Results Plasma CTSD levels were higher in patients with MetALD/ALD compared to healthy controls. While hepatic disease parameters (AST/ALT ratio, liver stiffness measurement) were higher at advanced histopathological stages (assessed by liver biopsy), plasma CTSD levels were already elevated at early histopathological stages. Furthermore, combining plasma CTSD levels with liver stiffness measurement and AST/ALT ratio yielded enhanced diagnostic precision (AUC 0.872) in detecting MetALD/ALD in contrast to the utilization of CTSD alone (AUC 0.804). Plasma CTSD levels remained elevated in abstainers. Conclusion Elevated levels of CTSD in the circulation can serve as an early indicator of MetALD/ALD. Impact and implications Alcohol-related liver disease is the leading cause of liver disease-related morbidity and mortality worldwide. However, the currently available non-invasive methods to diagnose MetALD/ALD are only able to detect advanced stages of MetALD/ALD. Here, we demonstrate that plasma levels of the lysosomal enzyme cathepsin D are already elevated at early stages of MetALD/ALD. Moreover, cathepsin D levels outperformed the currently available non-invasive methods to detect MetALD/ALD. Plasma levels of cathepsin D could therefore be a useful non-invasive marker for detection of MetALD/ALD.
Collapse
Affiliation(s)
- Mengying Li
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Tom Houben
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Albert V. Bitorina
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Dennis M. Meesters
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| | - Mads Israelsen
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Maria Kjærgaard
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Ger H. Koek
- Department of Internal Medicine Division of Gastroenterology and Hepatology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Tim Hendrikx
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Jef Verbeek
- Laboratory of Hepatology, Department of Chronic Diseases and Metabolism, KU Leuven, Belgium; Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Aleksander Krag
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Maja Thiele
- Center for Liver Research, Odense University Hospital and University of Southern Denmark, Kloevervaenget 10, entrance 112, DK-5000 Odense, Denmark
| | - Ronit Shiri-Sverdlov
- Department of Genetics and Cell Biology, Institute of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands
| |
Collapse
|
2
|
Salete-Granado D, Carbonell C, Puertas-Miranda D, Vega-Rodríguez VJ, García-Macia M, Herrero AB, Marcos M. Autophagy, Oxidative Stress, and Alcoholic Liver Disease: A Systematic Review and Potential Clinical Applications. Antioxidants (Basel) 2023; 12:1425. [PMID: 37507963 PMCID: PMC10376811 DOI: 10.3390/antiox12071425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Ethanol consumption triggers oxidative stress by generating reactive oxygen species (ROS) through its metabolites. This process leads to steatosis and liver inflammation, which are critical for the development of alcoholic liver disease (ALD). Autophagy is a regulated dynamic process that sequesters damaged and excess cytoplasmic organelles for lysosomal degradation and may counteract the harmful effects of ROS-induced oxidative stress. These effects include hepatotoxicity, mitochondrial damage, steatosis, endoplasmic reticulum stress, inflammation, and iron overload. In liver diseases, particularly ALD, macroautophagy has been implicated as a protective mechanism in hepatocytes, although it does not appear to play the same role in stellate cells. Beyond the liver, autophagy may also mitigate the harmful effects of alcohol on other organs, thereby providing an additional layer of protection against ALD. This protective potential is further supported by studies showing that drugs that interact with autophagy, such as rapamycin, can prevent ALD development in animal models. This systematic review presents a comprehensive analysis of the literature, focusing on the role of autophagy in oxidative stress regulation, its involvement in organ-organ crosstalk relevant to ALD, and the potential of autophagy-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Daniel Salete-Granado
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
| | - Cristina Carbonell
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - David Puertas-Miranda
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Víctor-José Vega-Rodríguez
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
| | - Marina García-Macia
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Instituto de Biología Funcional y Genómica (IBFG), Universidad de Salamanca, 37007 Salamanca, Spain
| | - Ana Belén Herrero
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain; (D.S.-G.); (C.C.); (D.P.-M.); (V.-J.V.-R.); (M.G.-M.); (A.B.H.)
- Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Unidad de Medicina Molecular, Departamento de Medicina, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
3
|
Thomes PG, Strupp MS, Donohue TM, Kubik JL, Sweeney S, Mahmud R, Schott MB, Schulze RJ, McNiven MA, Casey CA. Hydroxysteroid 17β-dehydrogenase 11 accumulation on lipid droplets promotes ethanol-induced cellular steatosis. J Biol Chem 2023; 299:103071. [PMID: 36849008 PMCID: PMC10060109 DOI: 10.1016/j.jbc.2023.103071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 02/02/2023] [Accepted: 02/20/2023] [Indexed: 02/27/2023] Open
Abstract
Lipid droplets (LDs) are fat-storing organelles enclosed by a phospholipid monolayer, which harbors membrane-associated proteins that regulate distinct LD functions. LD proteins are degraded by the ubiquitin-proteasome system (UPS) and/or by lysosomes. Because chronic ethanol (EtOH) consumption diminishes the hepatic functions of the UPS and lysosomes, we hypothesized that continuous EtOH consumption slows the breakdown of lipogenic LD proteins targeted for degradation, thereby causing LD accumulation. Here, we report that LDs from livers of EtOH-fed rats exhibited higher levels of polyubiquitylated-proteins, linked at either lysine 48 (directed to proteasome) or lysine 63 (directed to lysosomes) than LDs from pair-fed control rats. MS proteomics of LD proteins, immunoprecipitated with UB remnant motif antibody (K-ε-GG), identified 75 potential UB proteins, of which 20 were altered by chronic EtOH administration. Among these, hydroxysteroid 17β-dehydrogenase 11 (HSD17β11) was prominent. Immunoblot analyses of LD fractions revealed that EtOH administration enriched HSD17β11 localization to LDs. When we overexpressed HSD17β11 in EtOH-metabolizing VA-13 cells, the steroid dehydrogenase 11 became principally localized to LDs, resulting in elevated cellular triglycerides (TGs). Ethanol exposure augmented cellular TG, while HSD17β11 siRNA decreased both control and EtOH-induced TG accumulation. Remarkably, HSD17β11 overexpression lowered the LD localization of adipose triglyceride lipase. EtOH exposure further reduced this localization. Reactivation of proteasome activity in VA-13 cells blocked the EtOH-induced rises in both HSD17β11 and TGs. Our findings indicate that EtOH exposure blocks HSD17β11 degradation by inhibiting the UPS, thereby stabilizing HSD17β11 on LD membranes, to prevent lipolysis by adipose triglyceride lipase and promote cellular LD accumulation.
Collapse
Affiliation(s)
- Paul G Thomes
- Department of Veterans' Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA.
| | - Michael S Strupp
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Terence M Donohue
- Department of Veterans' Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Jacy L Kubik
- Department of Veterans' Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sarah Sweeney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - R Mahmud
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Micah B Schott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ryan J Schulze
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Mark A McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Carol A Casey
- Department of Veterans' Affairs, VA-Nebraska-Western Iowa Health Care System, Omaha, Nebraska, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
4
|
Thomes PG, Rensch G, Casey CA, Donohue TM. Ethanol Exposure to Ethanol-Oxidizing HEPG2 Cells Induces Intracellular Protein Aggregation. Cells 2023; 12:cells12071013. [PMID: 37048086 PMCID: PMC10093015 DOI: 10.3390/cells12071013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/05/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Background: Aggresomes are collections of intracellular protein aggregates. In liver cells of patients with alcoholic hepatitis, aggresomes appear histologically as cellular inclusions known as Mallory–Denk (M–D) bodies. The proteasome is a multicatalytic intracellular protease that catalyzes the degradation of both normal (native) and abnormal (misfolded and/or damaged) proteins. The enzyme minimizes intracellular protein aggregate formation by rapidly degrading abnormal proteins before they form aggregates. When proteasome activity is blocked, either by specific inhibitors or by intracellular oxidants (e.g., peroxynitrite, acetaldehyde), aggresome formation is enhanced. Here, we sought to verify whether inhibition of proteasome activity by ethanol exposure enhances protein aggregate formation in VL-17A cells, which are recombinant, ethanol-oxidizing HepG2 cells that express both alcohol dehydrogenase (ADH) and cytochrome P450 2E1 (CYP2E1). Methods: We exposed ethanol-non-oxidizing HepG2 cells (ADH−/CYP2E1−) or ethanol-oxidizing VL-17A (ADH+/CYP2E1+) to varying levels of ethanol for 24 h or 72 h. After these treatments, we stained cells for aggresomes (detected microscopically) and quantified their numbers and sizes. We also conducted flow cytometric analyses to confirm our microscopic findings. Additionally, aggresome content in liver cells of patients with alcohol-induced hepatitis was quantified. Results: After we exposed VL-17A cells to increasing doses of ethanol for 24 h or 72 h, 20S proteasome activity declined in response to rising ethanol concentrations. After 24 h of ethanol exposure, aggresome numbers in VL-17A cells were 1.8-fold higher than their untreated controls at all ethanol concentrations employed. After 72 h of ethanol exposure, mean aggresome numbers were 2.5-fold higher than unexposed control cells. The mean aggregate size in all ethanol-exposed VL-17A cells was significantly higher than in unexposed control cells but was unaffected by the duration of ethanol exposure. Co-exposure of cells to EtOH and rapamycin, the latter an autophagy activator, completely prevented EtOH-induced aggresome formation. In the livers of patients with alcohol-induced hepatitis (AH), the staining intensity of aggresomes was 2.2-fold higher than in the livers of patients without alcohol use disorder (AUD). Conclusions: We conclude that ethanol-induced proteasome inhibition in ethanol-metabolizing VL-17A hepatoma cells causes accumulation of protein aggregates. Notably, autophagy activation removes such aggregates. The significance of these findings is discussed.
Collapse
Affiliation(s)
- Paul G. Thomes
- Liver Study Unit, VA-Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- The Depts of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Biochemistry/Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Correspondence: ; Tel.: +1-402-995-3738; Fax: +1-402-449-0604
| | - Gage Rensch
- The Depts of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Carol A. Casey
- Liver Study Unit, VA-Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- The Depts of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Biochemistry/Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Terrence M. Donohue
- Liver Study Unit, VA-Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
- The Depts of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Biochemistry/Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
5
|
New-Aaron M, Thomes PG, Ganesan M, Dagur RS, Donohue TM, Kusum KK, Poluektova LY, Osna NA. Alcohol-Induced Lysosomal Damage and Suppression of Lysosome Biogenesis Contribute to Hepatotoxicity in HIV-Exposed Liver Cells. Biomolecules 2021; 11:1497. [PMID: 34680130 PMCID: PMC8533635 DOI: 10.3390/biom11101497] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 02/05/2023] Open
Abstract
Although the causes of hepatotoxicity among alcohol-abusing HIV patients are multifactorial, alcohol remains the least explored "second hit" for HIV-related hepatotoxicity. Here, we investigated whether metabolically derived acetaldehyde impairs lysosomes to enhance HIV-induced hepatotoxicity. We exposed Cytochrome P450 2E1 (CYP2E1)-expressing Huh 7.5 (also known as RLW) cells to an acetaldehyde-generating system (AGS) for 24 h. We then infected (or not) the cells with HIV-1ADA then exposed them again to AGS for another 48 h. Lysosome damage was assessed by galectin 3/LAMP1 co-localization and cathepsin leakage. Expression of lysosome biogenesis-transcription factor, TFEB, was measured by its protein levels and by in situ immunofluorescence. Exposure of cells to both AGS + HIV caused the greatest amount of lysosome leakage and its impaired lysosomal biogenesis, leading to intrinsic apoptosis. Furthermore, the movement of TFEB from cytosol to the nucleus via microtubules was impaired by AGS exposure. The latter impairment appeared to occur by acetylation of α-tubulin. Moreover, ZKSCAN3, a repressor of lysosome gene activation by TFEB, was amplified by AGS. Both these changes contributed to AGS-elicited disruption of lysosome biogenesis. Our findings indicate that metabolically generated acetaldehyde damages lysosomes and likely prevents their repair and restoration, thereby exacerbating HIV-induced hepatotoxicity.
Collapse
Affiliation(s)
- Moses New-Aaron
- Department of Environmental Health, Occupational Health, and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
| | - Paul G. Thomes
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Terrence M. Donohue
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Kharbanda K. Kusum
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Natalia A. Osna
- Department of Environmental Health, Occupational Health, and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (P.G.T.); (M.G.); (R.S.D.); (T.M.D.J.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| |
Collapse
|
6
|
Sweeter JM, Kudrna K, Hunt K, Thomes P, Dickey BF, Brody SL, Dickinson JD. Autophagy of mucin granules contributes to resolution of airway mucous metaplasia. Sci Rep 2021; 11:13037. [PMID: 34158522 PMCID: PMC8219712 DOI: 10.1038/s41598-021-91932-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/01/2021] [Indexed: 12/21/2022] Open
Abstract
Exacerbations of muco-obstructive airway diseases such as COPD and asthma are associated with epithelial changes termed mucous metaplasia (MM). Many molecular pathways triggering MM have been identified; however, the factors that regulate resolution are less well understood. We hypothesized that the autophagy pathway is required for resolution of MM by eliminating excess non-secreted intracellular mucin granules. We found increased intracellular levels of mucins Muc5ac and Muc5b in mice deficient in autophagy regulatory protein, Atg16L1, and that this difference was not due to defects in the known baseline or stimulated mucin secretion pathways. Instead, we found that, in mucous secretory cells, Lc3/Lamp1 vesicles colocalized with mucin granules particularly adjacent to the nucleus, suggesting that some granules were being eliminated in the autophagy pathway rather than secreted. Using a mouse model of MM resolution, we found increased lysosomal proteolytic activity that peaked in the days after mucin production began to decline. In purified lysosomal fractions, Atg16L1-deficient mice had reduced proteolytic degradation of Lc3 and Sqstm1 and persistent accumulation of mucin granules associated with impaired resolution of mucous metaplasia. In normal and COPD derived human airway epithelial cells (AECs), activation of autophagy by mTOR inhibition led to a reduction of intracellular mucin granules in AECs. Our findings indicate that during peak and resolution phases of MM, autophagy activity rather than secretion is required for elimination of some remaining mucin granules. Manipulation of autophagy activation offers a therapeutic target to speed resolution of MM in airway disease exacerbations.
Collapse
Affiliation(s)
- J M Sweeter
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Kudrna
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - K Hunt
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Thomes
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - B F Dickey
- Department of Pulmonary Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - S L Brody
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - J D Dickinson
- Pulmonary, Critical Care and Sleep Medicine Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
7
|
Percival BC, Latour YL, Tifft CJ, Grootveld M. Rapid Identification of New Biomarkers for the Classification of GM1 Type 2 Gangliosidosis Using an Unbiased 1H NMR-Linked Metabolomics Strategy. Cells 2021; 10:572. [PMID: 33807817 PMCID: PMC7998791 DOI: 10.3390/cells10030572] [Citation(s) in RCA: 4] [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: 12/08/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/04/2023] Open
Abstract
Biomarkers currently available for the diagnosis, prognosis, and therapeutic monitoring of GM1 gangliosidosis type 2 (GM1T2) disease are mainly limited to those discovered in targeted proteomic-based studies. In order to identify and establish new, predominantly low-molecular-mass biomarkers for this disorder, we employed an untargeted, multi-analyte approach involving high-resolution 1H NMR analysis coupled to a range of multivariate analysis and computational intelligence technique (CIT) strategies to explore biomolecular distinctions between blood plasma samples collected from GM1T2 and healthy control (HC) participants (n = 10 and 28, respectively). The relationship of these differences to metabolic mechanisms underlying the pathogenesis of GM1T2 disorder was also investigated. 1H NMR-linked metabolomics analyses revealed significant GM1T2-mediated dysregulations in ≥13 blood plasma metabolites (corrected p < 0.04), and these included significant upregulations in 7 amino acids, and downregulations in lipoprotein-associated triacylglycerols and alanine. Indeed, results acquired demonstrated a profound distinctiveness between the GM1T2 and HC profiles. Additionally, employment of a genome-scale network model of human metabolism provided evidence that perturbations to propanoate, ethanol, amino-sugar, aspartate, seleno-amino acid, glutathione and alanine metabolism, fatty acid biosynthesis, and most especially branched-chain amino acid degradation (p = 10-12-10-5) were the most important topologically-highlighted dysregulated pathways contributing towards GM1T2 disease pathology. Quantitative metabolite set enrichment analysis revealed that pathological locations associated with these dysfunctions were in the order fibroblasts > Golgi apparatus > mitochondria > spleen ≈ skeletal muscle ≈ muscle in general. In conclusion, results acquired demonstrated marked metabolic imbalances and alterations to energy demand, which are consistent with GM1T2 disease pathogenesis mechanisms.
Collapse
Affiliation(s)
- Benita C. Percival
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| | - Yvonne L. Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232-0252, USA;
| | - Cynthia J. Tifft
- Deputy Clinical Director, National Human Genome Research Institute, Director, National Institutes of Health, Bethesda, MD 20892-1205, USA;
| | - Martin Grootveld
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| |
Collapse
|
8
|
Kouroumalis E, Voumvouraki A, Augoustaki A, Samonakis DN. Autophagy in liver diseases. World J Hepatol 2021; 13:6-65. [PMID: 33584986 PMCID: PMC7856864 DOI: 10.4254/wjh.v13.i1.6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is the liver cell energy recycling system regulating a variety of homeostatic mechanisms. Damaged organelles, lipids and proteins are degraded in the lysosomes and their elements are re-used by the cell. Investigations on autophagy have led to the award of two Nobel Prizes and a health of important reports. In this review we describe the fundamental functions of autophagy in the liver including new data on the regulation of autophagy. Moreover we emphasize the fact that autophagy acts like a two edge sword in many occasions with the most prominent paradigm being its involvement in the initiation and progress of hepatocellular carcinoma. We also focused to the implication of autophagy and its specialized forms of lipophagy and mitophagy in the pathogenesis of various liver diseases. We analyzed autophagy not only in well studied diseases, like alcoholic and nonalcoholic fatty liver and liver fibrosis but also in viral hepatitis, biliary diseases, autoimmune hepatitis and rare diseases including inherited metabolic diseases and also acetaminophene hepatotoxicity. We also stressed the different consequences that activation or impairment of autophagy may have in hepatocytes as opposed to Kupffer cells, sinusoidal endothelial cells or hepatic stellate cells. Finally, we analyzed the limited clinical data compared to the extensive experimental evidence and the possible future therapeutic interventions based on autophagy manipulation.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71110, Greece
| | - Argryro Voumvouraki
- 1 Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54636, Greece
| | - Aikaterini Augoustaki
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece
| | - Dimitrios N Samonakis
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece.
| |
Collapse
|
9
|
Dagur RS, New-Aaron M, Ganesan M, Wang W, Romanova S, Kidambi S, Kharbanda KK, Poluektova LY, Osna NA. Alcohol-and-HIV-Induced Lysosomal Dysfunction Regulates Extracellular Vesicles Secretion in Vitro and in Liver-Humanized Mice. BIOLOGY 2021; 10:29. [PMID: 33466299 PMCID: PMC7824801 DOI: 10.3390/biology10010029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Alcohol abuse is common in people living with HIV-1 and dramaticallyenhances the severity of HIV-induced liver damage by inducing oxidative stress and lysosomaldysfunction in the liver cells. We hypothesize that the increased release of extracellular vesicles(EVs) in hepatocytes and liver humanized mouse model is linked to lysosome dysfunction. METHODS The study was performed on primary human hepatocytes and human hepatoma RLWXP-GFP (Huh7.5 cells stably transfected with CYP2E1 and XPack-GFP) cells and validated on ethanol-fed liverhumanizedfumarylacetoacetate hydrolase (Fah)-/-, Rag2-/-, common cytokine receptor gamma chainknockout (FRG-KO) mice. Cells and mice were infected with HIV-1ADA virus. RESULTS We observedan increase in the secretion of EVs associated with a decrease in lysosomal activity and expressionof lysosomal-associated membrane protein 1. Next-generation RNA sequencing of primary humanhepatocytes revealed 63 differentially expressed genes, with 13 downregulated and 50 upregulatedgenes in the alcohol-HIV-treated group. Upstream regulator analysis of differentially expressedgenes through Ingenuity Pathway Analysis identified transcriptional regulators affecting downstreamgenes associated with increased oxidative stress, lysosomal associated disease, and function andEVs biogenesis. Our in vitro findings were corroborated by in vivo studies on human hepatocytetransplantedhumanized mice, indicating that intensive EVs' generation by human hepatocytes andtheir secretion to serum was associated with increased oxidative stress and reduction in lysosomalactivities triggered by HIV infection and ethanol diet. CONCLUSION HIV-and-ethanol-metabolisminducedEVs release is tightly controlled by lysosome status in hepatocytes and participates in thedevelopment of double-insult-induced liver injury.
Collapse
Affiliation(s)
- Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.N.-A.); (M.G.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Moses New-Aaron
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.N.-A.); (M.G.); (K.K.K.)
- Department of Environmental, Agriculture and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68102, USA
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.N.-A.); (M.G.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Weimin Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; (W.W.); (L.Y.P.)
| | - Svetlana Romanova
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588, USA;
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.N.-A.); (M.G.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA; (W.W.); (L.Y.P.)
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.N.-A.); (M.G.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
10
|
New-Aaron M, Ganesan M, Dagur RS, Kharbanda KK, Poluektova LY, Osna NA. Obeticholic acid attenuates human immunodeficiency virus/alcohol metabolism-induced pro-fibrotic activation in liver cells. World J Hepatol 2020; 12:965-975. [PMID: 33312422 PMCID: PMC7701963 DOI: 10.4254/wjh.v12.i11.965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/16/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The morbidity and mortality of human immunodeficiency virus (HIV)-infection is often associated with liver disease, which progresses slowly into severe liver dysfunction. There are multiple insults which exacerbate HIV-related liver injury, including HIV-associated dysregulation of lipid metabolism and fat turnover, co-infections with hepatotropic viruses and alcohol abuse. As we reported before, exposure of hepatocytes to HIV and alcohol metabolites causes high oxidative stress, impairs proteasomal and lysosomal functions leading to accumulation of HIV in these cells, which end-ups with apoptotic cell death and finally promotes development of liver fibrosis. AIM To study whether obeticholic acid (OCA) prevents HIV/ethanol metabolism-induced hepatotoxicity and subsequent activation of hepatic stellate cells (HSC) by HIV+ apoptotic hepatocyte engulfment. METHODS Huh7.5-CYP (RLW) cells were exposed to HIV and acetaldehyde-generating system (AGS) in the presence or absence of OCA. In the cells, we measured the expression of HIV-related markers: HIVgagRNA-by real-time polymerase chain reaction (PCR), p24- by western blot, HIV DNA-by semi-nested PCR, integrated HIV DNA-by ddPCR. Lysosomal and proteasomal activities were measured using fluorometrically-labeled substrates. For hepatocyte apoptosis, cleaved caspase 3 and cleaved PARP were visualized by western blot and cytokeratin 18- by M30 ELISA-in supernatants. Apoptotic bodies were generated from untreated and HIV-treated RLW cells exposed to UV light. Pro-fibrotic activation of HSC was characterized by Col1A1 and transforming growth factor-β mRNAs, while inflammasome activation- by NLRP3, caspase 1, interleukin (IL)-6, IL-1β mRNA levels. RESULTS In RLW cells, OCA treatment attenuated HIV-AGS-induced accumulation of HIVgagRNA, HIV DNA and p24. OCA suppressed reactive oxygen species production and restored chymotrypsin-like proteasome activity as well as cathepsin B lysosome activity. OCA also decreased HIV-AGS-triggered apoptosis in RLW cells. Exposure of HIV-containing apoptotic hepatocytes to HSC prevented activation of inflammasome and induced pro-fibrotic activation in these cells. CONCLUSION We conclude that by suppressing oxidative stress and restoring proteasomal and lysosomal functions impaired by HIV and ethanol metabolism, OCA decreases accumulation of HIV in hepatocytes, leading to down-regulation of apoptosis in these cells. In addition, OCA reverses pro-fibrotic and inflammasome-related activation of HSC triggered by engulfment of HIV-containing apoptotic hepatocytes, potentially contributing to suppression of liver fibrosis development.
Collapse
Affiliation(s)
- Moses New-Aaron
- Department of Environmental, Agriculture and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68105, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
| | - Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, United States
| | - Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, United States
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, United States
| | - Larisa Y Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Natalia A Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, United States.
| |
Collapse
|
11
|
Yuan F, Xu Y, You K, Zhang J, Yang F, Li YX. Calcitriol alleviates ethanol-induced hepatotoxicity via AMPK/mTOR-mediated autophagy. Arch Biochem Biophys 2020; 697:108694. [PMID: 33232716 DOI: 10.1016/j.abb.2020.108694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 12/28/2022]
Abstract
Excessive ethanol consumption causes cellular damage, leading to fetal alcohol syndrome and alcohol liver diseases, which are frequently seen with vitamin D (VD) deficiency. A great deal of progress has been achieved in the mechanisms of ethanol-induced hepatocyte damage. However, there are limited intervention means to reduce or rescue hepatocytes damage caused by ethanol. On the basis of our preliminary limited screen process, calcitriol showed a positive effect on protecting hepatocyte viability. Therefore, the molecular basis is worth elucidating. We found that calcitriol pretreatment markedly improved the cell viability, decreased cell apoptosis and oxidative stress and alleviated the abnormal mitochondrial morphology and membrane potential of hepatocytes induced by ethanol. Notably, autophagy was significantly enhanced by calcitriol, as evident by the increasing number of autophagosomes and autolysosomes, upregulated LC3B-Ⅱ and ATG5 levels, and promotion of p62 degradation. Furthermore, calcitriol pretreatment increased the colocalization of GFP-LC3-labeled autophagosomes with mitochondria, suggesting that calcitriol effectively promoted ethanol-induced mitophagy in hepatocytes. In addition, the inhibition of autophagy attenuated the protective and preventive effect of calcitriol. Furthermore, the effect of calcitriol on autophagy was regulated by AMPK/mTOR signaling, and signaling transduction was dependent on the Vitamin D receptor (VDR). In conclusion, calcitriol ameliorates ethanol-induced hepatocyte damage by enhancing autophagy. It may offer a convenient preventive and hepatoprotective mean for people on occasional social drink.
Collapse
Affiliation(s)
- Fang Yuan
- School of Life Sciences, University of Science and Technology of China, 230027, Hefei, China; Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yingying Xu
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Kai You
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Jiaye Zhang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Fan Yang
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China
| | - Yin-Xiong Li
- Institute of Public Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China; Guangdong Provincial Key Laboratory of Biocomputing, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 510530, Guangzhou, China.
| |
Collapse
|
12
|
Xia SW, Wang ZM, Sun SM, Su Y, Li ZH, Shao JJ, Tan SZ, Chen AP, Wang SJ, Zhang ZL, Zhang F, Zheng SZ. Endoplasmic reticulum stress and protein degradation in chronic liver disease. Pharmacol Res 2020; 161:105218. [PMID: 33007418 DOI: 10.1016/j.phrs.2020.105218] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Endoplasmic reticulum (ER) stress is easily observed in chronic liver disease, which often causes accumulation of unfolded or misfolded proteins in the ER, leading to unfolded protein response (UPR). Regulating protein degradation is an integral part of UPR to relieve ER stress. The major protein degradation system includes the ubiquitin-proteasome system (UPS) and autophagy. All three arms of UPR triggered in response to ER stress can regulate UPS and autophagy. Accumulated misfolded proteins could activate these arms, and then generate various transcription factors to regulate the expression of UPS-related and autophagy-related genes. The protein degradation process regulated by UPR has great significance in many chronic liver diseases, including non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), viral hepatitis, liver fibrosis, and hepatocellular carcinoma(HCC). In most instances, the degradation of excessive proteins protects cells with ER stress survival from apoptosis. According to the specific functions of protein degradation in chronic liver disease, choosing to promote or inhibit this process is promising as a potential method for treating chronic liver disease.
Collapse
Affiliation(s)
- Si-Wei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhi-Min Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Su-Min Sun
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Su
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhang-Hao Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiang-Juan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shan-Zhong Tan
- Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - An-Ping Chen
- Department of Pathology, School of Medicine, Saint Louis University, MO 63104, USA
| | - Shi-Jun Wang
- Shandong University of Traditional Chinese Medicine, Jinan 250035, China
| | - Zi-Li Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shi-Zhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
13
|
Ganesan M, New-Aaron M, Dagur RS, Makarov E, Wang W, Kharbanda KK, Kidambi S, Poluektova LY, Osna NA. Alcohol Metabolism Potentiates HIV-Induced Hepatotoxicity: Contribution to End-Stage Liver Disease. Biomolecules 2019; 9:851. [PMID: 31835520 PMCID: PMC6995634 DOI: 10.3390/biom9120851] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
In an era of improved survival due to modern antiretroviral therapy, liver disease has become a major cause of morbidity and mortality, resulting in death in 15-17% of human immunodeficiency virus (HIV)-infected patients. Alcohol enhances HIV-mediated liver damage and promotes the progression to advanced fibrosis and cirrhosis. However, the mechanisms behind these events are uncertain. Here, we hypothesize that ethanol metabolism potentiates accumulation of HIV in hepatocytes, causing oxidative stress and intensive apoptotic cell death. Engulfment of HIV-containing apoptotic hepatocytes by non-parenchymal cells (NPCs) triggers their activation and liver injury progression. This study was performed on primary human hepatocytes and Huh7.5-CYP cells infected with HIV-1ADA, and major findings were confirmed by pilot data obtained on ethanol-fed HIV-injected chimeric mice with humanized livers. We demonstrated that ethanol exposure potentiates HIV accumulation in hepatocytes by suppressing HIV degradation by lysosomes and proteasomes. This leads to increased oxidative stress and hepatocyte apoptosis. Exposure of HIV-infected apoptotic hepatocytes to NPCs activates the inflammasome in macrophages and pro-fibrotic genes in hepatic stellate cells. We conclude that while HIV and ethanol metabolism-triggered apoptosis clears up HIV-infected hepatocytes, continued generation of HIV-expressing apoptotic bodies may be detrimental for progression of liver inflammation and fibrosis due to constant activation of NPCs.
Collapse
Affiliation(s)
- Murali Ganesan
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.G.); (M.N.-A.); (R.S.D.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Moses New-Aaron
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.G.); (M.N.-A.); (R.S.D.); (K.K.K.)
- Department of Environmental, Agriculture and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Raghubendra Singh Dagur
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.G.); (M.N.-A.); (R.S.D.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Edward Makarov
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (E.M.); (W.W.); (L.Y.P.)
| | - Weimin Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (E.M.); (W.W.); (L.Y.P.)
| | - Kusum K. Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.G.); (M.N.-A.); (R.S.D.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, NE 68588, USA;
| | - Larisa Y. Poluektova
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (E.M.); (W.W.); (L.Y.P.)
| | - Natalia A. Osna
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; (M.G.); (M.N.-A.); (R.S.D.); (K.K.K.)
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Environmental, Agriculture and Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE 68105, USA
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68105, USA; (E.M.); (W.W.); (L.Y.P.)
| |
Collapse
|
14
|
Donohue TM, Osna NA, Kharbanda KK, Thomes PG. Lysosome and proteasome dysfunction in alcohol-induced liver injury. LIVER RESEARCH 2019; 3:191-205. [DOI: 10.1016/j.livres.2019.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
15
|
Yan S, Khambu B, Hong H, Liu G, Huda N, Yin XM. Autophagy, Metabolism, and Alcohol-Related Liver Disease: Novel Modulators and Functions. Int J Mol Sci 2019; 20:ijms20205029. [PMID: 31614437 PMCID: PMC6834312 DOI: 10.3390/ijms20205029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023] Open
Abstract
Alcohol-related liver disease (ALD) is caused by over-consumption of alcohol. ALD can develop a spectrum of pathological changes in the liver, including steatosis, inflammation, cirrhosis, and complications. Autophagy is critical to maintain liver homeostasis, but dysfunction of autophagy has been observed in ALD. Generally, autophagy is considered to protect the liver from alcohol-induced injury and steatosis. In this review, we will summarize novel modulators of autophagy in hepatic metabolism and ALD, including autophagy-mediating non-coding RNAs (ncRNAs), and crosstalk of autophagy machinery and nuclear factors. We will also discuss novel functions of autophagy in hepatocytes and non-parenchymal hepatic cells during the pathogenesis of ALD and other liver diseases.
Collapse
Affiliation(s)
- Shengmin Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Bilon Khambu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Honghai Hong
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Gang Liu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Nazmul Huda
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
16
|
Ma Y, Chai H, Ding Q, Qian Q, Yan Z, Ding B, Dou X, Li S. Hepatic SIRT3 Upregulation in Response to Chronic Alcohol Consumption Contributes to Alcoholic Liver Disease in Mice. Front Physiol 2019; 10:1042. [PMID: 31474877 PMCID: PMC6707764 DOI: 10.3389/fphys.2019.01042] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/30/2019] [Indexed: 12/20/2022] Open
Abstract
Background Alcoholic liver disease (ALD) is a type of chronic liver disease caused by chronic ethanol overconsumption. The pathogenesis of ALD is complex and there is no effective clinical treatment thus far. SIRT3 is an NAD+-dependent deacetylase primarily located inside mitochondria, and reports on the effect of chronic alcohol exposure on liver SIRT3 expression are scarce. This study aims to investigate the effect of chronic alcohol consumption on hepatic SIRT3 expression and its role in alcoholic-induced liver injury. Methods Using the Lieber-DeCarli mouse model of ALD, we analyzed the regulation of SIRT3 and the effect of liver-specific knocking-down of SIRT3 on alcohol-induced liver injury. HepG2 and AML12 hepatocytes were employed to detect the biological function of SIRT3 on alcohol-induced hepatic cytotoxicity and its potential mechanism. Results Chronic alcohol exposure led to hepatic SIRT3 upregulation and liver-specific SIRT3 knockdown alleviated alcoholic feeding-induced liver injury and lipid accumulation, which is associated with improved autophagy induction. In addition, autophagy induction contributed to the cytoprotective effect of SIRT3 knockdown on ethanol-induced hepatocyte cell death. Conclusion In summary, our data suggest that hepatic SIRT3 upregulation in response to chronic alcohol exposure and liver-specific SIRT3 knockdown, induced autophagy activation further alleviating alcoholic-induced liver injury, which represents a novel mechanism in this process.
Collapse
Affiliation(s)
- Yue Ma
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Laboratory Animal Center, Zhejiang Academy of Medical Sciences, Hangzhou, China
| | - Hui Chai
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinchao Ding
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qianyu Qian
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhaoyuan Yan
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Ding
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- College of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China.,Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
| | - Songtao Li
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China.,College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
17
|
Wang Y, Yang P, Zhang B, Ding Y, Lei S, Hou Y, Guan X, Li Q. Hepatic NPC1L1 overexpression attenuates alcoholic autophagy in mice. Mol Med Rep 2019; 20:3224-3232. [PMID: 31432115 PMCID: PMC6755247 DOI: 10.3892/mmr.2019.10549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/27/2019] [Indexed: 01/25/2023] Open
Abstract
Alcohol consumption causes liver steatosis in humans. Metabolic disorders of lipids are one of the factors that cause liver steatosis in hepatocytes. Hepatic Niemann‑Pick C1‑like 1 (NPC1L1) regulates lipid homeostasis in mammals. The relationship between NPC1L1 and autophagy in those with a history of alcohol abuse is unclear. The present study aimed to investigate the function of NPC1L1 in the activation of hepatic autophagy in a mouse model with a human (h)NPC1L1 transgene under alcohol feeding conditions. The mice expressing hNPC1L1 (Ad‑L1) or controls (Ad‑null) were created by retro‑orbital adenovirus injection. The Ad‑L1 and Ad‑null mice were fed with alcohol or a non‑alcoholic diet to mimic chronic alcohol consumption in humans. Hepatic autophagy was demonstrated in isolated primary hepatocytes by monitoring autophagic vacuoles under fluorescence microscopy, and by western blotting for autophagic makers. Isolated hepatocytes from the livers of Ad‑L1 mice were treated with different doses of ezetimibe to study the restoration of autophagy. Chronic alcohol feeding caused liver injury and steatosis, shown by significantly higher levels of plasma alanine transaminase and aspartate transaminase activity, and by hematoxylin and eosin staining in Ad‑L1 and Ad‑null mice. Compared to Ad‑null control mice, the microtubule‑associated proteins 1A/1B light chain 3 (LC3) particles in the isolated hepatocytes of Ad‑L1 mice were decreased, both under alcohol and non‑alcoholic feeding. The ratio of LC3II/LC3I was significantly decreased, and the level of p62/sequestosome‑1 protein was significantly increased in Ad‑L1 mice compared with Ad‑null mice after alcohol feeding. Levels of LC3II protein were statistically increased in hepatocytes isolated from Ad‑L1 mice with ezetimibe treatment. The increase in LC3II expression was dose dependent. Within the tested range, it reached its highest level at 40 µM. The livers of Ad‑L1 mice represent a more human‑like state for the study of hepatic autophagy. Hepatic expression of human NPC1L1 resulted in an inhibition of autophagy; it may contribute to alcoholic fatty liver disease in humans.
Collapse
Affiliation(s)
- Youlin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Pan Yang
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Bo Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| | - Yao Ding
- Department of Oncology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Shun Lei
- Department of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Yuning Hou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaoqing Guan
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Qingwang Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P.R. China
| |
Collapse
|
18
|
Yan S, Zhou J, Chen X, Dong Z, Yin XM. Diverse Consequences in Liver Injury in Mice with Different Autophagy Functional Status Treated with Alcohol. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1744-1762. [PMID: 31199920 DOI: 10.1016/j.ajpath.2019.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023]
Abstract
Alcoholic fatty liver disease is often complicated by other pathologic insults, such as viral infection or high-fat diet. Autophagy plays a homeostatic role in the liver but can be compromised by alcohol, high-fat diet, or viral infection, which in turn affects the disease process caused by these etiologies. To understand the full impact of autophagy modulation on alcohol-induced liver injury, several genetic models of autophagy deficiency, which have different levels of functional alterations, were examined after acute binge or chronic-plus-binge treatment. Mice given alcohol with either mode and induced with deficiency in liver-specific Atg7 shortly after the induction of Atg7 deletion had elevated liver injury, indicating the protective role of autophagy. Constitutive hepatic Atg7-deficient mice, in which Atg7 was deleted in embryos, were more susceptible with chronic-plus-binge but not with acute alcohol treatment. Constitutive hepatic Atg5-deficient mice, in which Atg5 was deleted in embryos, were more susceptible with acute alcohol treatment, but liver injury was unexpectedly improved with the chronic-plus-binge regimen. A prolonged autophagy deficiency may complicate the hepatic response to alcohol treatment, likely in part due to endogenous liver injury. The complexity of the relationship between autophagy deficiency and alcohol-induced liver injury can thus be affected by the timing of autophagy dysfunction, the exact autophagy gene being affected, and the alcohol treatment regimen.
Collapse
Affiliation(s)
- Shengmin Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jun Zhou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Minimal Invasive Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyun Chen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China; Department of Cell Biology and Anatomy, Medical College of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
Yang L, Yang C, Thomes PG, Kharbanda KK, Casey CA, McNiven MA, Donohue TM. Lipophagy and Alcohol-Induced Fatty Liver. Front Pharmacol 2019; 10:495. [PMID: 31143122 PMCID: PMC6521574 DOI: 10.3389/fphar.2019.00495] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022] Open
Abstract
This review describes the influence of ethanol consumption on hepatic lipophagy, a selective form of autophagy during which fat-storing organelles known as lipid droplets (LDs) are degraded in lysosomes. During classical autophagy, also known as macroautophagy, all forms of macromolecules and organelles are sequestered in autophagosomes, which, with their cargo, fuse with lysosomes, forming autolysosomes in which the cargo is degraded. It is well established that excessive drinking accelerates intrahepatic lipid biosynthesis, enhances uptake of fatty acids by the liver from the plasma and impairs hepatic secretion of lipoproteins. All the latter contribute to alcohol-induced fatty liver (steatosis). Here, our principal focus is on lipid catabolism, specifically the impact of excessive ethanol consumption on lipophagy, which significantly influences the pathogenesis alcohol-induced steatosis. We review findings, which demonstrate that chronic ethanol consumption retards lipophagy, thereby exacerbating steatosis. This is important for two reasons: (1) Unlike adipose tissue, the liver is considered a fat-burning, not a fat-storing organ. Thus, under normal conditions, lipophagy in hepatocytes actively prevents lipid droplet accumulation, thereby maintaining lipostasis; (2) Chronic alcohol consumption subverts this fat-burning function by slowing lipophagy while accelerating lipogenesis, both contributing to fatty liver. Steatosis was formerly regarded as a benign consequence of heavy drinking. It is now recognized as the "first hit" in the spectrum of alcohol-induced pathologies that, with continued drinking, progresses to more advanced liver disease, liver failure, and/or liver cancer. Complete lipid droplet breakdown requires that LDs be digested to release their high-energy cargo, consisting principally of cholesteryl esters and triacylglycerols (triglycerides). These subsequently undergo lipolysis, yielding free fatty acids that are oxidized in mitochondria to generate energy. Our review will describe recent findings on the role of lipophagy in LD catabolism, how continuous heavy alcohol consumption affects this process, and the putative mechanism(s) by which this occurs.
Collapse
Affiliation(s)
- Li Yang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Changqing Yang
- Division of Gastroenterology and Hepatology, Digestive Disease Institute, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Paul G. Thomes
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Departments of Internal Medicine and of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kusum K. Kharbanda
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Departments of Internal Medicine and of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Carol A. Casey
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Departments of Internal Medicine and of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mark A. McNiven
- Division of Gastroenterology and Hepatology, Department of Biochemistry and Molecular Biology, Center for Basic Research in Digestive Diseases, Mayo Clinic, Rochester, MN, United States
| | - Terrence M. Donohue
- Research Service, Department of Veterans Affairs, Nebraska-Western Iowa Health Care System, Omaha, NE, United States
- Departments of Internal Medicine and of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
20
|
Thomes PG, Rasineni K, Yang L, Donohue TM, Kubik JL, McNiven MA, Casey CA. Ethanol withdrawal mitigates fatty liver by normalizing lipid catabolism. Am J Physiol Gastrointest Liver Physiol 2019; 316:G509-G518. [PMID: 30714813 PMCID: PMC6957361 DOI: 10.1152/ajpgi.00376.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
We are investigating the changes in hepatic lipid catabolism that contribute to alcohol-induced fatty liver. Following chronic ethanol (EtOH) exposure, abstinence from alcohol resolves steatosis. Here, we investigated the hepatocellular events that lead to this resolution by quantifying specific catabolic parameters that returned to control levels after EtOH was withdrawn. We hypothesized that, after its chronic consumption, EtOH withdrawal reactivates lipid catabolic processes that restore lipostasis. Male Wistar rats were fed control and EtOH liquid diets for 6 wk. Randomly chosen EtOH-fed rats were then fed control diet for 7 days. Liver triglycerides (TG), lipid peroxides, key markers of fatty acid (FA) metabolism, lipophagy, and autophagy were quantified. Compared with controls, EtOH-fed rats had higher hepatic triglycerides, lipid peroxides, and serum free fatty acids (FFA). The latter findings were associated with higher levels of FA transporters (FATP 2, 4, and 5) but lower quantities of peroxisome proliferator-activated receptor-α (PPAR-α), which governs FA oxidation. EtOH-fed animals also had lower nuclear levels of the autophagy-regulating transcription factor EB (TFEB), associated with lower hepatic lipophagy and autophagy. After EtOH-fed rats were refed control diet for 7 days, their serum FFA levels and those of FATPs fell to control (normal) levels, whereas PPAR-α levels rose to normal. Hepatic TG and malondialdehyde levels in EtOH-withdrawn rats declined to near control levels. EtOH withdrawal restored nuclear TFEB content, hepatic lipophagy, and autophagy activity to control levels. EtOH withdrawal reversed aberrant FA metabolism and restored lysosomal function to promote resolution of alcohol-induced fatty liver. NEW & NOTEWORTHY Here, using an animal model, we show mechanisms of reversal of fatty liver and injury following EtOH withdrawal. Our data indicate that reactivation of autophagy and lysosome function through the restoration of transcription factor EB contribute to reversal of fatty liver and injury following EtOH withdrawal.
Collapse
Affiliation(s)
- Paul G. Thomes
- 1The Liver Study Unit, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska,2Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Karuna Rasineni
- 1The Liver Study Unit, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska,2Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Li Yang
- 7Departmentof Internal Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Terrence M. Donohue
- 1The Liver Study Unit, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska,2Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska,3Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska,4Pathology and Microbiology; College of Medicine; University of Nebraska Medical Center, Omaha, Nebraska,5The Center for Environmental Toxicology; College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jacy L. Kubik
- 1The Liver Study Unit, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska
| | - Mark A. McNiven
- 6Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Carol A. Casey
- 1The Liver Study Unit, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska,2Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska,3Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
21
|
Khambu B, Wang L, Zhang H, Yin XM. The Activation and Function of Autophagy in Alcoholic Liver Disease. Curr Mol Pharmacol 2019; 10:165-171. [PMID: 26278385 DOI: 10.2174/1874467208666150817112654] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/29/2015] [Accepted: 08/07/2015] [Indexed: 02/07/2023]
Abstract
Autophagy is an important lysosome-mediated intracellular degradation pathway required for tissue homeostasis. Dysregulation of liver autophagy is closely associated with different liver diseases including alcoholic liver disease. Studies now indicate that autophagy may be induced or suppressed depending on the amount and the duration of ethanol treatment. Autophagy induced by ethanol serves as a protective mechanism, probably by selective degradation of the damaged mitochondria (mitophagy) and excess lipid droplets (lipophagy) and in turn attenuates alcohol-induced steatosis and liver injury. However, the detailed molecular mechanism of selective targeting of mitochondria and lipid is still unclear. Autophagy may possess other functions that protect hepatocytes from ethanol. Understanding these molecular entities would be essential in order to therapeutically module autophagy for treatment of alcoholic liver disease.
Collapse
Affiliation(s)
- Bilon Khambu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202. United States
| | - Lin Wang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202. United States
| | - Hao Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202. United States
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202. United States
| |
Collapse
|
22
|
Quercetin ameliorates autophagy in alcohol liver disease associated with lysosome through mTOR-TFEB pathway. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.10.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
23
|
Ethanol and C2 ceramide activate fatty acid oxidation in human hepatoma cells. Sci Rep 2018; 8:12923. [PMID: 30150688 PMCID: PMC6110824 DOI: 10.1038/s41598-018-31025-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/04/2018] [Indexed: 02/07/2023] Open
Abstract
Obesogenic lipids and the sphingolipid ceramide have been implicated as potential cofactors in alcoholic liver disease (ALD) patients. However, the mechanisms by which these lipids modulate lipid trafficking in ethanol-treated human liver cells to promote steatosis, an early stage of ALD, are poorly understood. We measured fatty acid (FA) uptake, triglyceride export, FA synthesis and FA oxidation in human hepatoma (VL-17A) cells in response to ethanol and the exogenous lipids oleate, palmitate and C2 ceramide. We found that in combination with ethanol, both oleate and palmitate promote lipid droplet accumulation while C2 ceramide inhibits lipid droplet accumulation by enhancing FA oxidation. Further, using both a pharmacologic and siRNA approach to reduce peroxisome proliferator-activated receptors α (PPARα) gene expression, we demonstrate that C2 ceramide abrogates ethanol-mediated suppression of FA oxidation through an indirect PPARα mechanism. Together, these data suggest that lipids interact differentially with ethanol to modulate hepatocellular lipid droplet accumulation and may provide novel targets for preventing the earliest stage of alcoholic liver disease, alcoholic steatosis.
Collapse
|
24
|
Eid N, Kondo Y. Ethanol-induced mitophagy in rat Sertoli cells: Implications for male fertility. Andrologia 2018; 50:e12820. [PMID: 28488740 DOI: 10.1111/and.12820] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2017] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a pro-survival mechanism involving lysosomal degradation of damaged cellular components following multiple forms of cellular stress. There is currently a lack of literature on the mechanism, and specifically on mitophagy (selective autophagy of damaged pro-apoptotic mitochondria) in Sertoli cells (SCs). Against such a background, the authors induced mitophagy in SCs of adult male rats using a single injection of ethanol (5 g/kg) and observed mitophagy in the SCs via transmission electron microscopy 24 hr later. In addition, we briefly discussed the possible clinical implications of enhanced autophagy and mitophagy in stressed SCs in our model and in other models of acute stress (e.g., heat and transplantation stress). Further studies on SC autophagy are required, as a full understanding of the molecular mechanisms controlling autophagy in stressed SCs may have therapeutic implications for infertility treatment.
Collapse
Affiliation(s)
- N Eid
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Y Kondo
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Takatsuki, Osaka, Japan
| |
Collapse
|
25
|
Menk M, Graw JA, Poyraz D, Möbius N, Spies CD, von Haefen C. Chronic Alcohol Consumption Inhibits Autophagy and Promotes Apoptosis in the Liver. Int J Med Sci 2018; 15:682-688. [PMID: 29910672 PMCID: PMC6001414 DOI: 10.7150/ijms.25393] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/25/2018] [Indexed: 01/27/2023] Open
Abstract
Background: Chronic alcohol consumption is a major cause of liver injury. However, the molecular mechanisms by which alcohol impairs hepatocellular function and induces cell death remain unclear. Macroautophagy (hereafter called 'autophagy') is a degradation pathway involved in the survival or death of cells during conditions of cellular stress. This study examines the effect of chronic alcohol consumption on hepatocellular autophagy in an animal model. Methods: During a 12-week period male Wistar rats were fed a Lieber-DeCarli diet containing 5% alcohol (EtOH group; n=10), or an isocaloric diet (control group; n=10). Hepatic expression of key regulatory autophagy proteins (e.g. Beclin-1, ATG-3, ATG-5, p62/SQSTM1 and LC3) were detected by real-time polymerase chain reaction and Western blot analysis. Markers of cellular stress and apoptotic cell death (e.g. HO-1, caspase-3, PARP-1 and Bcl-2) were determined, and levels of reduced and oxidized glutathione were measured. Results: Chronic alcohol consumption caused cellular and oxidative stress in the liver. Transcriptional and translational expression of Beclin-1 and ATG-5 was significantly impaired. The protein expression of LC3-I and LC3-II was significantly increased, while the ratio of LC3I/II remained unchanged in the EtOH group compared with controls. Hepatocellular expression of p62/SQSTM1 and markers of apoptotic cell death (such as cleaved caspase-3 and cleaved PARP-1) were significantly increased in the EtOH group indicating a disrupted autophagic flux and increased rate of apoptosis in the liver. Conclusions: In this model, chronic alcohol consumption impaired hepatocellular autophagy and induced apoptotic cell death. It appears that changes in autophagy might contribute to alcohol-induced structural and functional hepatocellular injury.
Collapse
Affiliation(s)
- Mario Menk
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jan Adriaan Graw
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| | - Deniz Poyraz
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| | - Nadine Möbius
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| | - Claudia D Spies
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM/CVK), Charité - University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Campus Virchow-Klinikum; Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
26
|
Eid N, Ito Y, Horibe A, Hamaoka H, Kondo Y. A Method for In Vivo Induction and Ultrastructural Detection of Mitophagy in Sertoli Cells. Methods Mol Biol 2018; 1748:103-112. [PMID: 29453568 DOI: 10.1007/978-1-4939-7698-0_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An emerging body of evidences based on in vitro studies indicate that mitophagy (selective autophagic clearance of damaged mitochondria) is a prosurvival mechanism, specifically under exposure to various stressors. Sertoli cells (SCs) play essential roles in maintenance of spermatogenesis via paracrine interactions with germ cells and other somatic cells in the testis; however, studies investigating mitophagy in SCs are still very few. In this chapter, we give a brief review of mechanisms and detection methods of mitophagy in SCs based on our recent publications on animal models of ethanol toxicity and current literature. In addition, we provide a method for induction and ultrastructural identification of mitophagy in SCs of adult Wistar rats using a single intraperitoneal injection (5 g/kg) of ethanol. Proper understanding of mitophagy features and mechanisms in SCs may have therapeutic implications for infertility associated with alcoholism and other diseases characterized by mitochondrial dysfunction.
Collapse
Affiliation(s)
- Nabil Eid
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Osaka, Japan.
| | - Yuko Ito
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Osaka, Japan
| | - Akio Horibe
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Osaka, Japan
| | - Hitomi Hamaoka
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Osaka, Japan
| | - Yoichi Kondo
- Division of Life Sciences, Department of Anatomy and Cell Biology, Osaka Medical College, Osaka, Japan
| |
Collapse
|
27
|
Wang L, Zhou J, Yan S, Lei G, Lee CH, Yin XM. Ethanol-triggered Lipophagy Requires SQSTM1 in AML12 Hepatic Cells. Sci Rep 2017; 7:12307. [PMID: 28951592 PMCID: PMC5614958 DOI: 10.1038/s41598-017-12485-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022] Open
Abstract
Ethanol-induced hepatic lipophagy plays an important cytoprotective role against liver injury, but its mechanism is not fully determined. In the present study, ethanol-induced lipophagy was studied in an immortalized mouse hepatocyte line, AML12. We found that ethanol treatment elevated lipid content in these cells, which could be regulated by autophagy. To determine the potential mechanism, we investigated the role of a key adaptor molecule SQSTM1/p62. SQSTM1 can bind to LC3 on autophagosomes and ubiquitinated molecules on cargos, thus facilitating the autophagic engulfment of the cargo. We found that both LC3 and SQSTM1 could colocalize with lipid droplets (LDs) following ethanol treatment. Colocalization of LC3 with LDs was significantly inhibited by SQSTM1 knockdown, which also reduced ethanol-induced lipid elevation. In addition, increased ubiquitin signals were found to colocalize with SQSTM1 on LDs in response to ethanol. Moreover, the SQSTM1 signal was colocalized with that of perilipin1, a major protein on LDs. Finally, perilipin1 knockdown significantly altered ethanol-induced lipophagy. Taken together, these data support a model in which autophagosomes were directed to the LDs via SQSTM1, which bound to ubiquitinated proteins, possibly including perilipin 1, on LDs. This study provides a potential mechanistic explanation to how ethanol induces lipophagy in hepatocytes.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,College of Animal Science and Veterinary Medicine, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China.,Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China.,Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City, Shandong Province, 271018, China
| | - Jun Zhou
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Center of Minimally Invasive Surgery, Xiangya 2nd Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shengmin Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Guangsheng Lei
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chao-Hung Lee
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
28
|
Rasineni K, Donohue TM, Thomes PG, Yang L, Tuma DJ, McNiven MA, Casey CA. Ethanol-induced steatosis involves impairment of lipophagy, associated with reduced Dynamin2 activity. Hepatol Commun 2017; 1:501-512. [PMID: 29152606 PMCID: PMC5678901 DOI: 10.1002/hep4.1063] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Lipid droplets (LDs), the organelles central to alcoholic steatosis, are broken down by lipophagy, a specialized form of autophagy. Here, we hypothesize that ethanol administration retards lipophagy by down-regulating Dynamin 2 (Dyn2), a protein that facilitates lysosome re-formation, contributing to hepatocellular steatosis. METHODS Primary hepatocytes were isolated from male Wistar rats fed Lieber-DeCarli control or EtOH liquid diets for 6-8 wk. Hepatocytes were incubated in complete medium (fed) or nutrient-free medium (fasting) with or without the Dyn2 inhibitor Dynasore or the Src inhibitor SU6656. Phosphorylated (active) forms of Src and Dyn2, and markers of autophagy were quantified by Western Blot. Co-localization of LDs-with autophagic machinery was determined by confocal microscopy. RESULTS In hepatocytes from pair-fed rats, LD breakdown was accelerated during fasting, as judged by smaller LDs and lower TG content when compared to hepatocytes in complete media. Fasting-induced TG loss in control hepatocytes was significantly blocked by either SU6656 or Dynasore. Compared to controls, hepatocytes from EtOH-fed rats had 66% and 40% lower content of pSrc and pDyn2, respectively, coupled with lower rate of fasting-induced TG loss. This slower rate of fasting-induced TG loss was blocked in cells co-incubated with Dynasore. Microscopic examination of EtOH-fed rat hepatocytes revealed increased co-localization of the autophagosome marker LC3 on LDs with a concomitant decrease in lysosome marker LAMP1. Whole livers and LD fractions of EtOH-fed rats exhibited simultaneous increase in LC3II and p62 over that of controls, indicating a block in lipophagy. CONCLUSION Chronic ethanol administration slowed the rate of hepatocyte lipophagy, owing in part to lower levels of phosphorylated Src kinase available to activate its substrate, Dyn2, thereby causing depletion of lysosomes for LD breakdown.
Collapse
Affiliation(s)
- Karuna Rasineni
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
| | - Terrence M. Donohue
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNE
- Department of Pathology and MicrobiologyCollege of Medicine, University of Nebraska Medical CenterOmahaNE
- Center for Environmental ToxicologyCollege of Public Health, University of Nebraska Medical CenterOmahaNE
| | - Paul G. Thomes
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
| | - Li Yang
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
- Tongji HospitalTongji University School of MedicineShanghaiChina
| | - Dean J. Tuma
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
| | - Mark A. McNiven
- Department of Biochemistry and Molecular BiologyMayo Clinic College of MedicineRochesterMN
| | - Carol A. Casey
- The Liver Study UnitVA Nebraska‐Western Iowa Health Care System (VA NWIHCS)OmahaNE
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNE
| |
Collapse
|
29
|
Horibe A, Eid N, Ito Y, Hamaoka H, Tanaka Y, Kondo Y. Upregulated Autophagy in Sertoli Cells of Ethanol-Treated Rats Is Associated with Induction of Inducible Nitric Oxide Synthase (iNOS), Androgen Receptor Suppression and Germ Cell Apoptosis. Int J Mol Sci 2017; 18:1061. [PMID: 28505146 PMCID: PMC5454973 DOI: 10.3390/ijms18051061] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/22/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022] Open
Abstract
This study was conducted to investigate the autophagic response of Sertoli cells (SCs) to acute ethanol toxicity using in vivo and in vitro models. Adult Wistar rats were intraperitoneally injected with either 5 g/kg ethanol or phosphate-buffered saline (for the control group) and sacrificed 0, 3, 6 and 24 h after injection. Compared to the control group, enhanced germ cell apoptosis was observed in the ethanol-treated rats (ETRs) in association with upregulation of iNOS and reduced expression of androgen receptor protein levels in SCs, which were resistant to apoptosis. Meanwhile, autophagy was upregulated in ETR SCs (peaking at 24 h) compared to the control group, as evidenced by transcription factor EB (TFEB) nuclear translocation, enhanced expression of microtubule-associated protein 1 light chain3-II (LC3-II), lysosome-associated membrane protein-2 (LAMP-2), pan cathepsin protein levels and reduced expression of p62. This upregulation of SC autophagy was confirmed ultrastructurally by enhanced formation of autophagic vacuoles and by immunofluorescent double labelling of autophagosomal and lysosomal markers. Study of cultured SCs confirmed enhanced autophagic response to ethanol toxicity, which was cytoprotective based on decreased viability of SCs upon blocking autophagy with 3-methyladenine (3-MA). The results highlighted the molecular mechanisms of prosurvival autophagy in ETR SCs for the first time, and may have significant implications for male fertility.
Collapse
Affiliation(s)
- Akio Horibe
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Nabil Eid
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Yuko Ito
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Hitomi Hamaoka
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Yoshihisa Tanaka
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Yoichi Kondo
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| |
Collapse
|
30
|
Yan S, Huda N, Khambu B, Yin XM. Relevance of autophagy to fatty liver diseases and potential therapeutic applications. Amino Acids 2017; 49:1965-1979. [PMID: 28478585 DOI: 10.1007/s00726-017-2429-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022]
Abstract
Autophagy is an evolutionarily conserved lysosome-mediated cellular degradation program. Accumulating evidence shows that autophagy is important to the maintenance of liver homeostasis. Autophagy involves recycling of cellular nutrients recycling as well as quality control of subcellular organelles. Autophagy deficiency in the liver causes various liver pathologies. Fatty liver disease (FLD) is characterized by the accumulation of lipids in hepatocytes and the dysfunction in energy metabolism. Autophagy is negatively affected by the pathogenesis of FLD and the activation of autophagy could ameliorate steatosis, which suggests a potential therapeutic approach to FLD. In this review, we will discuss autophagy and its relevance to liver diseases, especially FLD. In addition, we will discuss recent findings on potential therapeutic applications of autophagy modulators for FLD.
Collapse
Affiliation(s)
- Shengmin Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nazmul Huda
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Bilon Khambu
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiao-Ming Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
31
|
Schulze RJ, Rasineni K, Weller SG, Schott MB, Schroeder B, Casey CA, McNiven MA. Ethanol exposure inhibits hepatocyte lipophagy by inactivating the small guanosine triphosphatase Rab7. Hepatol Commun 2017; 1:140-152. [PMID: 29404450 PMCID: PMC5721426 DOI: 10.1002/hep4.1021] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/29/2017] [Indexed: 12/20/2022] Open
Abstract
Alcohol consumption is a well-established risk factor for the onset and progression of fatty liver disease. An estimated 90% of heavy drinkers are thought to develop significant liver steatosis. For these reasons, an increased understanding of the molecular basis for alcohol-induced hepatic steatosis is important. It has become clear that autophagy, a catabolic process of intracellular degradation and recycling, plays a key role in hepatic lipid metabolism. We have shown that Rab7, a small guanosine triphosphatase known to regulate membrane trafficking, acts as a key orchestrator of hepatocellular lipophagy, a selective form of autophagy in which lipid droplets (LDs) are specifically targeted for turnover by the autophagic machinery. Nutrient starvation results in Rab7 activation on the surface of the LD and lysosomal compartments, resulting in the mobilization of triglycerides stored within the LDs for energy production. Here, we examine whether the steatotic effects of alcohol exposure are a result of perturbations to the Rab7-mediated lipophagic pathway. Rats chronically fed an ethanol-containing diet accumulated significantly higher levels of fat in their hepatocytes. Interestingly, hepatocytes isolated from these ethanol-fed rats contained juxtanuclear lysosomes that exhibited impaired motility. These changes are similar to those we observed in Rab7-depleted hepatocytes. Consistent with these defects in the lysosomal compartment, we observed a marked 80% reduction in Rab7 activity in cultured hepatocytes as well as a complete block in starvation-induced Rab7 activation in primary hepatocytes isolated from chronic ethanol-fed animals. Conclusion: A mechanism is supported whereby ethanol exposure inhibits Rab7 activity, resulting in the impaired transport, targeting, and fusion of the autophagic machinery with LDs, leading to an accumulation of hepatocellular lipids and hepatic steatosis. (Hepatology Communications 2017;1:140-152).
Collapse
Affiliation(s)
- Ryan J. Schulze
- Department of Biochemistry and Molecular Biology and the Center for Digestive DiseasesMayo ClinicRochesterMN
| | - Karuna Rasineni
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
| | - Shaun G. Weller
- Department of Biochemistry and Molecular Biology and the Center for Digestive DiseasesMayo ClinicRochesterMN
| | - Micah B. Schott
- Department of Biochemistry and Molecular Biology and the Center for Digestive DiseasesMayo ClinicRochesterMN
| | - Barbara Schroeder
- Department of Biochemistry and Molecular Biology and the Center for Digestive DiseasesMayo ClinicRochesterMN
- Present address:
Helmholtz Zentrum München, Institute of Biological and Medical ImagingNeuherbergGermany
| | - Carol A. Casey
- Department of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE
- Research Service, VA Nebraska‐Western Iowa Health Care SystemOmahaNE
| | - Mark A. McNiven
- Department of Biochemistry and Molecular Biology and the Center for Digestive DiseasesMayo ClinicRochesterMN
| |
Collapse
|
32
|
Girault V, Gilard V, Marguet F, Lesueur C, Hauchecorne M, Ramdani Y, Laquerrière A, Marret S, Jégou S, Gonzalez BJ, Brasse-Lagnel C, Bekri S. Prenatal alcohol exposure impairs autophagy in neonatal brain cortical microvessels. Cell Death Dis 2017; 8:e2610. [PMID: 28182007 PMCID: PMC5386476 DOI: 10.1038/cddis.2017.29] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/31/2016] [Accepted: 01/10/2017] [Indexed: 02/06/2023]
Abstract
Brain developmental lesions are a devastating consequence of prenatal alcohol exposure (PAE). We recently showed that PAE affects cortical vascular development with major effects on angiogenesis and endothelial cell survival. The underlying molecular mechanisms of these effects remain poorly understood. This study aimed at characterizing the ethanol exposure impact on the autophagic process in brain microvessels in human fetuses with fetal alcohol syndrome (FAS) and in a PAE mouse model. Our results indicate that PAE induces an increase of autophagic vacuole number in human fetal and neonatal mouse brain cortical microvessels. Subsequently, ex vivo studies using green fluorescent protein (GFP)-LC3 mouse microvessel preparations revealed that ethanol treatment alters autophagy in endothelial cells. Primary cultures of mouse brain microvascular endothelial cells were used to characterize the underlying molecular mechanisms. LC3 and p62 protein levels were significantly increased in endothelial cells treated with 50 mM ethanol. The increase of autophagic vacuole number may be due to excessive autophagosome formation associated with the partial inhibition of the mammalian target of rapamycin pathway upon ethanol exposure. In addition, the progression from autophagosomes to autolysosomes, which was monitored using autophagic flux inhibitors and mRFP-EGFP vector, showed a decrease in the autolysosome number. Besides, a decrease in the Rab7 protein level was observed that may underlie the impairment of autophagosome-lysosome fusion. In addition, our results showed that ethanol-induced cell death is likely to be mediated by decreased mitochondrial integrity and release of apoptosis-inducing factor. Interestingly, incubation of cultured cells with rapamycin prevented ethanol effects on autophagic flux, ethanol-induced cell death and vascular plasticity. Taken together, these results are consistent with autophagy dysregulation in cortical microvessels upon ethanol exposure, which could contribute to the defects in angiogenesis observed in patients with FAS. Moreover, our results suggest that rapamycin represents a potential therapeutic strategy to reduce PAE-related brain developmental disorders.
Collapse
Affiliation(s)
- Virginie Girault
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
| | - Vianney Gilard
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Department of Neurosurgery, Rouen University Hospital, Rouen, France
| | - Florent Marguet
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Pathology Laboratory, Rouen University Hospital, Rouen, France
| | - Céline Lesueur
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | | | - Yasmina Ramdani
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
| | - Annie Laquerrière
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Pathology Laboratory, Rouen University Hospital, Rouen, France
| | - Stéphane Marret
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Department of Neonatal Pediatrics and Intensive Care and Neuropediatrics, Rouen University Hospital, Rouen, France
| | - Sylvie Jégou
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
| | | | | | - Soumeya Bekri
- Normandie University, UNIROUEN, INSERM U1245, NeoVasc Team, Rouen, France
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| |
Collapse
|
33
|
Neuman MG, French SW, Zakhari S, Malnick S, Seitz HK, Cohen LB, Salaspuro M, Voinea-Griffin A, Barasch A, Kirpich IA, Thomes PG, Schrum LW, Donohue TM, Kharbanda KK, Cruz M, Opris M. Alcohol, microbiome, life style influence alcohol and non-alcoholic organ damage. Exp Mol Pathol 2017; 102:162-180. [PMID: 28077318 DOI: 10.1016/j.yexmp.2017.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 02/06/2023]
Abstract
This paper is based upon the "8th Charles Lieber's Satellite Symposium" organized by Manuela G. Neuman at the Research Society on Alcoholism Annual Meeting, on June 25, 2016 at New Orleans, Louisiana, USA. The integrative symposium investigated different aspects of alcohol-induced liver disease (ALD) as well as non-alcohol-induced liver disease (NAFLD) and possible repair. We revealed the basic aspects of alcohol metabolism that may be responsible for the development of liver disease as well as the factors that determine the amount, frequency and which type of alcohol misuse leads to liver and gastrointestinal diseases. We aimed to (1) describe the immuno-pathology of ALD, (2) examine the role of genetics in the development of alcoholic hepatitis (ASH) and NAFLD, (3) propose diagnostic markers of ASH and non-alcoholic steatohepatitis (NASH), (4) examine age and ethnic differences as well as analyze the validity of some models, (5) develop common research tools and biomarkers to study alcohol-induced effects, 6) examine the role of alcohol in oral health and colon and gastrointestinal cancer and (7) focus on factors that aggravate the severity of organ-damage. The present review includes pre-clinical, translational and clinical research that characterizes ALD and NAFLD. Strong clinical and experimental evidence lead to recognition of the key toxic role of alcohol in the pathogenesis of ALD with simple fatty infiltrations and chronic alcoholic hepatitis with hepatic fibrosis or cirrhosis. These latter stages may also be associated with a number of cellular and histological changes, including the presence of Mallory's hyaline, megamitochondria, or perivenular and perisinusoidal fibrosis. Genetic polymorphisms of ethanol metabolizing enzymes and cytochrome p450 (CYP) 2E1 activation may change the severity of ASH and NASH. Other risk factors such as its co-morbidities with chronic viral hepatitis in the presence or absence of human deficiency virus were discussed. Dysregulation of metabolism, as a result of ethanol exposure, in the intestine leads to colon carcinogenesis. The hepatotoxic effects of ethanol undermine the contribution of malnutrition to the liver injury. Dietary interventions such as micro and macronutrients, as well as changes to the microbiota have been suggested. The clinical aspects of NASH, as part of the metabolic syndrome in the aging population, have been presented. The symposium addressed mechanisms and biomarkers of alcohol induced damage to different organs, as well as the role of the microbiome in this dialog. The microbiota regulates and acts as a key element in harmonizing immune responses at intestinal mucosal surfaces. It is known that microbiota is an inducer of proinflammatory T helper 17 cells and regulatory T cells in the intestine. The signals at the sites of inflammation mediate recruitment and differentiation in order to remove inflammatory inducers and promote tissue homeostasis restoration. The change in the intestinal microbiota also influences the change in obesity and regresses the liver steatosis. Evidence on the positive role of moderate alcohol consumption on heart and metabolic diseases as well on reducing steatosis have been looked up. Moreover nutrition as a therapeutic intervention in alcoholic liver disease has been discussed. In addition to the original data, we searched the literature (2008-2016) for the latest publication on the described subjects. In order to obtain the updated data we used the usual engines (Pub Med and Google Scholar). The intention of the eighth symposia was to advance the international profile of the biological research on alcoholism. We also wish to further our mission of leading the forum to progress the science and practice of translational research in alcoholism.
Collapse
Affiliation(s)
- Manuela G Neuman
- In Vitro Drug Safety and Biotechnology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | | - Stephen Malnick
- Department Internal Medicine, Kaplan Medical Centre and Hebrew University of Jerusalem, Rehovot, Israel
| | - Helmut K Seitz
- Centre of Alcohol Research, University of Heidelberg, Heidelberg, Germany
| | - Lawrence B Cohen
- Division of Gastroenterology, Sunnybrook Health Sciences Centre, Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mikko Salaspuro
- Research Unit on Acetaldehyde and Cancer, University of Helsinki, Helsinki, Finland
| | - Andreea Voinea-Griffin
- Public Health Science Texas A&M University, College of Dentistry, Dallas University, TX, USA
| | - Andrei Barasch
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Irina A Kirpich
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Paul G Thomes
- Department of Internal Medicine, Carolinas Medical Center, Charlotte, NC, USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Laura W Schrum
- Department of Internal Medicine, Carolinas Medical Center, Charlotte, NC, USA
| | - Terrence M Donohue
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kusum K Kharbanda
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, University of Nebraska Medical Center, Omaha, NE, USA; Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Marcus Cruz
- In Vitro Drug Safety and Biotechnology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mihai Opris
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Family Medicine Clinic CAR, Bucharest, Romania
| |
Collapse
|
34
|
Thomes PG, Brandon-Warner E, Li T, Donohue TM, Schrum LW. Rev-erb agonist and TGF-β similarly affect autophagy but differentially regulate hepatic stellate cell fibrogenic phenotype. Int J Biochem Cell Biol 2016; 81:137-147. [PMID: 27840152 DOI: 10.1016/j.biocel.2016.11.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 10/28/2016] [Accepted: 11/09/2016] [Indexed: 01/18/2023]
Abstract
We demonstrated that ligand-activated nuclear receptor Rev-erbα mitigates CCl4-induced liver fibrosis. Rev-erbα is also a novel regulator of autophagy, a crucial eukaryotic catabolic system in which lysosomes degrade substrates for energy generation. In hepatic stellate cells (HSC) autophagy is reportedly required for this purpose to activate HSCs during fibrogenesis. Here, we examined whether pharmacological activation of Rev-erb with its synthetic ligand SR9009 or treatment with the pro-fibrotic cytokine, TGF-β, each differentially modulate autophagy to regulate the HSC phenotype. We measured the effects of SR9009 on autophagy markers in a CCl4-induced liver fibrosis model. Using primary and immortalized HSCs in vitro, we quantified SR9009 and TGF-β effects on autophagy flux. Compared with vehicle-treated controls, livers from CCl4-treated mice exhibited lower AMPK, higher P70S6K phosphorylation, elevated P62 and lower levels of ATG proteins, indicating a disruption of autophagosome (AV) formation. SR9009 treatment prevented CCl4-induced P70S6K phosphorylation but did not affect CCl4-induced changes in AMPK, ATG proteins or P62. Analysis of autophagy markers and autophagy flux in primary HSCs or an immortalized human HSC line (LX2), revealed that SR9009 exposure down-regulated AV biogenesis. These events were associated with lower levels of fibrogenic gene expression, P70S6K phosphorylation and HSC proliferation. However, HSC exposure to TGF-β enhanced fibrogenic gene expression, P70S6K phosphorylation and HSC proliferation, while it simultaneously decelerated AV synthesis. The autophagy activator rapamycin and the autophagy inhibitor wortmannin each decreased HSC activation, P70S6K phosphorylation and HSC proliferation. Furthermore, knock-down of P70S6K using siRNA blocked basal and TGF-β-induced cell proliferation in human activated LX2. We conclude that SR9009 and TGF-β both similarly affected autophagy but, differentially regulated HSC fibrogenic phenotype through modulation of P70S6K, which is crucial for cell proliferation and fibrogenesis.
Collapse
Affiliation(s)
- Paul G Thomes
- Department of Internal Medicine, Carolinas Medical Center, Charlotte, NC, USA.
| | | | - Ting Li
- Department of Internal Medicine, Carolinas Medical Center, Charlotte, NC, USA
| | - Terrence M Donohue
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Laura W Schrum
- Department of Internal Medicine, Carolinas Medical Center, Charlotte, NC, USA.
| |
Collapse
|
35
|
Role of Autophagy in HIV Pathogenesis and Drug Abuse. Mol Neurobiol 2016; 54:5855-5867. [PMID: 27660273 DOI: 10.1007/s12035-016-0118-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 09/12/2016] [Indexed: 12/27/2022]
Abstract
Autophagy is a highly regulated process in which excessive cytoplasmic materials are captured and degraded during deprivation conditions. The unique nature of autophagy that clears invasive microorganisms has made it an important cellular defense mechanism in a variety of clinical situations. In recent years, it has become increasingly clear that autophagy is extensively involved in the pathology of HIV-1. To ensure survival of the virus, HIV-1 viral proteins modulate and utilize the autophagy pathway so that biosynthesis of the virus is maximized. At the same time, the abuse of illicit drugs such as methamphetamine, cocaine, morphine, and alcohol is thought to be a significant risk factor for the acquirement and progression of HIV-1. During drug-induced toxicity, autophagic activity has been proved to be altered in various cell types. Here, we review the current literature on the interaction between autophagy, HIV-1, and drug abuse and discuss the complex role of autophagy during HIV-1 pathogenesis in co-exposure to illicit drugs.
Collapse
|
36
|
Chigurupati H, Auddy B, Biyani M, Stohs SJ. Hepatoprotective Effects of a Proprietary Glycyrrhizin Product during Alcohol Consumption: A Randomized, Double-Blind, Placebo-Controlled, Crossover Study. Phytother Res 2016; 30:1943-1953. [DOI: 10.1002/ptr.5699] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/18/2016] [Accepted: 07/22/2016] [Indexed: 12/14/2022]
Affiliation(s)
| | - Biswajit Auddy
- Chigurupati Technologies Private Limited; Hyderabad India
| | - M. Biyani
- Chigurupati Technologies Private Limited; Hyderabad India
| | - Sidney J. Stohs
- Creighton University; 7068 Maumee Valley Court Frisco TX 75034 USA
| |
Collapse
|
37
|
Glade MJ, Meguid MM. A Glance At … ethanol consumption, GSH suppression, and oxidative liver damage. Nutrition 2016; 33:199-203. [PMID: 27644136 DOI: 10.1016/j.nut.2016.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 07/09/2016] [Indexed: 02/07/2023]
Affiliation(s)
| | - Michael M Meguid
- Professor Emeritus, Surgery, Neuroscience and Nutrition, Department of Surgery, University Hospital, Upstate Medical University, Syracuse, New York
| |
Collapse
|
38
|
Scheer MA, Schneider KJ, Finnigan RL, Maloney EP, Wells MA, Clemens DL. The Involvement of Acetaldehyde in Ethanol-Induced Cell Cycle Impairment. Biomolecules 2016; 6:biom6020017. [PMID: 27043646 PMCID: PMC4919912 DOI: 10.3390/biom6020017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 03/09/2016] [Accepted: 03/24/2016] [Indexed: 01/15/2023] Open
Abstract
Background: Hepatocytes metabolize the vast majority of ingested ethanol. This metabolic activity results in hepatic toxicity and impairs the ability of hepatocytes to replicate. Previous work by our group has shown that ethanol metabolism results in a G2/M cell cycle arrest. The intent of these studies was to discern the roles of acetaldehyde and reactive oxygen, two of the major by-products of ethanol metabolism, in the G2/M cell cycle arrest. Methods: To investigate the role of ethanol metabolites in the cell cycle arrest, VA-13 and VL-17A cells were used. These are recombinant Hep G2 cells that express alcohol dehydrogenase or alcohol dehydrogenase and cytochrome P450 2E1, respectively. Cells were cultured with or without ethanol, lacking or containing the antioxidants N-acetylcysteine (NAC) or trolox, for three days. Cellular accumulation was monitored by the DNA content of the cultures. The accumulation of the cyclin-dependent kinase, Cdc2 in the inactive phosphorylated form (p-Cdc2) and the cyclin-dependent kinase inhibitor p21 were determined by immunoblot analysis. Results: Cultures maintained in the presence of ethanol demonstrated a G2/M cell cycle arrest that was associated with a reduction in DNA content and increased levels of p-Cdc2 and p21, compared with cells cultured in its absence. Inclusion of antioxidants in the ethanol containing media was unable to rescue the cells from the cell cycle arrest or these ethanol metabolism-mediated effects. Additionally, culturing the cells in the presence of acetaldehyde alone resulted in increased levels of p-Cdc2 and p21. Conclusions: Acetaldehyde produced during ethanol oxidation has a major role in the ethanol metabolism-mediated G2/M cell cycle arrest, and the concurrent accumulation of p21 and p-Cdc2. Although reactive oxygen species are thought to have a significant role in ethanol-induced hepatocellular damage, they may have a less important role in the inability of hepatocytes to replace dead or damaged cells.
Collapse
Affiliation(s)
- Marc A Scheer
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Katrina J Schneider
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
- Nebraska and Western Iowa Veterans Administration Medical Center, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Rochelle L Finnigan
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Eamon P Maloney
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Mark A Wells
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| | - Dahn L Clemens
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA.
- Nebraska and Western Iowa Veterans Administration Medical Center, University of Nebraska Medical Center, Omaha, NE 68105, USA.
- Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68105, USA.
| |
Collapse
|
39
|
Clemens DL, Schneider KJ, Arkfeld CK, Grode JR, Wells MA, Singh S. Alcoholic pancreatitis: New insights into the pathogenesis and treatment. World J Gastrointest Pathophysiol 2016; 7:48-58. [PMID: 26909228 PMCID: PMC4753189 DOI: 10.4291/wjgp.v7.i1.48] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/23/2015] [Accepted: 11/11/2015] [Indexed: 02/06/2023] Open
Abstract
Acute pancreatitis is a necro-inflammatory disease of the exocrine pancreas that is characterized by inappropriate activation of zymogens, infiltration of the pancreas by inflammatory cells, and destruction of the pancreatic exocrine cells. Acute pancreatitis can progress to a severe life-threatening disease. Currently there is no pharmacotherapy to prevent or treat acute pancreatitis. One of the more common factors associated with acute pancreatitis is alcohol abuse. Although commonly associated with pancreatitis alcohol alone is unable to cause pancreatitis. Instead, it appears that alcohol and its metabolic by-products predispose the pancreas to damage from agents that normally do not cause pancreatitis, or to more severe disease from agents that normally cause mild pancreatic damage. Over the last 10 to 20 years, a tremendous amount of work has defined a number of alcohol-mediated biochemical changes in pancreatic cells. Among these changes are: Sustained levels of intracellular calcium, activation of the mitochondrial permeability transition pore, endoplasmic reticulum stress, impairment in autophagy, alteration in the activity of transcriptional activators, and colocalization of lysosomal and pancreatic digestive enzymes. Elucidation of these changes has led to a deeper understanding of the mechanisms by which ethanol predisposes acinar cells to damage. This greater understanding has revealed a number of promising targets for therapeutic intervention. It is hoped that further investigation of these targets will lead to the development of pharmacotherapy that is effective in treating and preventing the progression of acute pancreatitis.
Collapse
|
40
|
Abstract
Excessive ethanol exposure is detrimental to the brain. The developing brain is particularly vulnerable to ethanol such that prenatal ethanol exposure causes fetal alcohol spectrum disorders (FASD). Neuronal loss in the brain is the most devastating consequence and is associated with mental retardation and other behavioral deficits observed in FASD. Since alcohol consumption during pregnancy has not declined, it is imperative to elucidate the underlying mechanisms and develop effective therapeutic strategies. One cellular mechanism that acts as a protective response for the central nervous system (CNS) is autophagy. Autophagy regulates lysosomal turnover of organelles and proteins within cells, and is involved in cell differentiation, survival, metabolism, and immunity. We have recently shown that ethanol activates autophagy in the developing brain. The autophagic preconditioning alleviates ethanol-induced neuron apoptosis, whereas inhibition of autophagy potentiates ethanol-stimulated reactive oxygen species (ROS) and exacerbates ethanol-induced neuroapoptosis. The expression of genes encoding proteins required for autophagy in the CNS is developmentally regulated; their levels are much lower during an ethanol-sensitive period than during an ethanol-resistant period. Ethanol may stimulate autophagy through multiple mechanisms; these include induction of oxidative stress and endoplasmic reticulum stress, modulation of MTOR and AMPK signaling, alterations in BCL2 family proteins, and disruption of intracellular calcium (Ca2+) homeostasis. This review discusses the most recent evidence regarding the involvement of autophagy in ethanol-mediated neurotoxicity as well as the potential therapeutic approach of targeting autophagic pathways.
Collapse
Key Words
- AD, Alzheimer disease
- ALS, autophagy-lysosome system
- AMPK, adenosine 5′-monophosphate-activated protein kinase;
- ATG, autophagy-related
- CNS, central nervous system
- ER, endoplasmic reticulum
- FASD, fetal alcohol spectrum disorders
- FOXO3, forkhead box O3
- GSK3B, glycogen synthase kinase 3 β
- HD, Huntington disease, HNSCs, hippocampal neural stem cells
- LC3, microtubule-associated protein 1 light chain 3
- MTOR, mechanistic target of rapamycin (serine/threonine kinase)
- MTORC1, MTOR complex 1
- NFE2L2, nuclear factor, erythroid 2-like 2
- NOX, NADPH oxidase
- PD, Parkinson disease
- PI3K, class I phosphoinositide 3-kinase
- ROS, reactive oxygen species
- SQSTM1/p62, sequestosome 1
- TSC1/2, tuberous sclerosis 1/ 2
- UPR, unfolded protein response
- alcohol
- alcoholism
- development
- fetal alcohol spectrum disorders
- neurodegeneration
- oxidative stress
- protein degradation
Collapse
Affiliation(s)
- Jia Luo
- a Department of Pharmacology and Nutritional Sciences ; University of Kentucky College of Medicine ; Lexington , KY USA
| |
Collapse
|
41
|
Thomes PG, Trambly CS, Fox HS, Tuma DJ, Donohue TM. Acute and Chronic Ethanol Administration Differentially Modulate Hepatic Autophagy and Transcription Factor EB. Alcohol Clin Exp Res 2015; 39:2354-63. [PMID: 26556759 DOI: 10.1111/acer.12904] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 09/10/2015] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic ethanol (EtOH) consumption decelerates the catabolism of long-lived proteins, indicating that it slows hepatic macroautophagy (hereafter called autophagy) a crucial lysosomal catabolic pathway in most eukaryotic cells. Autophagy and lysosome biogenesis are linked. Both are regulated by the transcription factor EB (TFEB). Here, we tested whether TFEB can be used as a singular indicator of autophagic activity, by quantifying its nuclear content in livers of mice subjected to acute and chronic EtOH administration. We correlated nuclear TFEB to specific indices of autophagy. METHODS In acute experiments, we gavaged GFP-LC3(tg) mice with a single dose of EtOH or with phosphate buffered saline (PBS). We fed mice chronically by feeding them control or EtOH liquid diets. RESULTS Compared with PBS-gavaged controls, livers of EtOH-gavaged mice exhibited greater autophagosome (AV) numbers, a higher incidence of AV-lysosome co-localization, and elevated levels of free GFP, all indicating enhanced autophagy, which correlated with a higher nuclear content of TFEB. Compared with pair-fed controls, livers of EtOH-fed mice exhibited higher AV numbers, but had lower lysosome numbers, lower AV-lysosome co-localization, higher P62/SQSTM1 levels, and lower free GFP levels. The latter findings correlated with lower nuclear TFEB levels in EtOH-fed mice. Thus, enhanced autophagy after acute EtOH gavage correlated with a higher nuclear TFEB content. Conversely, chronic EtOH feeding inhibited hepatic autophagy, associated with a lower nuclear TFEB content. CONCLUSIONS Our findings suggest that the effect of acute EtOH gavage on hepatic autophagy differs significantly from that after chronic EtOH feeding. Each regimen distinctly affects TFEB localization, which in turn, regulates hepatic autophagy and lysosome biogenesis.
Collapse
Affiliation(s)
- Paul G Thomes
- Liver Study Unit, Department of Veterans Affairs, VA Nebraska-Western Iowa Health Care System (NWIHCS), Omaha, Nebraska.,Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Casey S Trambly
- Liver Study Unit, Department of Veterans Affairs, VA Nebraska-Western Iowa Health Care System (NWIHCS), Omaha, Nebraska.,Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Howard S Fox
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dean J Tuma
- Liver Study Unit, Department of Veterans Affairs, VA Nebraska-Western Iowa Health Care System (NWIHCS), Omaha, Nebraska.,Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Terrence M Donohue
- Liver Study Unit, Department of Veterans Affairs, VA Nebraska-Western Iowa Health Care System (NWIHCS), Omaha, Nebraska.,Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.,The Center for Environmental Health and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
42
|
Eid N, Ito Y, Otsuki Y. Mitophagy in steatotic hepatocytes of ethanol-treated wild-type and Parkin knockout mice. Am J Physiol Gastrointest Liver Physiol 2015; 309:G513-G514. [PMID: 26374875 DOI: 10.1152/ajpgi.00254.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- Nabil Eid
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Daigaku machi, Takatsuki, Osaka, Japan
| | - Yuko Ito
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Daigaku machi, Takatsuki, Osaka, Japan
| | - Yoshinori Otsuki
- Department of Anatomy and Cell Biology, Division of Life Sciences, Osaka Medical College, Daigaku machi, Takatsuki, Osaka, Japan
| |
Collapse
|
43
|
Autophagy in alcoholic liver disease, self-eating triggered by drinking. Clin Res Hepatol Gastroenterol 2015; 39 Suppl 1:S2-6. [PMID: 26186877 PMCID: PMC5582974 DOI: 10.1016/j.clinre.2015.05.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 05/22/2015] [Accepted: 05/27/2015] [Indexed: 02/07/2023]
Abstract
Macroautophagy (autophagy) is an evolutionarily conserved mechanism. It is important for normal cellular function and also plays critical roles in the etiology and pathogenesis of a number of human diseases. In alcohol-induced liver disease, autophagy is a protective mechanism against the liver injury caused by alcohol. Autophagy is activated in acute ethanol treatment but could be suppressed in chronic and/or high dose treatment of alcohol. The selective removal of lipid droplets and/or damaged mitochondria is likely the major mode of autophagy in reducing liver injury. Understanding the dynamics of the autophagy process and the approach to modulate autophagy could help finding new ways to battle against alcohol-induced liver injury.
Collapse
|
44
|
Elucidating mechanisms of toxicity using phenotypic data from primary human cell systems--a chemical biology approach for thrombosis-related side effects. Int J Mol Sci 2015; 16:1008-29. [PMID: 25569083 PMCID: PMC4307287 DOI: 10.3390/ijms16011008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/23/2014] [Indexed: 12/31/2022] Open
Abstract
Here we describe a chemical biology approach for elucidating potential toxicity mechanisms for thrombosis-related side effects. This work takes advantage of a large chemical biology data set comprising the effects of known, well-characterized reference agents on the cell surface levels of tissue factor (TF) in a primary human endothelial cell-based model of vascular inflammation, the BioMAP® 3C system. In previous work with the Environmental Protection Agency (EPA) for the ToxCast™ program, aryl hydrocarbon receptor (AhR) agonists and estrogen receptor (ER) antagonists were found to share an usual activity, that of increasing TF levels in this system. Since human exposure to compounds in both chemical classes is associated with increased incidence of thrombosis-related side effects, we expanded this analysis with a large number of well-characterized reference compounds in order to better understand the underlying mechanisms. As a result, mechanisms for increasing (AhR, histamine H1 receptor, histone deacetylase or HDAC, hsp90, nuclear factor kappa B or NFκB, MEK, oncostatin M receptor, Jak kinase, and p38 MAPK) and decreasing (vacuolar ATPase or V-ATPase) and mTOR) TF expression levels were uncovered. These data identify the nutrient, lipid, bacterial, and hypoxia sensing functions of autophagy as potential key regulatory points controlling cell surface TF levels in endothelial cells and support the mechanistic hypothesis that these functions are associated with thrombosis-related side effects in vivo.
Collapse
|
45
|
Ni HM, Bhakta A, Wang S, Li Z, Manley S, Huang H, Copple B, Ding WX. Role of hypoxia inducing factor-1β in alcohol-induced autophagy, steatosis and liver injury in mice. PLoS One 2014; 9:e115849. [PMID: 25536043 PMCID: PMC4275262 DOI: 10.1371/journal.pone.0115849] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 11/27/2014] [Indexed: 12/12/2022] Open
Abstract
Chronic alcohol causes liver hypoxia and steatosis, which eventually develops into alcoholic liver disease (ALD). While it has been known that alcohol consumption activates hepatic hypoxia inducing factor-1α (HIF-1α), conflicting results regarding the role of HIF-1α in alcohol-induced liver injury and steatosis in mice have been reported. In the present study, we aimed to use hepatocyte-specific HIF-1β knockout mice to eliminate the possible compensatory effects of the single knockout of the 1α subunit of HIF to study the role of HIFs in ALD. C57BL/6 wild type mice were treated with acute ethanol to mimic human binge drinking. Matched wild-type and hepatocyte specific HIF-1β knockout mice were also subjected to a recently established Gao-binge alcohol model to mimic chronic plus binge conditions, which is quite common in human alcoholics. We found that acute alcohol treatment increased BNIP3 and BNIP3L/NIX expression in primary cultured hepatocytes and in mouse livers, suggesting that HIF may be activated in these models. We further found that hepatocyte-specific HIF-1β knockout mice developed less steatosis and liver injury following the Gao-binge model or acute ethanol treatment compared with their matched wild type mice. Mechanistically, protection against Gao-binge treatment-induced steatosis and liver injury was likely associated with increased FoxO3a activation and subsequent induction of autophagy in hepatocyte-specific HIF-1β knockout mice.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Amar Bhakta
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Shaogui Wang
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhenrui Li
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sharon Manley
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Heqing Huang
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Bryan Copple
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
46
|
Abstract
The pathogenic mechanism underlying alcoholic fatty liver (AFL) is not clear. Autophagy is a self-digestion process that is critical for the maintenance of cellular homeostasis and regulation of lipid metabolism. We investigated the role of autophagy and autophagic flux in hepatic injury induced by chronic ethanol feeding in mice. C57BL/6 mice were fed a Lieber-DeCarli ethanol diet (ED) to induce AFL or an isocaloric control diet for 6 weeks. Chloroquine (CQ, 10 mg/kg, intra-peritoneally [i.p.]) or rapamycin (Rapa, 5 mg/kg, i.p.) were administered during the last 2 weeks of the experimental period. Chronic ethanol feeding induced AFL with focal necrosis associated with increased levels of hepatic triglyceride. This phenomenon was aggravated by CQ, an inhibitor of autophagy, and attenuated by Rapa, an inducer of autophagy. Expression of microtubule-associated protein 1 light chain 3 (LC3)-II and sequestosome1/p62 significantly increased in the ED group. Moreover, accumulation of autophagosomes was observed by transmission electron microscopy in chronic ethanol-treated mice. Chronic ethanol consumption decreased protein expression of LC3 lipidation-related proteins Atg3 and Atg7, and the lysosomal proteins lysosome-associated membrane protein-2 and Rab7, and increased the protein expression of calpain 1 and phosphorylated mammalian target of rapamycin. Taken together, these findings suggest that chronic ethanol consumption leads to impairment of autophagic flux, which contributes to ethanol-induced liver injury.
Collapse
|
47
|
Donohue TM, Thomes PG. Ethanol-induced oxidant stress modulates hepatic autophagy and proteasome activity. Redox Biol 2014; 3:29-39. [PMID: 25462063 PMCID: PMC4297932 DOI: 10.1016/j.redox.2014.10.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 02/07/2023] Open
Abstract
In this review, we describe research findings on the effects of alcohol exposure on two major catabolic systems in liver cells: the ubiquitin-proteasome system (UPS) and autophagy. These hydrolytic systems are not unique to liver cells; they exist in all eukaryotic tissues and cells. However, because the liver is the principal site of ethanol metabolism, it sustains the greatest damage from heavy drinking. Thus, the focus of this review is to specifically describe how ethanol oxidation modulates the activities of the UPS and autophagy and the mechanisms by which these changes contribute to the pathogenesis of alcohol-induced liver injury. Here, we describe the history and the importance of cellular hydrolytic systems, followed by a description of each catabolic pathway and the differential modulation of each by ethanol exposure. Overall, the evidence for an involvement of these catabolic systems in the pathogenesis of alcoholic liver disease is quite strong. It underscores their importance, not only as effective means of cellular recycling and eventual energy generation, but also as essential components of cellular defense.
Collapse
Affiliation(s)
- Terrence M Donohue
- Research Service (151), VA-Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, College of Medicine, USA; Department of Biochemistry and Molecular Biology, College of Medicine, USA; Department of Pathology and Microbiology, College of Medicine, USA; The Center for Environmental Health and Toxicology, College of Public Health, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Paul G Thomes
- Research Service (151), VA-Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, College of Medicine, USA
| |
Collapse
|
48
|
Williams JA, Manley S, Ding WX. New advances in molecular mechanisms and emerging therapeutic targets in alcoholic liver diseases. World J Gastroenterol 2014; 20:12908-12933. [PMID: 25278688 PMCID: PMC4177473 DOI: 10.3748/wjg.v20.i36.12908] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/07/2014] [Accepted: 04/16/2014] [Indexed: 02/06/2023] Open
Abstract
Alcoholic liver disease is a major health problem in the United States and worldwide. Chronic alcohol consumption can cause steatosis, inflammation, fibrosis, cirrhosis and even liver cancer. Significant progress has been made to understand key events and molecular players for the onset and progression of alcoholic liver disease from both experimental and clinical alcohol studies. No successful treatments are currently available for treating alcoholic liver disease; therefore, development of novel pathophysiological-targeted therapies is urgently needed. This review summarizes the recent progress on animal models used to study alcoholic liver disease and the detrimental factors that contribute to alcoholic liver disease pathogenesis including miRNAs, S-adenosylmethionine, Zinc deficiency, cytosolic lipin-1β, IRF3-mediated apoptosis, RIP3-mediated necrosis and hepcidin. In addition, we summarize emerging adaptive protective effects induced by alcohol to attenuate alcohol-induced liver pathogenesis including FoxO3, IL-22, autophagy and nuclear lipin-1α.
Collapse
|
49
|
Farnesoid X receptor regulates forkhead Box O3a activation in ethanol-induced autophagy and hepatotoxicity. Redox Biol 2014; 2:991-1002. [PMID: 25460735 PMCID: PMC4215528 DOI: 10.1016/j.redox.2014.08.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 08/17/2014] [Accepted: 08/25/2014] [Indexed: 02/07/2023] Open
Abstract
Alcoholic liver disease encompasses a wide spectrum of pathogenesis including steatosis, fibrosis, cirrhosis, and alcoholic steatohepatitis. Autophagy is a lysosomal degradation process that degrades cellular proteins and damaged/excess organelles, and serves as a protective mechanism in response to various stresses. Acute alcohol treatment induces autophagy via FoxO3a-mediated autophagy gene expression and protects against alcohol-induced steatosis and liver injury in mice. Farnesoid X Receptor (FXR) is a nuclear receptor that regulates cellular bile acid homeostasis. In the present study, wild type and FXR knockout (KO) mice were treated with acute ethanol for 16 h. We found that ethanol treated-FXR KO mice had exacerbated hepatotoxicity and steatosis compared to wild type mice. Furthermore, we found that ethanol treatment had decreased expression of various essential autophagy genes and several other FoxO3 target genes in FXR KO mice compared with wild type mice. Mechanistically, we did not find a direct interaction between FXR and FoxO3. Ethanol-treated FXR KO mice had increased Akt activation, increased phosphorylation of FoxO3 resulting in decreased FoxO3a nuclear retention and DNA binding. Furthermore, ethanol treatment induced hepatic mitochondrial spheroid formation in FXR KO mice but not in wild type mice, which may serve as a compensatory alternative pathway to remove ethanol-induced damaged mitochondria in FXR KO mice. These results suggest that lack of FXR impaired FoxO3a-mediated autophagy and in turn exacerbated alcohol-induced liver injury. FXR knockout mice are more susceptible to acute ethanol-induced steatosis and liver injury due to defective hepatic autophagy. FXR knockout mice had decreased FoxO3a activation and reduced expression of autophagy related genes in the liver after acute ethanol treatment. FXR knockout mice had increased mitochondrial spheroid formation after acute ethanol treatment.
Collapse
|
50
|
Liver autophagy in anorexia nervosa and acute liver injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:701064. [PMID: 25250330 PMCID: PMC4163421 DOI: 10.1155/2014/701064] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
Autophagy, a lysosomal catabolic pathway for long-lived proteins and damaged organelles, is crucial for cell homeostasis, and survival under stressful conditions. During starvation, autophagy is induced in numerous organisms ranging from yeast to mammals, and promotes survival by supplying nutrients and energy. In the early neonatal period, when transplacental nutrients supply is interrupted, starvation-induced autophagy is crucial for neonates' survival. In adult animals, autophagy provides amino acids and participates in glucose metabolism following starvation. In patients with anorexia nervosa, autophagy appears initially protective, allowing cells to copes with nutrient deprivation. However, when starvation is critically prolonged and when body mass index reaches 13 kg/m(2) or lower, acute liver insufficiency occurs with features of autophagic cell death, which can be observed by electron microscopy analysis of liver biopsy samples. In acetaminophen overdose, a classic cause of severe liver injury, autophagy is induced as a protective mechanism. Pharmacological enhancement of autophagy protects against acetaminophen-induced necrosis. Autophagy is also activated as a rescue mechanism in response to Efavirenz-induced mitochondrial dysfunction. However, Efavirenz overdose blocks autophagy leading to liver cell death. In conclusion, in acute liver injury, autophagy appears as a protective mechanism that can be however blocked or overwhelmed.
Collapse
|