251
|
Volarevic V, Misirkic M, Vucicevic L, Paunovic V, Simovic Markovic B, Stojanovic M, Milovanovic M, Jakovljevic V, Micic D, Arsenijevic N, Trajkovic V, Lukic ML. Metformin aggravates immune-mediated liver injury in mice. Arch Toxicol 2014; 89:437-50. [PMID: 24770553 DOI: 10.1007/s00204-014-1263-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 04/15/2014] [Indexed: 12/16/2022]
Abstract
Hepatotoxicity of the antidiabetic drug metformin has been reported, but the underlying mechanisms remain unclear. We here investigated the effect of metformin in immune-mediated liver damage. While not hepatotoxic alone, metformin (200 mg/kg) aggravated concanavalin A (Con A, 12 mg/kg)-induced hepatitis, an experimental model of T cell-mediated liver injury, in both relatively resistant BALB/c and highly susceptible C57Bl/6 mice. Metformin + Con A-treated mice had elevated serum levels of pro-inflammatory cytokines TNF-α and IFN-γ, accompanied by a massive mononuclear cell infiltration in the liver. This was associated with the higher numbers of CD4(+) T cells producing TNF-α, IFN-γ and IL-17, CD4(+) T cells expressing chemokine receptor CXCR3 and activation marker CD27, CD4(+)CD62L(-)CCR7(-) and CD8(+)CD62L(-)CCR7(-) effector memory cells, IFN-γ producing NK cells, IL-4 and IL-17 producing NKT cells and IL-12 producing macrophages/dendritic cells. The percentage of CD4(+)CXCR3(+)Tbet(+)IL-10(+) and CD4(+)CD69(+)CD25(-) regulatory T cells was reduced. Metformin stimulated inducible nitric oxide synthase (iNOS) expression in the liver and spleen, and genetic deletion of iNOS attenuated the hepatotoxicity of metformin. Metformin increased the autophagic light chain 3 conversion and mRNA expression of important autophagy-inducing (beclin-1, Atg5 and GABARAP) and pro-apoptotic (p21, p27, Puma, Noxa, Bax, Bad, Bak1, Bim and Apaf1), but not anti-apoptotic molecules (Bcl-xL, survivin and XIAP), which correlated with the apoptotic caspase-3/PARP cleavage in the liver. The autophagy inhibitor chloroquine (20 mg/kg) prevented liver injury and apoptotic changes induced by metformin. Therefore, metformin aggravates immune-mediated hepatitis by promoting autophagy and activation of immune cells, affecting effector, as well as liver-specific regulatory T cells and iNOS expression.
Collapse
Affiliation(s)
- Vladislav Volarevic
- Centre for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozara Markovica Street, 34 000, Kragujevac, Serbia,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
252
|
Bao L, Chandra PK, Moroz K, Zhang X, Thung SN, Wu T, Dash S. Impaired autophagy response in human hepatocellular carcinoma. Exp Mol Pathol 2014; 96:149-154. [PMID: 24369267 PMCID: PMC4364514 DOI: 10.1016/j.yexmp.2013.12.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Autophagy is a cellular lysosomal degradation mechanism that has been implicated in chronic liver diseases and hepatocellular carcinoma (HCC). Association of autophagy defect with the development of human HCC has been shown in transgenic mouse model. AIM We performed this study to verify whether a defect in autophagy would play a role in human hepatocellular carcinoma (HCC). METHODS Archival tissue sections of 20 patients with HCC with or without hepatitis C virus (HCV) infection were studied. All slides were immunostained using monoclonal antibodies to p62 and glypican-3 with appropriate positive and negative controls. The expression of p62 and glycican-3 in the HCC and the surrounding non-tumor was semiquantitated. The cytoplasmic staining was graded as negative, weak or strong. RESULTS Positive p62 staining was found in 20 out of 20 (100%) HCCs and negative staining was observed in 20 out of 20 non-tumor areas and cirrhotic nodules. Positive glypican-3 staining was found in 70% of HCCs and negative staining was seen in all non-tumor areas. An autophagy defect leading to increased expression of p62 and glypican-3 was also seen in the HCC cell line (Huh-7.5), but not in the primary human hepatocytes. Activation of cellular autophagy in Huh-7.5 cells efficiently cleared p62 and glypican-3 expression and inhibition of autophagy induced the expression of p62 and glypican-3. CONCLUSIONS This study shows that p62 is increased in HCC compared to the surrounding non-tumorous liver tissue suggesting that human HCCs are autophagy defective. We provide further evidence that glypican-3 expression in HCC may also be related to defective autophagy. Our study indicates that p62 immunostain may represent a novel marker for HCC.
Collapse
Affiliation(s)
- Lili Bao
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Partha K Chandra
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Krzysztof Moroz
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Xuchen Zhang
- The Lillian and Henry M. Stratton-Hans Popper Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Swan N Thung
- The Lillian and Henry M. Stratton-Hans Popper Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, 1430 Tulane Avenue, New Orleans, LA 70112, USA.
| |
Collapse
|
253
|
Ju C, Roth RA. PKCs: pernicious kinase culprits in acetaminophen pathogenesis. Hepatology 2014; 59:1229-31. [PMID: 24677191 DOI: 10.1002/hep.26923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 09/06/2013] [Accepted: 10/31/2013] [Indexed: 12/07/2022]
Affiliation(s)
- Cynthia Ju
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO
| | | |
Collapse
|
254
|
Pi*Z heterozygous alpha-1 antitrypsin states accelerate parenchymal but not biliary cirrhosis. Eur J Gastroenterol Hepatol 2014; 26:412-7. [PMID: 24518491 DOI: 10.1097/meg.0000000000000061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The degree to which heterozygous forms of alpha-1 antitrypsin (A1AT), principally MZ, causes liver disease is uncertain. If heterozygosity is a relevant cofactor, over-representation in patients with end-stage liver disease would be predicted. We therefore assessed the prevalence and disease-related distribution of A1AT heterozygosity in the largest cohort to date for this purpose. METHODS We retrospectively analysed 1036 patients assessed for liver transplantation at our unit between 2003 and 2010. A1AT heterozygotes were identified on the basis of isoelectric focusing and/or histology, showing A1AT globule deposition consistent with an abnormal phenotype. RESULTS Z-allele frequency was the highest in patients with nonalcoholic steatohepatitis (NASH) cirrhosis (20.3%), followed by patients with 'other parenchymal' diseases (11.9%), alcohol-related liver disease (9.9%), autoimmune disease (8.6%), hepatitis C (6.1%), hepatitis B (3.0%) and biliary disease (1.9%). Compared with the heterozygote frequency in the general European population of 9.0%, the heterozygote frequency was significantly higher among patients with NASH cirrhosis (P≤0.0001) and lower in the biliary subgroup (P=0.004). The prevalence of MZ heterozygosity was significantly increased in cirrhosis because of both alcohol (9.9%) and NASH (17.3%) compared with the general European population (2.8%; P<0.0001). CONCLUSION Accumulation of misfolded A1AT aggregates appears to accelerate progression, in which the hepatocyte is the key injured cell. Heterozygous A1AT states worsen prognosis, particularly in NASH and alcohol-related cirrhosis, and should be identified at presentation. In cases in which genetic screening is not readily available, a low threshold for isoelectric focusing and routine specific histochemical staining of liver biopsy specimens are warranted to identify these patients.
Collapse
|
255
|
de Luxán-Delgado B, Caballero B, Potes Y, Rubio-González A, Rodríguez I, Gutiérrez-Rodríguez J, Solano JJ, Coto-Montes A. Melatonin administration decreases adipogenesis in the liver of ob/ob mice through autophagy modulation. J Pineal Res 2014; 56:126-33. [PMID: 24134701 DOI: 10.1111/jpi.12104] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/15/2013] [Indexed: 01/07/2023]
Abstract
Despite efforts to curb the incidence of obesity and its comorbidities, this condition remains the fifth leading cause of death worldwide. To identify ways to reduce this global effect, we investigated the actions of daily melatonin administration on oxidative stress parameters and autophagic processes as a possible treatment of obesity in ob/ob mice. The involvement of melatonin in many physiological functions, such as the regulation of seasonal body weight variation, glucose uptake, or adiposity, and the role of this indoleamine as an essential antioxidant, has become the focus of numerous anti-obesity studies. Here, we examined the oxidative status in the livers of obese melatonin-treated and untreated mice, observing a decrease in the oxidative stress levels through elevated catalase activity. ROS-mediated autophagy was downregulated in the liver of melatonin-treated animals and was accompanied by significant accumulation of p62. Autophagy is closely associated with adipogenesis; in this study, we report that melatonin-treated obese mice also showed reduced adiposity, as demonstrated by diminished body weight and reduced peroxisome proliferator-activated receptor gamma expression. Based on these factors, it is reasonable to assume that oxidative stress and autophagy play important roles in obesity, and therefore, melatonin could be an interesting target molecule for the development of a potential therapeutic agent to curb body weight.
Collapse
Affiliation(s)
- Beatriz de Luxán-Delgado
- Department of Morphology and Cellular Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | |
Collapse
|
256
|
Lin CW, Lo S, Perng DS, Wu DBC, Lee PH, Chang YF, Kuo PL, Yu ML, Yuan SSF, Hsieh YC. Complete activation of autophagic process attenuates liver injury and improves survival in septic mice. Shock 2014; 41:241-249. [PMID: 24365881 DOI: 10.1097/shk.0000000000000111] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The accumulation of autophagosomes in the terminal step of the autophagic process has recently emerged as a potentially maladaptive process in the septic heart and lung. However, the role of autophagy in the septic liver has not been ascertained. This study was investigated by first examining the entire sequence of the autophagic process in the liver of septic mice. Second, a novel pharmacotherapeutic approach was utilized to treat sepsis with autophagy enhancer/inhibitor. Sepsis was induced by cecal ligation and puncture (CLP). C57BL/6 mice received autophagy enhancer carbamazepine (CBZ), autophagy inhibitor 3-methyladenine (inhibition of autophagosomal formation), or chloroquine (impairment of autophagosomal clearance). We found that the whole autophagic process was activated at 4 h after CLP; however, it did not proceed to completion during the 4- to 24-h time period, as indicated by accumulated autophagosomes and decreased autophagic flux. Carbamazepine, which induced complete activation of the autophagic process, improved CLP survival. This protective effect was also associated with decreased cell death, inflammatory responses, and hepatic injury. However, disruption of autophagosomal clearance with chloroquine abolished the above protective effects in CBZ-treated CLP mice. 3-Methyladenine, which resulted in inhibition of the autophagosomal formation, did not show any above beneficial effects in CLP mice. Impaired autophagosome-lysome fusion resulting in incomplete activation of autophagy may contribute to sepsis-induced liver injury. Treatment with CBZ may serve a protective role in the septic liver, possibly through the effect of complete activation of autophagic process.
Collapse
Affiliation(s)
- Chih-Wen Lin
- *Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University; †Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University; ‡Department of Plastic and Reconstructive Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan; §Canniesburn Plastic Surgery Unit, Royal Infirmary, Glasgow, UK; ∥Jeffrey Cheah School of Medicine and Health Sciences, Monash University Sunway Campus, Malaysia; ¶Division of General Surgery, Department of Surgery, and **Department of Medical Research, E-Da Hospital, I-Shou University; ††School of Medicine, College of Medicine, Kaohsiung Medical University; ‡‡Hepatobiliary Division, Department of Internal Medicine, §§Translational Research Center and Cancer Center, and ∥∥Department of Obstetrics & Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
257
|
Puri P, Chandra A. Autophagy modulation as a potential therapeutic target for liver diseases. J Clin Exp Hepatol 2014; 4:51-9. [PMID: 25755534 PMCID: PMC4017203 DOI: 10.1016/j.jceh.2014.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/01/2014] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a critical intracellular pathway which maintains cellular function by lysosomal degradation of damaged proteins and organelles besides elimination of invading pathogens. Its primary function is to prevent cell death. Autophagy has diverse physiological functions namely; starvation adaptation, prevention of tumorigenesis, energy homeostasis, intracellular quality control and degradation of abnormal intracellular protein aggregates. Understanding the molecular mechanisms of autophagy has given key insights into the pathogenesis of various diseases like Non Alcoholic Steato-Hepatitis, Hepatitis B and C infections, Alpha-1 antitrypsin deficiency and hepatocellular carcinoma. Pharmacological modulation of autophagy may have a therapeutic potential in management of these liver diseases.
Collapse
Key Words
- AMPk, adenosine monophosphate-activated protein linase
- AT, antitrypsin
- ER, endoplasmic reticulum
- HBV, hepatitis B virus
- HCC, hepatocellular carcinoma
- HCV, hepatitis C virus
- HSC, hepatic stellate cells
- NASH, Non Alcoholic Steato-Hepatitis
- STEBPs, sterol regulatory element-binding proteins
- TG, triglyceride
- ULK1, Uncoordinated 51-like kinase 1
- autophagosome
- autophagy
- liver diseases
- mTORC1, mTOR complex 1
- mTORC2, mTOR complex 2
Collapse
Affiliation(s)
- Pankaj Puri
- Department of Gastroenterology, Army Hospital (Research and Referral), Dhaula Kuan, New Delhi 110010, India,Address for correspondence: Pankaj Puri, Department of Gastroenterology, Army Hospital (Research and Referral), Dhaula Kuan, New Delhi 110010, India.
| | - Alok Chandra
- Department of Gastroenterology, Command Hospital (EC), Alipore Road, Kolkata 700027, West Bengal, India
| |
Collapse
|
258
|
Abstract
Studies performed in the liver in the 1960s led to the identification of lysosomes and the discovery of autophagy, the process by which intracellular proteins and organelles are degraded in lysosomes. Early studies in hepatocytes also uncovered how nutritional status regulates autophagy and how various circulating hormones modulate the activity of this catabolic process in the liver. The intensive characterization of hepatic autophagy over the years has revealed that lysosome-mediated degradation is important not only for maintaining liver homeostasis in normal physiological conditions, but also for an adequate response of this organ to stressors such as proteotoxicity, metabolic dysregulation, infection and carcinogenesis. Autophagic malfunction has also been implicated in the pathogenesis of common liver diseases, suggesting that chemical manipulation of this process might hold potential therapeutic value. In this Review--intended as an introduction to the topic of hepatic autophagy for clinical scientists--we describe the different types of hepatic autophagy, their role in maintaining homeostasis in a healthy liver and the contribution of autophagic malfunction to liver disease.
Collapse
|
259
|
Wang Z, Han W, Sui X, Fang Y, Pan H. Autophagy: A novel therapeutic target for hepatocarcinoma (Review). Oncol Lett 2014; 7:1345-1351. [PMID: 24765136 PMCID: PMC3997714 DOI: 10.3892/ol.2014.1916] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/27/2014] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a highly conserved intracellular degradation process and plays an important role in hepatocarcinogenesis. Available data show that autophagy is involved in anti-hepatocarcinoma (HCC) therapies. Autophagy regulation involves a novel target for overcoming therapeutic resistance and sensitizing HCC to currently therapeutic methods. This is a systematic review on the interface of autophagy and the development of HCC and outlining the role of autophagy in current anti-HCC approaches. Understanding the significance of autophagy in anti-HCC therapy may offer a novel therapeutic target for improving anti-cancer efficacy and prolong survival for HCC patients.
Collapse
Affiliation(s)
- Zhanggui Wang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Xinbing Sui
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
260
|
Lan SH, Wu SY, Zuchini R, Lin XZ, Su IJ, Tsai TF, Lin YJ, Wu CT, Liu HS. Autophagy suppresses tumorigenesis of hepatitis B virus-associated hepatocellular carcinoma through degradation of microRNA-224. Hepatology 2014; 59:505-17. [PMID: 23913306 PMCID: PMC4298796 DOI: 10.1002/hep.26659] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 07/24/2013] [Indexed: 12/12/2022]
Abstract
UNLABELLED In hepatocellular carcinoma (HCC), dysregulated expression of microRNA-224 (miR-224) and impaired autophagy have been reported separately. However, the relationship between them has not been explored. In this study we determined that autophagy is down-regulated and inversely correlated with miR-224 expression in hepatitis B virus (HBV)-associated HCC patient specimens. These results were confirmed in liver tumors of HBV X gene transgenic mice. Furthermore, miR-224 was preferentially recruited and degraded during autophagic progression demonstrated by real-time polymerase chain reaction and miRNA in situ hybridization electron microscopy after extraction of autophagosomes. Our in vitro study demonstrated that miR-224 played an oncogenic role in hepatoma cell migration and tumor formation through silencing its target gene Smad4. In HCC patients, the expression of low-Atg5, high-miR-224, and low-Smad4 showed significant correlation with HBV infection and a poor overall survival rate. Autophagy-mediated miR-224 degradation and liver tumor suppression were further confirmed by the autophagy inducer amiodarone and miR-224 antagonist using an orthotopic SD rat model. CONCLUSION A noncanonical pathway links autophagy, miR-224, Smad4, and HBV-associated HCC. These findings open a new avenue for the treatment of HCC.
Collapse
Affiliation(s)
- Sheng-Hui Lan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Shan-Ying Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Roberto Zuchini
- Graduate Institute of Clinical Medicine, College of Medicine, National Cheng Kung UniversityTainan, Taiwan,Department of Internal Medicine, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Xi-Zhang Lin
- Department of Internal Medicine, College of Medicine, National Cheng Kung UniversityTainan, Taiwan
| | - Ih-Jen Su
- National Health Research InstitutesDivision of Clinical Research, Tainan, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming UniversityTaipei, Taiwan
| | - Yen-Ju Lin
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research InstituteHsinchu, Taiwan
| | - Cheng-Tao Wu
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research InstituteHsinchu, Taiwan
| | - Hsiao-Sheng Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung UniversityTainan, Taiwan,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung UniversityTainan, Taiwan,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung UniversityTainan, Taiwan,Address reprint requests to: Hsiao-Sheng Liu, Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, #1 University road, Tainan, Taiwan; Tel: +1886-6-2353535, ext. 5630; Fax: 1886-6-2082705; E-mail:
| |
Collapse
|
261
|
Shen M, Lu J, Dai W, Wang F, Xu L, Chen K, He L, Cheng P, Zhang Y, Wang C, Wu D, Yang J, Zhu R, Zhang H, Zhou Y, Guo C. Ethyl pyruvate ameliorates hepatic ischemia-reperfusion injury by inhibiting intrinsic pathway of apoptosis and autophagy. Mediators Inflamm 2013; 2013:461536. [PMID: 24453420 PMCID: PMC3886226 DOI: 10.1155/2013/461536] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/26/2013] [Accepted: 11/28/2013] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Hepatic ischemia-reperfusion (I/R) injury is a pivotal clinical problem occurring in many clinical conditions such as transplantation, trauma, and hepatic failure after hemorrhagic shock. Apoptosis and autophagy have been shown to contribute to cell death in hepatic I/R injury. Ethyl pyruvate, a stable and simple lipophilic ester, has been shown to have anti-inflammatory properties. In this study, the purpose is to explore both the effect of ethyl pyruvate on hepatic I/R injury and regulation of intrinsic pathway of apoptosis and autophagy. METHODS Three doses of ethyl pyruvate (20 mg/kg, 40 mg/kg, and 80 mg/kg) were administered 1 h before a model of segmental (70%) hepatic warm ischemia was established in Balb/c mice. All serum and liver tissues were obtained at three different time points (4 h, 8 h, and 16 h). RESULTS Alanine aminotransferase (ALT), aspartate aminotransferase (AST), and pathological features were significantly ameliorated by ethyl pyruvate (80 mg/kg). The expression of Bcl-2, Bax, Beclin-1, and LC3, which play an important role in the regulation of intrinsic pathway of apoptosis and autophagy, was also obviously decreased by ethyl pyruvate (80 mg/kg). Furthermore, ethyl pyruvate inhibited the HMGB1/TLR4/ NF-κb axis and the release of cytokines (TNF-α and IL-6). CONCLUSION Our results showed that ethyl pyruvate might attenuate to hepatic I/R injury by inhibiting intrinsic pathway of apoptosis and autophagy, mediated partly through downregulation of HMGB1/TLR4/ NF-κb axis and the competitive interaction with Beclin-1 of HMGB1.
Collapse
Affiliation(s)
- Miao Shen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Fan Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ling Xu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Lei He
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Ping Cheng
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yan Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chengfen Wang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Dong Wu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Jing Yang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Rong Zhu
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Huawei Zhang
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Yinqun Zhou
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, The Tenth People's Hospital of Tongji University, Shanghai 200072, China
| |
Collapse
|
262
|
Huang YY, Gusdon AM, Qu S. Cross-talk between the thyroid and liver: A new target for nonalcoholic fatty liver disease treatment. World J Gastroenterol 2013; 19:8238-8246. [PMID: 24363514 PMCID: PMC3857446 DOI: 10.3748/wjg.v19.i45.8238] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 10/04/2013] [Accepted: 10/18/2013] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been recognized as the most common liver metabolic disease, and it is also a burgeoning health problem that affects one-third of adults and is associated with obesity and insulin resistance now. Thyroid hormone (TH) and its receptors play a fundamental role in lipid metabolism and lipid accumulation in the liver. It is found that thyroid receptor and its isoforms exhibit tissue-specific expression with a variety of functions. TRβ1 is predominantly expressed in the brain and adipose tissue and TRβ2 is the major isoform in the liver, kidney and fat. They have different functions and play important roles in lipid metabolism. Recently, there are many studies on the treatment of NAFLD with TH and its analogues. We review here that thyroid hormone and TR are a potential target for pharmacologic treatments. Lipid metabolism and lipid accumulation can be regulated and reversed by TH and its analogues.
Collapse
|
263
|
Excess iron modulates endoplasmic reticulum stress-associated pathways in a mouse model of alcohol and high-fat diet-induced liver injury. J Transl Med 2013; 93:1295-312. [PMID: 24126888 DOI: 10.1038/labinvest.2013.121] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 12/12/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is an important pathogenic mechanism for alcoholic (ALD) and nonalcoholic fatty liver disease (NAFLD). Iron overload is an important cofactor for liver injury in ALD and NAFLD, but its role in ER stress and associated stress signaling pathways is unclear. To investigate this, we developed a murine model of combined liver injury by co-feeding the mildly iron overloaded, the hemochromatosis gene-null (Hfe(-/)) mouse ad libitum with ethanol and a high-fat diet (HFD) for 8 weeks. This co-feeding led to profound steatohepatitis, significant fibrosis, and increased apoptosis in the Hfe(-/-) mice as compared with wild-type (WT) controls. Iron overload also led to induction of unfolded protein response (XBP1 splicing, activation of IRE-1α and PERK, as well as sequestration of GRP78) and ER stress (increased CHOP protein expression) following HFD and ethanol. This is associated with a muted autophagic response including reduced LC3-I expression and impaired conjugation to LC3-II, reduced beclin-1 protein, and failure of induction of autophagy-related proteins (Atg) 3, 5, 7, and 12. As a result of the impaired autophagy, levels of the sequestosome protein p62 were most elevated in the Hfe(-/-) group co-fed ethanol and HFD. Iron overload reduces the activation of adenosine monophosphate protein kinase associated with ethanol and HFD feeding. We conclude that iron toxicity may modulate hepatic stress signaling pathways by impairing adaptive cellular compensatory mechanisms in alcohol- and obesity-induced liver injury.
Collapse
|
264
|
Yang J, Carra S, Zhu WG, Kampinga HH. The regulation of the autophagic network and its implications for human disease. Int J Biol Sci 2013; 9:1121-33. [PMID: 24339733 PMCID: PMC3858585 DOI: 10.7150/ijbs.6666] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 06/28/2013] [Indexed: 12/19/2022] Open
Abstract
Autophagy has attracted a lot of attention in recent years. More and more proteins and signaling pathways have been discovered that somehow feed into the autophagy regulatory pathways. Regulation of autophagy is complex and condition-specific, and in several diseases, autophagic fluxes are changed. Here, we review the most well-established concepts in this field as well as the reported signaling pathways or components which steer the autophagy machinery. Furthermore, we will highlight how autophagic fluxes are changed in various diseases either as cause for or as response to deal with an altered cellular homeostasis and how modulation of autophagy might be used as potential therapy for such diseases.
Collapse
Affiliation(s)
- Jing Yang
- 1. Department of Cell Biology; University Medical Center Groningen, University of Groningen; Groningen, The Netherlands. ; 2. Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education); Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing 100191, China
| | | | | | | |
Collapse
|
265
|
Wu D, Cederbaum AI. Inhibition of autophagy promotes CYP2E1-dependent toxicity in HepG2 cells via elevated oxidative stress, mitochondria dysfunction and activation of p38 and JNK MAPK. Redox Biol 2013; 1:552-65. [PMID: 24273738 PMCID: PMC3836279 DOI: 10.1016/j.redox.2013.10.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 10/27/2013] [Indexed: 02/07/2023] Open
Abstract
Autophagy has been shown to be protective against drug and alcohol-induced liver injury. CYP2E1 plays a role in the toxicity of ethanol, carcinogens and certain drugs. Inhibition of autophagy increased ethanol-toxicity and accumulation of fat in wild type and CYP2E1 knockin mice but not in CYP2E1 knockout mice as well as in HepG2 cells expressing CYP2E1 (E47 cells) but not HepG2 cells lacking CYP2E1 (C34 cells). The goal of the current study was to evaluate whether modulation of autophagy can affect CYP2E1-dependent cytotoxicity in the E47 cells. The agents used to promote CYP2E1 -dependent toxicity were a polyunsaturated fatty acid, arachidonic acid (AA), buthionine sulfoximine (BSO), which depletes GSH, and CCl4, which is metabolized to the CCl3 radical. These three agents produced a decrease in E47 cell viability which was enhanced upon inhibition of autophagy by 3-methyladenine (3-MA) or Atg 7 siRNA. Toxicity was lowered by rapamycin which increased autophagy and was much lower to the C34 cells which do not express CYP2E1. Toxicity was mainly necrotic and was associated with an increase in reactive oxygen production and oxidative stress; 3-MA increased while rapamycin blunted the oxidative stress. The enhanced toxicity and ROS formation produced when autophagy was inhibited was prevented by the antioxidant N-Acetyl cysteine. AA, BSO and CCl4 produced mitochondrial dysfunction, lowered cellular ATP levels and elevated mitochondrial production of ROS. This mitochondrial dysfunction was enhanced by inhibition of autophagy with 3-MA but decreased when autophagy was increased by rapamycin. The mitogen activated protein kinases p38 MAPK and JNK were activated by AA especially when autophagy was inhibited and chemical inhibitors of p38 MAPK and JNK lowered the elevated toxicity of AA produced by 3-MA. These results show that autophagy was protective against the toxicity produced by several agents known to be activated by CYP2E1. Since CYP2E1 plays an important role in the toxicity of ethanol, drugs and carcinogens and is activated under various pathophysiological conditions such as diabetes, NASH and obesity, attempts to stimulate autophagy may be beneficial in preventing/lowering CYP2E1/ethanol liver injury.
Collapse
Key Words
- 3-MA, 3-methyadenine
- AA, arachidonic acid
- Autophagy
- BSO, L-buthionine sulfoximine
- C34 cells, HepG2 cells which do not express CYP2E1
- CCl4, carbon tetrachloride
- CYP2E1
- CYP2E1, cytochrome P4502E1, E47 cells, HepG2 cells which express CYP2E1
- Cox IV, cytochrome oxidase subunit 4
- Cytotoxicity
- DCFDA, 2′-7′-dichlorofluorescin-diacetate
- GSH, reduced glutathione
- JNK
- MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium-bromide
- Mitochondria dysfunction
- NAC, N-acetyl-cysteine
- P38 MAPK
- ROS
- ROS, reactive oxygen species
- Rap, rapamycin
- SOD, superoxide dismutase
- TBARs, thiobarbituric acid-reactive substances
Collapse
Affiliation(s)
| | - Arthur I. Cederbaum
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, Box 1603, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
266
|
Liu K, Lou J, Wen T, Yin J, Xu B, Ding W, Wang A, Liu D, Zhang C, Chen D, Li N. Depending on the stage of hepatosteatosis, p53 causes apoptosis primarily through either DRAM-induced autophagy or BAX. Liver Int 2013; 33:1566-74. [PMID: 23875779 PMCID: PMC4283711 DOI: 10.1111/liv.12238] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 05/23/2013] [Accepted: 05/28/2013] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Apoptosis mediated by p53 plays a pathological role in the progression of hepatosteatosis. It is noteworthy that p53 can promote the expression of damage-regulated autophagy modulator (DRAM), an inducer of autophagy-mediated apoptosis. However, the relationship between p53-mediated apoptosis and autophagy in hepatosteatosis remains elusive. This study aimed to examine how p53 orchestrates autophagy and apoptosis to affect hepatosteatosis. METHODS HepG2 cells were treated with oleic acid (OA) for 24 h to induce hepatosteatosis. Mice were fed a high-fat diet for 20 or 40 weeks to induce hepatosteatosis. RESULTS OA induced a dose-dependent increase in steatosis severity and apoptosis. OA also induced autophagy, which was a critical inducer of apoptosis in mild steatosis induced by 400 μM OA, but not in the more severe steatosis induced by 800 and 1200 μM OA. p53 inhibition by siRNA mostly blocked OA-induced apoptosis and autophagy. Moreover, OA-induced autophagy was DRAM-dependent and primarily occurred in the mitochondria (mitophagy), where DRAM was localized. In severe steatosis induced by 1200 μM OA, apoptosis was mainly dependent on p53-induced expression of BAX, which was also localized to the mitochondria. Our in vivo study showed that p53 expression increased in both mild and severe hepatosteatosis. Increased DRAM expression and autophagy were identified in mild hepatosteatosis, whereas greater BAX expression was observed in severe hepatosteatosis. CONCLUSIONS p53 may induce apoptosis via different mechanisms. DRAM-mediated mitophagy is a primary apoptotic inducer in mild hepatosteatosis, whereas p53-induced BAX expression mainly induces apoptosis in severe hepatosteatosis.
Collapse
Affiliation(s)
- Kai Liu
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China,
* Both authors contributed equally to this work
| | - Jinli Lou
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China,
* Both authors contributed equally to this work
| | - Tao Wen
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Jiming Yin
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Bin Xu
- Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Wei Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical UniversityBeijing, China
| | - Anna Wang
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Daojie Liu
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Chao Zhang
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Dexi Chen
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China
| | - Ning Li
- Infectious Disease Medical Center, Beijing YouAn Hospital, Capital Medical UniversityBeijing, China,Division of HCC Research, Beijing Institute of HepatologyBeijing, China,Correspondence, Ning Li, MD., Beijing You'an Hospital, Capital Medical University, 8 Xi Tou Tiao, You An Men Wai, Feng Tai, 100069 Beijing, China, Tel: +86 10 6329 2337, Fax: +86 10 6305 7109, e-mail:
| |
Collapse
|
267
|
Lake AD, Novak P, Hardwick RN, Flores-Keown B, Zhao F, Klimecki WT, Cherrington NJ. The adaptive endoplasmic reticulum stress response to lipotoxicity in progressive human nonalcoholic fatty liver disease. Toxicol Sci 2013; 137:26-35. [PMID: 24097666 DOI: 10.1093/toxsci/kft230] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) may progress from simple steatosis to severe, nonalcoholic steatohepatitis (NASH) in 7%-14% of the U.S. population through a second "hit" in the form of increased oxidative stress and inflammation. Endoplasmic reticulum (ER) stress signaling and the unfolded protein response (UPR) are triggered when high levels of lipids and misfolded proteins alter ER homeostasis creating a lipotoxic environment within NAFLD livers. The objective of this study was to determine the coordinate regulation of ER stress-associated genes in the progressive stages of human NAFLD. Human liver samples categorized as normal, steatosis, NASH (Fatty), and NASH (Not Fatty) were analyzed by individual Affymetrix GeneChip Human 1.0 ST microarrays, immunoblots, and immunohistochemistry. A gene set enrichment analysis was performed on autophagy, apoptosis, lipogenesis, and ER stress/UPR gene categories. An enrichment of downregulated genes in the ER stress-associated lipogenesis and ER stress/UPR gene categories was observed in NASH. Conversely, an enrichment of upregulated ER stress-associated genes for autophagy and apoptosis gene categories was observed in NASH. Protein expression of the adaptive liver response protein STC2 and the transcription factor X-box binding protein 1 spliced (XBP-1s) were significantly elevated among NASH samples, whereas other downstream ER stress proteins including CHOP, ATF4, and phosphorylated JNK and eIF2α were not significantly changed in disease progression. Increased nuclear accumulation of total XBP-1 protein was observed in steatosis and NASH livers. The findings reveal the presence of a coordinated, adaptive transcriptional response to hepatic ER stress in human NAFLD.
Collapse
|
268
|
Fouraschen SMG, de Ruiter PE, Kwekkeboom J, de Bruin RWF, Kazemier G, Metselaar HJ, Tilanus HW, van der Laan LJW, de Jonge J. mTOR signaling in liver regeneration: Rapamycin combined with growth factor treatment. World J Transplant 2013; 3:36-47. [PMID: 24255881 PMCID: PMC3832859 DOI: 10.5500/wjt.v3.i3.36] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/28/2013] [Accepted: 06/19/2013] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the effects of mammalian target of rapamycin (mTOR) inhibition on liver regeneration and autophagy in a surgical resection model.
METHODS: C57BL/6 mice were subjected to a 70% partial hepatectomy (PH) and treated intraperitoneally every 24 h with a combination of the mTOR inhibitor rapamycin (2.5 mg/kg per day) and the steroid dexamethasone (2.0 mg/kg per day) in phosphate buffered saline (PBS) or with PBS alone as vehicle control. In the immunosuppressant group, part of the group was treated subcutaneously 4 h prior to and 24 h after PH with a combination of human recombinant interleukin 6 (IL-6; 500 μg/kg per day) and hepatocyte growth factor (HGF; 100 μg/kg per day) in PBS. Animals were sacrificed 2, 3 or 5 d after PH and liver tissue and blood were collected for further analysis. Immunohistochemical staining for 5-Bromo-2’-deoxyuridine (BrdU) was used to quantify hepatocyte proliferation. Western blotting was used to detect hepatic microtubule-associated protein 1 light chain 3 (LC3)-II protein expression as a marker for autophagy. Hepatic gene expression levels of proliferation-, inflammation- and angiogenesis-related genes were examined by real-time reverse transcription-polymerase chain reaction and serum bilirubin and transaminase levels were analyzed at the clinical chemical core facility of the Erasmus MC-University Medical Center.
RESULTS: mTOR inhibition significantly suppressed regeneration, shown by decreased hepatocyte proliferation (2% vs 12% BrdU positive hepatocyte nuclei at day 2, P < 0.01; 0.8% vs 1.4% at day 5, P = 0.02) and liver weight reconstitution (63% vs 76% of initial total liver weight at day 3, P = 0.04), and furthermore increased serum transaminase levels (aspartate aminotransferase 641 U/L vs 185 U/L at day 2, P = 0.02). Expression of the autophagy marker LC3-II, which was reduced during normal liver regeneration, increased after mTOR inhibition (46% increase at day 2, P = 0.04). Hepatic gene expression showed an increased inflammation-related response [tumor necrosis factor (TNF)-α 3.2-fold upregulation at day 2, P = 0.03; IL-1Ra 6.0-fold upregulation at day 2 and 42.3-fold upregulation at day 5, P < 0.01] and a reduced expression of cell cycle progression and angiogenesis-related factors (HGF 40% reduction at day 2; vascular endothelial growth factor receptor 2 50% reduction at days 2 and 5; angiopoietin 1 60% reduction at day 2, all P≤ 0.01). Treatment with the regeneration stimulating cytokine IL-6 and growth factor HGF could overcome the inhibitory effect on liver weight (75% of initial total liver weight at day 3, P = 0.02 vs immunosuppression alone and P = 0.90 vs controls) and partially reversed gene expression changes caused by rapamycin (TNF-α and IL-1Ra levels at day 2 were restored to control levels). However, no significant changes in hepatocyte proliferation, serum injury markers or autophagy were found.
CONCLUSION: mTOR inhibition severely impairs liver regeneration and increases autophagy after PH. These effects are partly reversed by stimulation of the IL-6 and HGF pathways.
Collapse
|
269
|
Primary hepatocytes and their cultures in liver apoptosis research. Arch Toxicol 2013; 88:199-212. [PMID: 24013573 DOI: 10.1007/s00204-013-1123-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/29/2013] [Indexed: 01/18/2023]
Abstract
Apoptosis not only plays a key role in physiological demise of defunct hepatocytes, but is also associated with a plethora of acute and chronic liver diseases as well as with hepatotoxicity. The present paper focuses on the modelling of this mode of programmed cell death in primary hepatocyte cultures. Particular attention is paid to the activation of spontaneous apoptosis during the isolation of hepatocytes from the liver, its progressive manifestation upon the subsequent establishment of cell cultures and simultaneously to strategies to counteract this deleterious process. In addition, currently applied approaches to experimentally induce controlled apoptosis in this in vitro setting for mechanistic research purposes and thereby its detection using relevant biomarkers are reviewed.
Collapse
|
270
|
|
271
|
Li Q, Li L, Wang F, Chen J, Zhao Y, Wang P, Nilius B, Liu D, Zhu Z. Dietary capsaicin prevents nonalcoholic fatty liver disease through transient receptor potential vanilloid 1-mediated peroxisome proliferator-activated receptor δ activation. Pflugers Arch 2013; 465:1303-16. [PMID: 23605066 DOI: 10.1007/s00424-013-1274-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 03/19/2013] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by hepatic lipid deposition and coincides often with cardiometabolic diseases. Several dietary factors attenuate NAFLD. Here, we report beneficial effects of chronic dietary capsaicin intake on NAFLD which is mediated by the transient receptor potential vanilloid 1 (TRPV1) activation. The results showed that TRPV1 activation by capsaicin reduced free fatty acids (FFAs) induced the intracellular lipid droplets in HepG2 cells and prevented fatty liver in vivo. Chronic dietary capsaicin promoted lipolysis by increasing hepatic phosphorylated hormone-sensitive lipase (phospho-HSL), carnitine palmitoyltransferase 1 (CPT1), and peroxisome proliferator-activated receptor δ (PPARδ) in wild-type (WT) mice. This effect was absent in TRPV1(-/-) mice. Dietary capsaicin did not affect lipogenesis, as indicated by the detection of hepatic fatty acid synthase (FAS), sterol regulatory element-binding protein-1 (SREBP-1), PPARα, and liver X receptor (LXR) in mice. Importantly, TRPV1 causes PPARδ activation which significantly increased the expression of autophagy-related proteins, such as light chain 3 (LC3)II, Beclin1, Atg5, and Atg7 in HepG2 cells. In the in vivo study, TRPV1 activation by dietary capsaicin enhanced hepatic PPARδ and autophagy-related proteins and reduced hepatic enzymes and inflammatory factor in WT but not TRPV1(-/-) mice. TRPV1 activation by dietary capsaicin prevents NAFLD through PPARδ-dependent autophagy enhancement in mice. Dietary capsaicin may represent a beneficial intervention in populations at high risk for NAFLD.
Collapse
Affiliation(s)
- Qiang Li
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing 400042, China
| | | | | | | | | | | | | | | | | |
Collapse
|
272
|
Convergent mechanisms for dysregulation of mitochondrial quality control in metabolic disease: implications for mitochondrial therapeutics. Biochem Soc Trans 2013; 41:127-33. [PMID: 23356271 DOI: 10.1042/bst20120231] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mitochondrial dysfunction is associated with a broad range of pathologies including diabetes, ethanol toxicity, metabolic syndrome and cardiac failure. It is now becoming clear that maintaining mitochondrial quality through a balance between biogenesis, reserve capacity and mitophagy is critical in determining the response to metabolic or xenobiotic stress. In diseases associated with metabolic stress, such as Type II diabetes and non-alcoholic and alcoholic steatosis, the mitochondria are subjected to multiple 'hits' such as hypoxia and oxidative and nitrative stress, which can overwhelm the mitochondrial quality control pathways. In addition, the underlying mitochondrial genetics that evolved to accommodate high-energy demand, low-calorie supply environments may now be maladapted to modern lifestyles (low-energy demand, high-calorie environments). The pro-oxidant and pro-inflammatory environment of a sedentary western lifestyle has been associated with modified redox cell signalling pathways such as steatosis, hypoxic signalling, inflammation and fibrosis. These data suggest that loss of mitochondrial quality control is intimately associated with the aberrant activation of redox cell signalling pathways under pathological conditions. In the present short review, we discuss evidence from alcoholic liver disease supporting this concept, the insights obtained from experimental models and the application of bioenergetic-based therapeutics in the context of maintaining mitochondrial quality.
Collapse
|
273
|
Zaouali MA, Boncompagni E, Reiter RJ, Bejaoui M, Freitas I, Pantazi E, Folch-Puy E, Abdennebi HB, Garcia-Gil FA, Roselló-Catafau J. AMPK involvement in endoplasmic reticulum stress and autophagy modulation after fatty liver graft preservation: a role for melatonin and trimetazidine cocktail. J Pineal Res 2013; 55:65-78. [PMID: 23551302 DOI: 10.1111/jpi.12051] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/22/2013] [Indexed: 12/11/2022]
Abstract
Ischemia/reperfusion injury (IRI) associated with liver transplantation plays an important role in the induction of graft injury. Prolonged cold storage remains a risk factor for liver graft outcome, especially when steatosis is present. Steatotic livers exhibit exacerbated endoplasmic reticulum (ER) stress that occurs in response to cold IRI. In addition, a defective liver autophagy correlates well with liver damage. Here, we evaluated the combined effect of melatonin and trimetazidine as additives to IGL-1 solution in the modulation of ER stress and autophagy in steatotic liver grafts through activation of AMPK. Steatotic livers were preserved for 24 hr (4°C) in UW or IGL-1 solutions with or without MEL + TMZ and subjected to 2-hr reperfusion (37°C). We assessed hepatic injury (ALT and AST) and function (bile production). We evaluated ER stress (GRP78, PERK, and CHOP) and autophagy (beclin-1, ATG7, LC3B, and P62). Steatotic livers preserved in IGL-1 + MEL + TMZ showed lower injury and better function as compared to those preserved in IGL-1 alone. IGL-1 + MEL + TMZ induced a significant decrease in GRP78, pPERK, and CHOP activation after reperfusion. This was consistent with a major activation of autophagic parameters (beclin-1, ATG7, and LC3B) and AMPK phosphorylation. The inhibition of AMPK induced an increase in ER stress and a significant reduction in autophagy. These data confirm the close relationship between AMPK activation and ER stress and autophagy after cold IRI. The addition of melatonin and TMZ to IGL-1 solution improved steatotic liver graft preservation through AMPK activation, which reduces ER stress and increases autophagy.
Collapse
Affiliation(s)
- Mohamed Amine Zaouali
- Experimental Hepatic Ischemia-Reperfusion Unit, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas, IDIBAPS-Ciberehd, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
274
|
Codogno P, Meijer AJ. Autophagy in the liver. J Hepatol 2013; 59:389-91. [PMID: 23669287 DOI: 10.1016/j.jhep.2013.02.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Patrice Codogno
- INSERM U845, Necker Medical School, University Paris-Descartes, 75015 Paris, France.
| | | |
Collapse
|
275
|
Brain JG, Robertson H, Thompson E, Humphreys EH, Gardner A, Booth TA, Jones DEJ, Afford SC, von Zglinicki T, Burt AD, Kirby JA. Biliary epithelial senescence and plasticity in acute cellular rejection. Am J Transplant 2013; 13:1688-702. [PMID: 23750746 PMCID: PMC3746108 DOI: 10.1111/ajt.12271] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 03/26/2013] [Indexed: 01/25/2023]
Abstract
Biliary epithelial cells (BEC) are important targets in some liver diseases, including acute allograft rejection. Although some injured BEC die, many can survive in function compromised states of senescence or phenotypic de-differentiation. This study was performed to examine changes in the phenotype of BEC during acute liver allograft rejection and the mechanism driving these changes. Liver allograft sections showed a positive correlation (p < 0.0013) between increasing T cell mediated acute rejection and the number of BEC expressing the senescence marker p21(WAF1/Cip) or the mesenchymal marker S100A4. This was modeled in vitro by examination of primary or immortalized BEC after acute oxidative stress. During the first 48 h, the expression of p21(WAF1/Cip) was increased transiently before returning to baseline. After this time BEC showed increased expression of mesenchymal proteins with a decrease in epithelial markers. Analysis of TGF-β expression at mRNA and protein levels also showed a rapid increase in TGF-β2 (p < 0.006) following oxidative stress. The epithelial de-differentiation observed in vitro was abrogated by pharmacological blockade of the ALK-5 component of the TGF-β receptor. These data suggest that stress induced production of TGF-β2 by BEC can modify liver allograft function by enhancing the de-differentiation of local epithelial cells.
Collapse
Affiliation(s)
- J G Brain
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - H Robertson
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - E Thompson
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - E H Humphreys
- Centre for Liver Research, School of Infection and Immunity University of BirminghamBirmingham, UK,NIHR BRU Queen Elizabeth Hospital BirminghamUK
| | - A Gardner
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - T A Booth
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - D E J Jones
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK
| | - S C Afford
- Centre for Liver Research, School of Infection and Immunity University of BirminghamBirmingham, UK,NIHR BRU Queen Elizabeth Hospital BirminghamUK
| | - T von Zglinicki
- Institute for Ageing and Health, Newcastle UniversityNewcastle upon Tyne, UK
| | - A D Burt
- Clinical Deanery, Newcastle UniversityNewcastle upon Tyne, UK
| | - J A Kirby
- Institute of Cellular Medicine, Newcastle UniversityNewcastle upon Tyne, UK,*Corresponding author: John A. Kirby,
| |
Collapse
|
276
|
Knockdown of TIGAR by RNA interference induces apoptosis and autophagy in HepG2 hepatocellular carcinoma cells. Biochem Biophys Res Commun 2013; 437:300-6. [DOI: 10.1016/j.bbrc.2013.06.072] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 06/20/2013] [Indexed: 11/18/2022]
|
277
|
Liu A, Fang H, Dahmen U, Dirsch O. Chronic lithium treatment protects against liver ischemia/reperfusion injury in rats. Liver Transpl 2013; 19:762-72. [PMID: 23696274 DOI: 10.1002/lt.23666] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/06/2013] [Indexed: 12/30/2022]
Abstract
Lithium has long been widely used in the treatment of bipolar mood disorders. Recent studies have demonstrated that lithium is able to decrease ischemia/reperfusion (I/R) injury in the brain, kidneys, and heart. Because lithium may act on a number of stress and survival pathways, it is of great interest to explore this compound also in the setting of liver I/R injury. In this study, we aimed to evaluate the effects of lithium in a model of liver I/R injury in rats. Chronic treatment with lithium (2 mmol/kg for 3 days before ischemia) decreased I/R injury, whereas acute treatment with a single dose of lithium (2 mmol/kg 1 hour before ischemia) did not confer any protection in a partial hepatic I/R model. Furthermore, rats subjected to chronic lithium treatment had a significantly better survival rate (60%) than saline-treated rats (27%) in a total hepatic I/R survival model. Chronic lithium treatment protected against liver I/R injury, as indicated by lower serum aminotransferase levels, fewer I/R-associated histopathological changes, lower hepatic inflammatory cytokine levels, less neutrophil infiltration, and lower hepatic high-mobility group box expression and serum levels. The mechanism of action of lithium appears to involve its ability to inhibit glycogen synthase kinase 3β activation, modulate mitogen-activated protein kinase activation, inhibit hepatic apoptosis, and induce autophagy. On the basis of these data, we conclude that lithium treatment may be a simple and applicable preconditioning intervention for protecting against liver I/R injury.
Collapse
Affiliation(s)
- Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | |
Collapse
|
278
|
Fang H, Liu A, Dahmen U, Dirsch O. Dual role of chloroquine in liver ischemia reperfusion injury: reduction of liver damage in early phase, but aggravation in late phase. Cell Death Dis 2013; 4:e694. [PMID: 23807223 PMCID: PMC3702304 DOI: 10.1038/cddis.2013.225] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The anti-malaria drug chloroquine is well known as autophagy inhibitor. Chloroquine has also been used as anti-inflammatory drugs to treat inflammatory diseases. We hypothesized that chloroquine could have a dual effect in liver ischemia/reperfusion (I/R) injury: chloroquine on the one hand could protect the liver against I/R injury via inhibition of inflammatory response, but on the other hand could aggravate liver I/R injury through inhibition of autophagy. Rats (n=6 per group) were pre-treated with chloroquine (60 mg/kg, i.p.) 1 h before warm ischemia, and they were continuously subjected to a daily chloroquine injection for up to 2 days. Rats were killed 0.5, 6, 24 and 48 h after reperfusion. At the early phase (i.e., 0–6 h after reperfusion), chloroquine treatment ameliorated liver I/R injury, as indicated by lower serum aminotransferase levels, lower hepatic inflammatory cytokines and fewer histopathologic changes. In contrast, chloroquine worsened liver injury at the late phase of reperfusion (i.e., 24–48 h after reperfusion). The mechanism of protective action of chloroquine appeared to involve its ability to modulate mitogen-activated protein kinase activation, reduce high-mobility group box 1 release and inflammatory cytokines production, whereas chloroquine worsened liver injury via inhibition of autophagy and induction of hepatic apoptosis at the late phase. In conclusion, chloroquine prevents ischemic liver damage at the early phase, but aggravates liver damage at the late phase in liver I/R injury. This dual role of chloroquine should be considered when using chloroquine as an inhibitor of inflammation or autophagy in I/R injury.
Collapse
Affiliation(s)
- H Fang
- Department of Pathophysiology, Anhui Medical University, Hefei 230032, China
| | | | | | | |
Collapse
|
279
|
Nepal S, Park PH. Activation of autophagy by globular adiponectin attenuates ethanol-induced apoptosis in HepG2 cells: involvement of AMPK/FoxO3A axis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:2111-25. [PMID: 23688633 DOI: 10.1016/j.bbamcr.2013.05.013] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/09/2013] [Accepted: 05/10/2013] [Indexed: 02/07/2023]
Abstract
Hepatocellular apoptosis is an important pathological entity of alcoholic liver disease. Previously, we have shown that globular adiponectin (gAcrp) protects liver cells from ethanol-induced apoptosis by modulating an array of signaling pathways. In the present study, we investigated the role of autophagy induction by gAcrp in the suppression of ethanol-induced apoptosis and its potential mechanism(s) in liver cells. Here, we demonstrated that gAcrp significantly restores ethanol-induced suppression of autophagy-related genes, including Beclin-1 and microtubule-associated protein light chain (LC3B) both in primary rat hepatocytes and human hepatoma cell line (HepG2). Globular adiponectin also restored autophagosome formation suppressed by ethanol treatment in HepG2. Furthermore, inhibition of gAcrp-induced autophagic process by knock-down of LC3B prevented protection from ethanol-induced apoptosis. In particular, the autophagic process induced by gAcrp was involved in the suppression of ethanol-induced activation of caspase-8 and expression of Bax. Moreover, knock-down of AMPK by small interfering RNA (siRNA) blocked gAcrp-induced expression of genes related to autophagy, which in turn prevented protection from ethanol-induced apoptosis, suggesting that AMPK plays an important role in the induction of autophagy and protection of liver cells by gAcrp. Finally, we also showed that gAcrp treatment induces translocation of the forkhead box O member protein, FoxO3A, into the nucleus, which may play a role in the induction of autophagy-related genes. Taken together, our data demonstrated that gAcrp protects liver cells from ethanol-induced apoptosis via induction of autophagy. Further, the AMPK-FoxO3A axis plays a cardinal role in gAcrp-induced autophagy and subsequent inhibition of ethanol-induced apoptosis.
Collapse
Affiliation(s)
- Saroj Nepal
- College of Pharmacy, Yeungnam University, Gyeongsangbuk-do, Republic of Korea
| | | |
Collapse
|
280
|
Sid B, Verrax J, Calderon PB. Role of AMPK activation in oxidative cell damage: Implications for alcohol-induced liver disease. Biochem Pharmacol 2013; 86:200-9. [PMID: 23688501 DOI: 10.1016/j.bcp.2013.05.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 05/03/2013] [Accepted: 05/08/2013] [Indexed: 02/08/2023]
Abstract
Chronic alcohol consumption is a well-known risk factor for liver disease. Progression of alcohol-induced liver disease (ALD) is a multifactorial process that involves a number of genetic, nutritional and environmental factors. Experimental and clinical studies increasingly show that oxidative damage induced by ethanol contributes in many ways to the pathogenesis of alcohol hepatoxicity. Oxidative stress appears to activate AMP-activated protein kinase (AMPK) signaling system, which has emerged in recent years as a kinase that controls the redox-state and mitochondrial function. This review focuses on the most recent insights concerning the activation of AMPK by reactive oxygen species (ROS), and describes recent evidences supporting the hypothesis that AMPK signaling pathways play an important role in promoting cell viability under conditions of oxidative stress, such as during alcohol exposure. We suggest that AMPK activation by ROS can promote cell survival by inducing autophagy, mitochondrial biogenesis and expression of genes involved in antioxidant defense. Hence, increased intracellular concentrations of ROS may represent a general mechanism for enhancement of AMPK-mediated cellular adaptation, including maintenance of redox homeostasis. On the other hand, AMPK inhibition in the liver by ethanol appears to play a key role in the development of steatosis induced by chronic alcohol consumption. Although more studies are needed to assess the functions of AMPK during oxidative stress, AMPK may be a possible therapeutic target in the particular case of alcohol-induced liver disease.
Collapse
Affiliation(s)
- Brice Sid
- Université Catholique de Louvain, Louvain Drug Research Institute, Toxicology and Cancer Biology Research Group GTOX, Brussels, Belgium
| | | | | |
Collapse
|
281
|
Ketogenic essential amino acids replacement diet ameliorated hepatosteatosis with altering autophagy-associated molecules. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1605-12. [PMID: 23669346 DOI: 10.1016/j.bbadis.2013.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 02/06/2023]
Abstract
Ketogenic amino acid (KAA) replacement diet has been shown to cure hepatic steatosis, a serious liver disease associated with diverse metabolic defects. In this study, we investigated the effects of KAA replacement diet on nutrition sensing signaling pathway and analyzed whether induction of hepatic autophagy was involved. Mice are fed with high fat diet (HFD) or KAA replacement in high-fat diet (30% fat in food; HFD)-fed (HFD(KAAR)) and sacrificed at 8, 12, 16 weeks after initiation of experimental food. Hepatic autophagy was analyzed in protein expression of several autophagy-associated molecules and in light chain-3 green fluorescent protein (LC-3 GFP) transgenic mice. HFD(KAAR) showed increased AMP-activated protein kinase (AMPK) phosphorylation and enhanced liver kinase B1 (LKB1) expression compared to control HFD-fed mice. The KAA-HFD-induced activation of AMPK was associated with an increased protein expression of sirtuin 1 (Sirt1), decreased forkhead box protein O3a (Foxo3a) level, and suppression of mammalian target of rapamycin (mTOR) phosphorylation compared with the HFD-fed mice. The intervention study revealed that a KAA-replacement diet also ameliorated all the established metabolic and autophagy defects in the HFD-fed mice, suggesting that a KAA-replacement diet can be used therapeutically in established diseases. These results indicate that KAA replacement in food could be a novel strategy to combat hepatic steatosis and metabolic abnormalities likely involvement of an induction of autophagy.
Collapse
|
282
|
Lin CW, Zhang H, Li M, Xiong X, Chen X, Chen X, Dong XX, Yin XM. Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in mice. J Hepatol 2013; 58:993-9. [PMID: 23339953 PMCID: PMC3634371 DOI: 10.1016/j.jhep.2013.01.011] [Citation(s) in RCA: 349] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 12/19/2012] [Accepted: 01/03/2013] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Pharmacological approaches can potentially improve fatty liver condition in alcoholic and non-alcoholic fatty liver diseases. The salutary effects of reducing lipid synthesis or promoting lipid oxidation have been well reported, but the benefits of increasing lipid degradation have yet to be well explored. Macroautophagy is a cellular degradation process that can remove subcellular organelles including lipid droplets. We thus investigated whether pharmacological modulation of macroautophagy could be an effective approach to alleviate fatty liver condition and liver injury. METHODS C57BL/6 mice were given ethanol via intraperitoneal injection (acute) or by a 4-week oral feeding regime (chronic), or high fat diet for 12 weeks. An autophagy enhancer, carbamazepine or rapamycin, or an autophagy inhibitor, chloroquine, was given before sacrifice. Activation of autophagy, level of hepatic steatosis, and blood levels of triglycerides, liver enzyme, glucose and insulin were measured. RESULTS In both acute and chronic ethanol condition, macroautophagy was activated. Carbamazepine, as well as rapamycin, enhanced ethanol-induced macroautophagy in hepatocytes in vitro and in vivo. Hepatic steatosis and liver injury were exacerbated by chloroquine, but alleviated by carbamazepine. The protective effects of carbamazepine and rapamycin in reducing steatosis and in improving insulin sensitivity were also demonstrated in high fat diet-induced non-alcoholic fatty liver condition. CONCLUSIONS These findings indicate that pharmacological modulation of macroautophagy in the liver can be an effective strategy for reducing fatty liver condition and liver injury.
Collapse
Affiliation(s)
- Chih-Wen Lin
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hao Zhang
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Min Li
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Xiwen Xiong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Xi Chen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Xiaoyun Chen
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Xiaocheng X. Dong
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Xiao-Ming Yin
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
283
|
Lin CW, Zhang H, Li M, Xiong X, Chen X, Chen X, Dong XC, Yin XM. Pharmacological promotion of autophagy alleviates steatosis and injury in alcoholic and non-alcoholic fatty liver conditions in mice. J Hepatol 2013. [PMID: 23339953 DOI: 10.1124/10.1016/j.jhep.2013.01.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND & AIMS Pharmacological approaches can potentially improve fatty liver condition in alcoholic and non-alcoholic fatty liver diseases. The salutary effects of reducing lipid synthesis or promoting lipid oxidation have been well reported, but the benefits of increasing lipid degradation have yet to be well explored. Macroautophagy is a cellular degradation process that can remove subcellular organelles including lipid droplets. We thus investigated whether pharmacological modulation of macroautophagy could be an effective approach to alleviate fatty liver condition and liver injury. METHODS C57BL/6 mice were given ethanol via intraperitoneal injection (acute) or by a 4-week oral feeding regime (chronic), or high fat diet for 12 weeks. An autophagy enhancer, carbamazepine or rapamycin, or an autophagy inhibitor, chloroquine, was given before sacrifice. Activation of autophagy, level of hepatic steatosis, and blood levels of triglycerides, liver enzyme, glucose and insulin were measured. RESULTS In both acute and chronic ethanol condition, macroautophagy was activated. Carbamazepine, as well as rapamycin, enhanced ethanol-induced macroautophagy in hepatocytes in vitro and in vivo. Hepatic steatosis and liver injury were exacerbated by chloroquine, but alleviated by carbamazepine. The protective effects of carbamazepine and rapamycin in reducing steatosis and in improving insulin sensitivity were also demonstrated in high fat diet-induced non-alcoholic fatty liver condition. CONCLUSIONS These findings indicate that pharmacological modulation of macroautophagy in the liver can be an effective strategy for reducing fatty liver condition and liver injury.
Collapse
Affiliation(s)
- Chih-Wen Lin
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | | | | | | | | | | | | |
Collapse
|
284
|
Cui J, Gong Z, Shen HM. The role of autophagy in liver cancer: molecular mechanisms and potential therapeutic targets. Biochim Biophys Acta Rev Cancer 2013; 1836:15-26. [PMID: 23428608 DOI: 10.1016/j.bbcan.2013.02.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 02/08/2013] [Accepted: 02/08/2013] [Indexed: 02/07/2023]
Abstract
Autophagy is an evolutionarily conserved pathway for degradation of cytoplasmic proteins and organelles via lysosome. Proteins coded by the autophagy-related genes (Atgs) are the core molecular machinery in control of autophagy. Among the various biological functions of autophagy identified so far, the link between autophagy and cancer is probably among the most extensively studied and is often viewed as controversial. Autophagy might exert a dual role in cancer development: autophagy can serve as an anti-tumor mechanism, as defective autophagy (e.g., heterozygous knockdown Beclin 1 and Atg7 in mice) promotes the malignant transformation and spontaneous tumors. On the other hand, autophagy functions as a protective or survival mechanism in cancer cells against cellular stress (e.g., nutrient deprivation, hypoxia and DNA damage) and hence promotes tumorigenesis and causes resistance to therapeutic agents. Liver cancer is one of the common cancers with well-established etiological factors including hepatitis virus infection and environmental carcinogens such as aflatoxin and alcohol exposure. In recent years, the involvement of autophagy in liver cancer has been increasingly studied. Here, we aim to provide a systematic review on the close cross-talks between autophagy and liver cancer, and summarize the current status in development of novel liver cancer therapeutic approaches by targeting autophagy. It is believed that understanding the molecular mechanisms underlying the autophagy modulation and liver cancer development may provoke the translational studies that ultimately lead to new therapeutic strategies for liver cancer.
Collapse
Affiliation(s)
- Jianzhou Cui
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | |
Collapse
|
285
|
Alterations in the redox state and liver damage: hints from the EASL Basic School of Hepatology. J Hepatol 2013; 58:365-74. [PMID: 23023012 DOI: 10.1016/j.jhep.2012.09.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/27/2012] [Accepted: 09/19/2012] [Indexed: 12/19/2022]
Abstract
The importance of a correct balance between oxidative and reductive events has been shown to have a paramount effect on cell function for quite a long time. However, in spite of this body of rapidly growing evidence, the implication of the alteration of the redox state in human disease has been so far much less appreciated. Liver diseases make no exception. Although not fully comprehensive, this article reports what discussed during an EASL Basic School held in 2012 in Trieste, Italy, where the effect of the alteration of the redox state was addressed in different experimental and human models. This translational approach resulted in further stressing the concept that this topic should be expanded in the future not only to better understand how oxidative stress may be linked to a liver damage but also, perhaps more important, how this may be the target for better, more focused treatments. In parallel, understanding how alteration of the redox balance may be associated with liver damage may help define sensitive and ideally early biomarkers of the disorder.
Collapse
|
286
|
Ghosh S, Kaplan KJ, Schrum LW, Bonkovsky HL. Cytoskeletal proteins: shaping progression of hepatitis C virus-induced liver disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 302:279-319. [PMID: 23351713 DOI: 10.1016/b978-0-12-407699-0.00005-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatitis C virus (HCV) infection, which results in chronic hepatitis C (CHC) in most patients (70-85%), is a major cause of liver disease and remains a major therapeutic challenge. The mechanisms determining liver damage and the key factors that lead to a high rate of CHC remain imperfectly understood. The precise role of cytoskeletal (CS) proteins in HCV infection remains to be determined. Some studies including our recent study have demonstrated that changes occur in the expression of CS proteins in HCV-infected hepatocytes. A variety of host proteins interact with HCV proteins. Association between CS and HCV proteins may have implications in future design of CS protein-targeted therapy for the treatment for HCV infection. This chapter will focus on the interaction between host CS and viral proteins to signify the importance of this event in HCV entry, replication and transportation.
Collapse
Affiliation(s)
- Sriparna Ghosh
- Liver-Biliary-Pancreatic Center, Carolinas Medical Center, and School of Medicine, University of North Carolina, Carolinas Medical Center, Charlotte, NC, USA.
| | | | | | | |
Collapse
|
287
|
Su J, Zhou L, Kong X, Yang X, Xiang X, Zhang Y, Li X, Sun L. Endoplasmic reticulum is at the crossroads of autophagy, inflammation, and apoptosis signaling pathways and participates in the pathogenesis of diabetes mellitus. J Diabetes Res 2013; 2013:193461. [PMID: 23762873 PMCID: PMC3673337 DOI: 10.1155/2013/193461] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 05/08/2013] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease, and its incidence is growing worldwide. The endoplasmic reticulum (ER) is a central component of cellular functions and is involved in protein folding and trafficking, lipid synthesis, and maintenance of calcium homeostasis. The ER is also a sensor of both intra- and extracellular stress and thus participates in monitoring and maintaining cellular homeostasis. Therefore, the ER is one site of interaction between environmental signals and a cell's biological function. The ER is tightly linked to autophagy, inflammation, and apoptosis, and recent evidence suggests that these processes are related to the pathogenesis of DM and its complications. Thus, the ER has been considered an intersection integrating multiple stress responses and playing an important role in metabolism-related diseases including DM. Here, we review the relationship between the ER and autophagy, inflammation, and apoptosis in DM to better understand the molecular mechanisms of this disease.
Collapse
Affiliation(s)
- Jing Su
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Lei Zhou
- Department of Pathology, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xiaoxia Kong
- Institute of Hypoxia Research, School of Basic Medical Sciences, Wenzhou Medical College, Wenzhou, Zhejiang 325035, China
| | - Xiaochun Yang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Xiyan Xiang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Yu Zhang
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Xiaoning Li
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Liankun Sun
- Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
- *Liankun Sun:
| |
Collapse
|
288
|
CYP2E1-catalyzed alcohol metabolism: role of oxidant generation in interferon signaling, antigen presentation and autophagy. Subcell Biochem 2013; 67:177-97. [PMID: 23400922 DOI: 10.1007/978-94-007-5881-0_6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cytochrome P450 2E1 (CYP2E1) is one of two major enzymes that catalyze ethanol oxidation in the liver. CYP2E1 is also unique because it is inducible, as its hepatic content rises after continuous (chronic) ethanol administration, thereby accelerating the rate of ethanol metabolism and affording greater tolerance to heavy alcohol consumption. However, the broad substrate specificity of CYP2E1 and its capacity to generate free radicals from alcohol and other hepatotoxins, places CYP2E1 as a central focus of not only liver toxicity, but also as an enzyme that regulates cytokine signaling, antigen presentation, and macromolecular degradation, all of which are crucial to liver cell function and viability. Here, we describe our own and other published work relevant to the importance of CYP2E1-catalyzed ethanol oxidation and how this catalysis affects the aforementioned cellular processes to produce liver injury.
Collapse
|
289
|
Hill BG, Benavides GA, Lancaster JR, Ballinger S, Dell’Italia L, Zhang J, Darley-Usmar VM. Integration of cellular bioenergetics with mitochondrial quality control and autophagy. Biol Chem 2012; 393:1485-1512. [PMID: 23092819 PMCID: PMC3594552 DOI: 10.1515/hsz-2012-0198] [Citation(s) in RCA: 371] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2012] [Accepted: 06/22/2012] [Indexed: 02/06/2023]
Abstract
Bioenergetic dysfunction is emerging as a cornerstone for establishing a framework for understanding the pathophysiology of cardiovascular disease, diabetes,cancer and neurodegeneration. Recent advances in cellular bioenergetics have shown that many cells maintain a substantial bioenergetic reserve capacity, which is a prospective index of ‘ healthy ’ mitochondrial populations.The bioenergetics of the cell are likely regulated by energy requirements and substrate availability. Additionally,the overall quality of the mitochondrial population and the relative abundance of mitochondria in cells and tissues also impinge on overall bioenergetic capacity and resistance to stress. Because mitochondria are susceptible to damage mediated by reactive oxygen/nitrogen and lipid species, maintaining a ‘ healthy ’ population of mitochondria through quality control mechanisms appears to be essential for cell survival under conditions of pathological stress. Accumulating evidence suggest that mitophagy is particularly important for preventing amplification of initial oxidative insults, which otherwise would further impair the respiratory chain or promote mutations in mitochondrial DNA (mtDNA). The processes underlying the regulation of mitophagy depend on several factors, including the integrity of mtDNA, electron transport chain activity, and the interaction and regulation of the autophagic machinery. The integration and interpretation of cellular bioenergetics in the context of mitochondrial quality control and genetics is the theme of this review.
Collapse
Affiliation(s)
- Bradford G. Hill
- Diabetes and Obesity Center, Institute of Molecular Cardiology, and Department of Medicine, University of Louisville, Louisville, KY
- Departments of Biochemistry and Molecular Biology and Physiology and Biophysics, University of Louisville, Louisville, KY
| | - Gloria A. Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jack R. Lancaster
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Physiology and Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Scott Ballinger
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lou Dell’Italia
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Jianhua Zhang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Victor M. Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Medicine, Center for Heart Failure Research, University of Alabama at Birmingham, Birmingham, AL 35294
- Department of Environmental Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
290
|
Sivertsson L, Synnergren J, Jensen J, Björquist P, Ingelman-Sundberg M. Hepatic differentiation and maturation of human embryonic stem cells cultured in a perfused three-dimensional bioreactor. Stem Cells Dev 2012; 22:581-94. [PMID: 22970843 DOI: 10.1089/scd.2012.0202] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Drug-induced liver injury is a serious and frequently occurring adverse drug reaction in the clinics and is hard to predict during preclinical studies. Today, primary hepatocytes are the most frequently used cell model for drug discovery and prediction of toxicity. However, their use is marred by high donor variability regarding drug metabolism and toxicity, and instable expression levels of liver-specific genes such as cytochromes P450. An in vitro model system based on human embryonic stem cells (hESC), with their unique properties of pluripotency and self-renewal, has potential to provide a stable and unlimited supply of human hepatocytes. Much effort has been made to direct hESC toward the hepatic lineage, mostly using 2-dimensional (2D) cultures. Although the results are encouraging, these cells lack important functionality. Here, we investigate if hepatic differentiation of hESC can be improved by using a 3-dimensional (3D) bioreactor system. Human ESCs were differentiated toward the hepatic lineage using the same cells in either the 3D or 2D system. A global transcriptional analysis identified important differences between the 2 differentiation regimes, and we identified 10 pathways, highly related to liver functions, which were significantly upregulated in cells differentiated in the bioreactor compared to 2D control cultures. The enhanced hepatic differentiation observed in the bioreactor system was also supported by immunocytochemistry. Taken together, our results suggest that hepatic differentiation of hESC is improved when using this 3D bioreactor technology as compared to 2D culture systems.
Collapse
Affiliation(s)
- Louise Sivertsson
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
291
|
PI3K-PKB hyperactivation augments human plasmacytoid dendritic cell development and function. Blood 2012; 120:4982-91. [PMID: 23091295 DOI: 10.1182/blood-2012-02-413229] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are considered potential tools or targets for immunotherapy. However, current knowledge concerning methodologies to manipulate their development or function remains limited. Here, we investigated the role of the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (PKB)-mammalian target of rapamycin (mTOR) axis in human pDC development, survival, and function. In vitro pDC generation from human cord blood-derived CD34(+) hematopoietic progenitors was reduced by pharmacologic inhibition of PI3K, PKB, or mTOR activity, and peripheral blood pDCs required PI3K-PKB-mTOR signaling to survive. Accordingly, activity of this pathway in circulating pDCs correlated with their abundance in peripheral blood. Importantly, introduction of constitutively active PKB or pharmacologic inhibition of negative regulator phosphatase and tensin homolog (PTEN) resulted in increased pDC numbers in vitro and in vivo. Furthermore, MHC class II and costimulatory molecule expression, and production of IFN-α and TNF-α, were augmented, which could be explained by enhanced IRF7 and NF-κB activation. Finally, the numerically and functionally impaired pDCs of chronic hepatitis B patients demonstrated reduced PI3K-PKB-mTOR activity. In conclusion, intact PI3K-PKB-mTOR signaling regulates development, survival, and function of human pDCs, and pDC development and functionality can be promoted by PI3K-PKB hyperactivation. Manipulation of this pathway or its downstream targets could be used to improve the generation and function of pDCs to augment immunity.
Collapse
|
292
|
Wu D, Wang X, Zhou R, Yang L, Cederbaum AI. Alcohol steatosis and cytotoxicity: the role of cytochrome P4502E1 and autophagy. Free Radic Biol Med 2012; 53:1346-57. [PMID: 22819980 PMCID: PMC3436962 DOI: 10.1016/j.freeradbiomed.2012.07.005] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 07/06/2012] [Accepted: 07/06/2012] [Indexed: 02/07/2023]
Abstract
The goal of the current study was to evaluate whether CYP2E1 plays a role in binge-ethanol induced steatosis and if autophagy impacts CYP2E1-mediated hepatotoxicity, oxidative stress and fatty liver formation produced by ethanol. Wild type (WT), CYP2E1 knockin (KI) and CYP2E1 knockout (KO) mice were gavaged with 3g/kg body wt ethanol twice a day for four days. This treatment caused fatty liver, elevation of CYP2E1 and oxidative stress in WT and KI mice but not KO mice. Autophagy was impaired in ethanol-treated KI mice compared to KO mice as reflected by a decline in the LC3-II/LC3-I ratio and lower total LC-3 and Beclin-1 levels coupled to increases in P62, pAKT/AKT and mTOR. Inhibition of macroautophagy by administration of 3-methyladenine enhanced the binge ethanol hepatotoxicity, steatosis and oxidant stress in CYP2E1 KI, but not CYP2E1 KO mice. Stimulation of autophagy by rapamycin blunted the elevated steatosis produced by binge ethanol. Treatment of HepG2 E47 cells which express CYP2E1 with 100mM ethanol for 8 days increased fat accumulation and oxidant stress but decreased autophagy. Ethanol had no effect on these reactions in HepG2 C34 cells which do not express CYP2E1. Inhibition of autophagy elevated ethanol toxicity, lipid accumulation and oxidant stress in the E47, but not C34 cells. The antioxidant N-acetylcysteine, and CYP2E1 inhibitor chlormethiazole blunted these effects of ethanol. These results indicate that CYP2E1 plays an important role in binge ethanol-induced fatty liver. We propose that CYP2E1-derived reactive oxygen species inhibit autophagy, which subsequently causes accumulation of lipid droplets. Inhibition of autophagy promotes binge ethanol induced hepatotoxicity, steatosis and oxidant stress via CYP2E1.
Collapse
Affiliation(s)
- Defeng Wu
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029
| | - Xiaodong Wang
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029
| | - Richard Zhou
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029
| | - Lili Yang
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029
| | - Arthur I. Cederbaum
- Department of Pharmacology & Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
293
|
Lavallard VJ, Meijer AJ, Codogno P, Gual P. Autophagy, signaling and obesity. Pharmacol Res 2012; 66:513-25. [PMID: 22982482 DOI: 10.1016/j.phrs.2012.09.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 12/28/2022]
Abstract
Autophagy is a cellular pathway crucial for development, differentiation, survival and homeostasis. Autophagy can provide protection against aging and a number of pathologies such as cancer, neurodegeneration, cardiac disease and infection. Recent studies have reported new functions of autophagy in the regulation of cellular processes such as lipid metabolism and insulin sensitivity. Important links between the regulation of autophagy and obesity including food intake, adipose tissue development, β cell function, insulin sensitivity and hepatic steatosis exist. This review will provide insight into the current understanding of autophagy, its regulation, and its role in the complications associated with obesity.
Collapse
Affiliation(s)
- Vanessa J Lavallard
- INSERM, U1065, Equipe 8 «Complications hépatiques de l'obésité», Nice, France
| | | | | | | |
Collapse
|
294
|
Hokari A, Ishikawa T, Tajiri H, Matsuda T, Ishii O, Matsumoto N, Okuse C, Takahashi H, Kurihara T, Kawahara KI, Maruyama I, Zeniya M. Efficacy of MK615 for the treatment of patients with liver disorders. World J Gastroenterol 2012; 18:4118-26. [PMID: 22919243 PMCID: PMC3422791 DOI: 10.3748/wjg.v18.i31.4118] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 05/10/2012] [Accepted: 05/26/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the hepatoprotective effect of MK615, a Japanese apricot extract, in an animal model, and its clinical therapeutic effect.
METHODS: Wistar rats were administered physiological saline (4 mL/kg) or MK615 solution (4 mL/kg) for 7 d. On the sixth d, acute hepatic injury was induced by administering a single intraperitoneal injection (ip) of D-galactosamine hydrochloride (D-GalN) (600 mg/kg). Plasma levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were determined, and liver tissues were used for histopathological analysis. Fifty-eight patients with liver disorders [hepatitis C (n = 40), non-alcoholic fatty liver disease (n = 15), and autoimmune liver disease (n = 3)] were orally administered commercially available Misatol ME-containing MK615 (13 g/d) daily for 12 wk. Blood and urine were sampled immediately before and 6 wk, 12 wk, and 16 wk after the start of intake to measure various biochemical parameters. The percentage change in ALT and AST levels after 12 wk from the pre-intake baseline served as a primary endpoint.
RESULTS: D-GalN effectively induced acute hepatic injury in the rats. At 48 h after the ip injection of D-GalN, the plasma levels of ALT (475.6 ± 191.5 IU/L vs 225.3 ± 194.2 IU/L, P < 0.05) and AST (1253.9 ± 223.4 IU/L vs 621.9 ± 478.2 IU/L, P < 0.05) in the MK615 group were significantly lower than the control group. Scattered single cell necrosis, loss of hepatocytes, and extensive inflammatory cell infiltration were observed in hepatic tissue samples collected from the control group. However, these findings were less pronounced in the group receiving MK615. At the end of the clinical study, serum ALT and AST levels were significantly decreased compared with pre-intake baseline levels from 103.5 ± 58.8 IU/L to 71.8 ± 39.3 IU/L (P < 0.05) and from 93.5 ± 55.6 IU/L to 65.5 ± 34.8 IU/L (P < 0.05), respectively. A reduction of ≥ 30% from the pre-study baseline ALT level was observed in 26 (45%) of the 58 patients, while 25 (43%) patients exhibited similar AST level reductions. The chronic hepatitis C group exhibited significant ALT and AST level reductions from 93.4 ± 51.1 IU/L to 64.6 ± 35.1 IU/L (P < 0.05) and from 94.2 ± 55.5 IU/L to 67.2 ± 35.6 IU/L (P < 0.05), respectively. A reduction of ≥ 30% from the pre-study baseline ALT level was observed in 20 (50%) of the 40 patients. ALT levels in both the combined ursodeoxycholic acid (UDCA) treatment and the UDCA uncombined groups were significantly lower after Misatol ME administration. MK615 protected hepatocytes from D-GalN-induced cytotoxicity in rats. Misatol ME decreased elevated ALT and AST levels in patients with liver disorders.
CONCLUSION: These results suggest that MK615 and Misatol ME are promising hepatoprotective agents for patients with liver disorders.
Collapse
|
295
|
Abstract
Hypoxic hepatitis (HH), an acute liver injury also known as 'ischaemic hepatitis' or 'shock liver', is frequently observed in intensive care units. HH is heralded by a massive but transient rise in serum aminotransferase activities caused by anoxic necrosis of centrilobular liver cells. Cardiac failure, respiratory failure and toxic-septic shock are the main underlying conditions accounting for more than 90% of cases, but HH may also occur in other circumstances. Until recently, liver ischaemia, i.e. a drop in hepatic blood flow, was considered the leading, and even the sole, hemodynamic mechanism responsible for HH, and it was generally held that a shock state was required. In reality, other hemodynamic mechanisms of hypoxia, such as passive congestion of the liver, arterial hypoxaemia and dysoxia, play an important role while a shock state is observed in only 50% of cases. Accordingly, 'ischaemic hepatitis' and 'shock liver' are misnomers. Therapy of HH depends primarily on the nature of the underlying condition. The prognosis is poor, with more than half of patients dying during the hospital stay.
Collapse
Affiliation(s)
- Jean Henrion
- Service d'Hépato-Gastroentérologie, Hôpital de Jolimont, Haine-Saint-Paul, Belgium.
| |
Collapse
|
296
|
Ni HM, Williams JA, Yang H, Shi YH, Fan J, Ding WX. Targeting autophagy for the treatment of liver diseases. Pharmacol Res 2012; 66:463-74. [PMID: 22871337 DOI: 10.1016/j.phrs.2012.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 12/19/2022]
Abstract
Autophagy is a lysosomal degradation pathway that can degrade bulk cytoplasm and superfluous or damaged organelles, such as mitochondria, to maintain cellular homeostasis. It is now known that dysregulation of autophagy can cause pathogenesis of numerous human diseases. Here, we discuss the critical roles that autophagy plays in the pathogenesis of liver diseases such as non-alcoholic and alcoholic fatty liver, drug-induced liver injury, protein aggregate-related liver diseases, viral hepatitis, fibrosis, aging and liver cancer. In particular, we discuss the emerging therapeutic potential by pharmacological modulation of autophagy for these liver diseases.
Collapse
Affiliation(s)
- Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, KS 66160, United States
| | | | | | | | | | | |
Collapse
|
297
|
Evankovich J, Zhang R, Cardinal JS, Zhang L, Chen J, Huang H, Beer-Stolz D, Billiar TR, Rosengart MR, Tsung A. Calcium/calmodulin-dependent protein kinase IV limits organ damage in hepatic ischemia-reperfusion injury through induction of autophagy. Am J Physiol Gastrointest Liver Physiol 2012; 303:G189-98. [PMID: 22575222 PMCID: PMC3404570 DOI: 10.1152/ajpgi.00051.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sterile inflammatory insults, such as ischemia-reperfusion (I/R) injury, result from pathogenic factors, including damage-associated molecular pattern signaling, activation of innate immunity, and upregulation of proinflammatory cytokines. At the same time, a number of protective, or prosurvival, pathways are also activated, and the extent of end-organ damage is ultimately determined by the balance between these two systems. In liver I/R, members of the calcium/calmodulin-dependent protein kinase (CaMK) family are known to be activated, but their individual roles are largely unknown. In this study, we show that one CaMK member, CaMKIV, is protective in hepatic I/R by activating the prosurvival pathway of autophagy in hepatocytes. CaMKIV knockout mice experience significantly worse organ damage after I/R and are deficient in hepatocyte autophagic signaling. Restoration of autophagic signaling with rapamycin reduces organ damage in CaMKIV knockout mice to wild-type levels. In vitro, we show that CaMKIV activation induces autophagy in mouse hepatocytes, and that CaMKIV activation protects hepatocytes from oxidative stress-induced cell death. In conclusion, the protective autophagic signaling pathway serves to reduce organ damage following I/R and is regulated by activation of CaMKIV signaling in hepatocytes.
Collapse
Affiliation(s)
- John Evankovich
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Ruilin Zhang
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Jon S. Cardinal
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Lemeng Zhang
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Junda Chen
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Hai Huang
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Donna Beer-Stolz
- 2Department of Cell Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Matthew R. Rosengart
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| | - Allan Tsung
- 1Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania; and
| |
Collapse
|
298
|
LeCluyse EL, Witek RP, Andersen ME, Powers MJ. Organotypic liver culture models: meeting current challenges in toxicity testing. Crit Rev Toxicol 2012; 42:501-48. [PMID: 22582993 PMCID: PMC3423873 DOI: 10.3109/10408444.2012.682115] [Citation(s) in RCA: 248] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Revised: 03/26/2012] [Accepted: 03/30/2012] [Indexed: 02/07/2023]
Abstract
Prediction of chemical-induced hepatotoxicity in humans from in vitro data continues to be a significant challenge for the pharmaceutical and chemical industries. Generally, conventional in vitro hepatic model systems (i.e. 2-D static monocultures of primary or immortalized hepatocytes) are limited by their inability to maintain histotypic and phenotypic characteristics over time in culture, including stable expression of clearance and bioactivation pathways, as well as complex adaptive responses to chemical exposure. These systems are less than ideal for longer-term toxicity evaluations and elucidation of key cellular and molecular events involved in primary and secondary adaptation to chemical exposure, or for identification of important mediators of inflammation, proliferation and apoptosis. Progress in implementing a more effective strategy for in vitro-in vivo extrapolation and human risk assessment depends on significant advances in tissue culture technology and increasing their level of biological complexity. This article describes the current and ongoing need for more relevant, organotypic in vitro surrogate systems of human liver and recent efforts to recreate the multicellular architecture and hemodynamic properties of the liver using novel culture platforms. As these systems become more widely used for chemical and drug toxicity testing, there will be a corresponding need to establish standardized testing conditions, endpoint analyses and acceptance criteria. In the future, a balanced approach between sample throughput and biological relevance should provide better in vitro tools that are complementary with animal testing and assist in conducting more predictive human risk assessment.
Collapse
Affiliation(s)
- Edward L LeCluyse
- The Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA.
| | | | | | | |
Collapse
|
299
|
Sinha RA, You SH, Zhou J, Siddique MM, Bay BH, Zhu X, Privalsky ML, Cheng SY, Stevens RD, Summers SA, Newgard CB, Lazar MA, Yen PM. Thyroid hormone stimulates hepatic lipid catabolism via activation of autophagy. J Clin Invest 2012; 122:2428-2438. [PMID: 22684107 PMCID: PMC3386813 DOI: 10.1172/jci60580] [Citation(s) in RCA: 222] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 04/26/2012] [Indexed: 12/13/2022] Open
Abstract
For more than a century, thyroid hormones (THs) have been known to exert powerful catabolic effects, leading to weight loss. Although much has been learned about the molecular mechanisms used by TH receptors (TRs) to regulate gene expression, little is known about the mechanisms by which THs increase oxidative metabolism. Here, we report that TH stimulation of fatty acid β-oxidation is coupled with induction of hepatic autophagy to deliver fatty acids to mitochondria in cell culture and in vivo. Furthermore, blockade of autophagy by autophagy-related 5 (ATG5) siRNA markedly decreased TH-mediated fatty acid β-oxidation in cell culture and in vivo. Consistent with this model, autophagy was altered in livers of mice expressing a mutant TR that causes resistance to the actions of TH as well as in mice with mutant nuclear receptor corepressor (NCoR). These results demonstrate that THs can regulate lipid homeostasis via autophagy and help to explain how THs increase oxidative metabolism.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Seo-Hee You
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mobin M. Siddique
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Boon-Huat Bay
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Xuguang Zhu
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Martin L. Privalsky
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Sheue-Yann Cheng
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert D. Stevens
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Scott A. Summers
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Christopher B. Newgard
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mitchell A. Lazar
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Paul M. Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore.
Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
Department of Anatomy, Yong Loo Lin School of Medicine, Department of Anatomy, National University of Singapore, Singapore.
Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
Department of Microbiology, UCD, Davis, California, USA.
Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
300
|
Sinha RA, You SH, Zhou J, Siddique MM, Bay BH, Zhu X, Privalsky ML, Cheng SY, Stevens RD, Summers SA, Newgard CB, Lazar MA, Yen PM. Thyroid hormone stimulates hepatic lipid catabolism via activation of autophagy. J Clin Invest 2012. [PMID: 22684107 DOI: 10.1172/jci160580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For more than a century, thyroid hormones (THs) have been known to exert powerful catabolic effects, leading to weight loss. Although much has been learned about the molecular mechanisms used by TH receptors (TRs) to regulate gene expression, little is known about the mechanisms by which THs increase oxidative metabolism. Here, we report that TH stimulation of fatty acid β-oxidation is coupled with induction of hepatic autophagy to deliver fatty acids to mitochondria in cell culture and in vivo. Furthermore, blockade of autophagy by autophagy-related 5 (ATG5) siRNA markedly decreased TH-mediated fatty acid β-oxidation in cell culture and in vivo. Consistent with this model, autophagy was altered in livers of mice expressing a mutant TR that causes resistance to the actions of TH as well as in mice with mutant nuclear receptor corepressor (NCoR). These results demonstrate that THs can regulate lipid homeostasis via autophagy and help to explain how THs increase oxidative metabolism.
Collapse
Affiliation(s)
- Rohit Anthony Sinha
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|