1
|
Lim HJ, Kwak HJ. Selective PPARδ Agonist GW501516 Protects Against LPS-Induced Macrophage Inflammation and Acute Liver Failure in Mice via Suppressing Inflammatory Mediators. Molecules 2024; 29:5189. [PMID: 39519830 PMCID: PMC11547330 DOI: 10.3390/molecules29215189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammation is critical in the development of acute liver failure (ALF). Peroxisome proliferator-activated receptor delta (PPARδ) regulates anti-inflammatory responses and is protective in several diseases such as obesity and cancer. However, the beneficial effects and underlying mechanisms of PPARδ agonist GW501516 in ALF remain unclear. This study investigated the molecular mechanisms underlying the anti-inflammatory effects of GW501516 in macrophages and assessed its protective potential against lipopolysaccharide (LPS)/galactosamine (GalN)-induced ALF. In vivo administration of GW501516 significantly reduced LPS/GalN-induced hepatotoxicity, as evidenced by lower mortality, decreased liver damage, and attenuated secretion of IL-1β, IL-6, and TNF-α. GW501516 treatment also decreased LPS-induced nitric oxide synthase 2 (NOS2) expression and nitric oxide (NO) production in RAW264.7 cells, an effect reversed by PPARδ siRNA. Additionally, GW501516 inhibited LPS-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK), suggesting that inactivation of these MAPKs contributes to its effects. The secretion of IL-6, TNF-α, and NF-κB DNA-binding activity were also suppressed by GW501516, while the nuclear translocation of the NF-κB p65 subunit was unaffected. In conclusion, our findings suggest that GW501516 exerts protective effects in ALF by inhibiting the production of inflammatory mediators. Therefore, GW501516 may act as a potential agent for developing anti-inflammatory therapies for ALF.
Collapse
Affiliation(s)
- Hyun-Joung Lim
- Division of Cardiovascular Diseases Research, Department of Chronic Diseases Convergence, National Institute of Health, Cheongju 28159, Republic of Korea;
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
2
|
Feng Z, Bao S, Kong L, Chen X. MicroRNA-378 inhibits hepatocyte apoptosis during acute liver failure by targeting caspase-9 in mice. GASTROENTEROLOGIA Y HEPATOLOGIA 2023; 46:124-134. [PMID: 35964807 DOI: 10.1016/j.gastrohep.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 06/18/2022] [Accepted: 07/16/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Acute liver failure (ALF) is a severe and potentially lethal clinical syndrome. It has been demonstrated that micro ribonucleic acids (miRNAs) are crucial mediators of nearly all pathological processes, including liver disease. OBJECTIVE The present study investigates the role of miR-378 in ALF. An ALF mouse model was induced using intraperitoneal injections of d-galactosamine/lipopolysaccharide (d-GalN/LPS). A hepatocyte cell line and miR-378 analogue were used in vitro to investigate the possible roles of miR-378 in ALF. METHODS The expressions of miR-378 and predicted target genes were measured via reverse transcription-quantitative polymerase chain reaction and western blotting, and cell apoptosis was assayed using flow cytometry. RESULTS Compared with mice in the control group, the mice challenged with d-GalN/LPS showed higher levels of alanine aminotransferase, aspartate aminotransferase, tumour necrosis factor-alpha and interleukin-6, more severe liver damage and increased numbers of apoptotic hepatocytes. Hepatic miR-378 was distinctly downregulated, while messenger RNA and protein levels of cysteinyl aspartate specific proteinase 9 (caspase-9) were upregulated in the ALF model. Furthermore, miR-378 was downregulated in d-GalN/TNF-induced hepatocyte cells, and miR-378 was found to inhibit hepatocyte apoptosis by targeting caspase-9. CONCLUSION Together, the present results indicate that miR-378 is a previously unrecognised post-ALF hepatocyte apoptosis regulator and may be a potential therapeutic target in the context of ALF.
Collapse
Affiliation(s)
- Zhiwen Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Shenghua Bao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Lianbao Kong
- Department of Liver Transplantation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaopeng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.
| |
Collapse
|
3
|
Pu Y, Yang Z, Mo X. Protective Effect of Luteolin on D-Galactosamine (D-Gal)/Lipopolysaccharide (LPS) Induced Hepatic Injury by in Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:2252705. [PMID: 34368345 PMCID: PMC8342164 DOI: 10.1155/2021/2252705] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/19/2021] [Indexed: 01/13/2023]
Abstract
To observe the effects of luteolin on galactosamine (D-Gal)/lipopolysaccharide (LPS) induced liver injury in mice. Male C57BL/6 mice were randomly divided into 4 groups: normal control group, D-GaI/LPS group, D-GaI/LPS + luteolin (Lu, 20 mg/kg), and D-GaI/LPS + luteolin (Lu, 40 mg/kg). Mice in the normal control group and D-GaI/LPS group were given distilled water while other groups were given drugs in 7 days by gavage. 4 hours after the continuous administration, Gal (700 mg/kg) and LPS (10 mg/kg) were injected intraperitoneally. Mice in the normal control group were given the same volume of vegetable oil solution. 24 h after the establishment of the mice model, blood and liver samples were collected. Hematoxylin (HE) staining was used to observe the changes of hepatic histopathology. Alanine aminotransferase (ALT) and glutamic oxalacetic transaminase (AST) in serum, interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor (TNF-α) were measured by related kits. Western blotting was used to demonstrate the expression levels of related inflammation proteins. Lu significantly reduced levels of proinflammatory cytokines including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) in serum and liver. Lu restored the pathological changes after galactosamine (D-Gal)/lipopolysaccharide (LPS) treatment. In addition, Lu regulated proteins levels of the NLRP3/NF-κB pathway in liver. Lu exhibited therapeutical effects on D-GaI/LPS induced liver injury in mice which might be related to the regulation of the NLRP3/NF-κB pathway.
Collapse
Affiliation(s)
- Yiwei Pu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhaocong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Jia F, Deng F, Xu P, Li S, Wang X, Hu P, Ren H, Tong S, Yin W. NOD1 Agonist Protects Against Lipopolysaccharide and D-Galactosamine-Induced Fatal Hepatitis Through the Upregulation of A20 Expression in Hepatocytes. Front Immunol 2021; 12:603192. [PMID: 33746949 PMCID: PMC7969647 DOI: 10.3389/fimmu.2021.603192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 02/17/2021] [Indexed: 01/13/2023] Open
Abstract
Increasing evidence suggests that NODs are involved in liver diseases; however, the underlying mechanisms remain obscure. In the present study, we analyzed the effect of NOD1 agonist pretreatment on acute liver failure induced by lipopolysaccharide (LPS) in D-galactosamine (D-GalN)-sensitized mice. We found that pretreatment with the NOD1 agonist markedly reduced LPS/D-GalN-induced mortality, elevation of serum ALT levels, and hepatocyte apoptosis. The protective effect of NOD1 agonist was independent of tumor necrosis factor (TNF)-α inhibition. NOD1 agonist pretreatment also attenuated TNF-α/D-GalN-induced apoptotic liver damage. The anti-apoptotic protein A20 expression was more pronounced in NOD1 agonist pretreated mice than in controls, and knockdown of A20 abrogated the protective effect of NOD1 agonist on LPS/D-GalN-induced liver injury and hepatocyte apoptosis. Further experiments showed that NOD1 agonist-induced A20 upregulation required the presence of kupffer cells and TNF-α. Taken together, our data strongly indicate that NOD1 is involved in the regulation of liver injury and could be a potential therapeutic target for liver diseases.
Collapse
Affiliation(s)
- Fang Jia
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Department of Endocrinology, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an Jiaotong University, Xi'an, China
| | - Fuxue Deng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an Jiaotong University, Xi'an, China
| | - Pan Xu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiying Li
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xuefu Wang
- School of Pharmacy, Anhui Medical University, Hefei, China
| | - Peng Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiwen Tong
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenwei Yin
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Wu S, Huang X, Sun W, Chen L, Huang Y, Wang Y, Luo E, Qin A, Zhao W, Gan J. Role of the microRNA‑214/Bax axis in the progression of acute liver failure. Mol Med Rep 2020; 22:117-126. [PMID: 32377732 PMCID: PMC7248488 DOI: 10.3892/mmr.2020.11123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 02/28/2020] [Indexed: 12/25/2022] Open
Abstract
Acute liver failure (ALF) is a fatal liver disease characterized by severe hepatocyte destruction. MicroRNAs (miRNAs/miRs) have been reported to serve a key role in a number of liver diseases. Therefore, the aim of the present study was to investigate the role and underlying mechanism of miR-214 in ALF. ALF murine and hepatocyte models were established using D-galactosamine (D-GalN) and lipopolysaccharide (LPS) or D-GalN + tumor necrosis factor (TNF)-α, respectively. The expression levels of miR-214 and Bax were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and/or western blotting. Furthermore, an automatic biochemical analyzer was used to measure the levels of aspartate aminotransferase (AST) or alanine aminotransferase (ALT). The levels of TNF-α and interleukin (IL)-6 were detected by ELISA and RT-qPCR. In addition, TUNEL staining and flow cytometry were used to analyze cell apoptosis, and the protein expression of caspase-3 was determined by western blotting. It was identified that the levels of AST and ALT were increased and that hepatocyte apoptosis was enhanced in the D-GalN/LPS-stimulated group compared with the control. Furthermore, higher expression of caspase-3 was observed in the D-GalN/LPS-stimulated group. In addition, it was demonstrated that miR-214 was downregulated, while Bax was upregulated in D-GalN/LPS-stimulated mice and D-GalN/TNF-α-stimulated BNLCL2 cells. Moreover, in D-GalN/TNF-α-stimulated BNLCL2 cells, miR-214 overexpression suppressed apoptosis and decreased TNF-α and IL-6 levels, and these effects were reversed by the Bax plasmid. It was also identified that overexpression of miR-214 significantly decreased Bax mRNA and protein expression levels in vitro. Collectively, the present results suggested that miR-214 inhibited hepatocyte apoptosis during ALF development via targeting Bax, thus indicating that miR-214 may be a potential target for ALF treatment.
Collapse
Affiliation(s)
- Shaohong Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Xiaoping Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Wei Sun
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Li Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Yan Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Yan Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Erping Luo
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Ailan Qin
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Weifeng Zhao
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| | - Jianhe Gan
- Department of Infectious Diseases, The First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
6
|
Jing ZT, Liu W, Xue CR, Wu SX, Chen WN, Lin XJ, Lin X. AKT activator SC79 protects hepatocytes from TNF-α-mediated apoptosis and alleviates d-Gal/LPS-induced liver injury. Am J Physiol Gastrointest Liver Physiol 2019; 316:G387-G396. [PMID: 30629471 DOI: 10.1152/ajpgi.00350.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a highly pleiotropic cytokine executing biological functions as diverse as cell proliferation, metabolic activation, inflammatory responses, and cell death. TNF-α can induce multiple mechanisms to initiate apoptosis in hepatocytes leading to the subsequent liver injury. Since the phosphoinositide-3-kinase/protein kinase B (PI3K/Akt) pathway is known to have a protective role in death factor-mediated apoptosis, it is our hypothesis that activation of Akt may represent a therapeutic strategy to alleviate TNF-α-induced hepatocyte apoptosis and liver injury. We report here that the Akt activator SC79 protects hepatocytes from TNF-α-induced apoptosis and protects mice from d-galactosamine (d-Gal)/lipopolysaccharide (LPS)-induced TNF-α-mediated liver injury and damage. SC79 not only enhances the nuclear factor-κB (NF-κB) prosurvival signaling in response to TNF-α stimulation, but also increases the expression of cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein L and S (FLIPL/S), which consequently inhibits the activation of procaspase-8. Furthermore, pretreatment of the PI3K/Akt inhibitor LY294002 reverses all the SC79-induced hepatoprotective effects. These results strongly indicate that SC79 protects against TNF-α-induced hepatocyte apoptosis and suggests that SC79 is likely a promising therapeutic agent for ameliorating the development of liver injury. NEW & NOTEWORTHY SC79 protects hepatocytes from TNF-α-mediated apoptosis and mice from Gal/LPS-induced liver injury and damage. Cytoprotective effects of SC79 against TNF-α act through both AKT-mediated activation of NF-κB and upregulation of FLIPL/S.
Collapse
Affiliation(s)
- Zhen-Tang Jing
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China
| | - Wei Liu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University , Fuzhou , China
| | - Chao-Rong Xue
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China
| | - Shu-Xiang Wu
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China
| | - Wan-Nan Chen
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University , Fuzhou , China
| | - Xin-Jian Lin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China
| | - Xu Lin
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University , Fuzhou , China.,Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University , Fuzhou , China
| |
Collapse
|
7
|
A systems pharmacology-oriented discovery of a new therapeutic use of the TCM formula Liuweiwuling for liver failure. Sci Rep 2018; 8:5645. [PMID: 29618826 PMCID: PMC5884779 DOI: 10.1038/s41598-018-21515-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 02/06/2018] [Indexed: 02/08/2023] Open
Abstract
Multiple components of traditional Chinese medicine (TCM) formulae determine their treatment targets for multiple diseases as opposed to a particular disease. However, discovering the unexplored therapeutic potential of a TCM formula remains challenging and costly. Inspired by the drug repositioning methodology, we propose an integrated strategy to feasibly identify new therapeutic uses for a formula composed of six herbs, Liuweiwuling. First, we developed a comprehensive systems approach to enrich drug compound-liver disease networks to analyse the major predicted diseases of Liuweiwuling and discover its potential effect on liver failure. The underlying mechanisms were subsequently predicted to mainly attribute to a blockade of hepatocyte apoptosis via a synergistic combination of multiple effects. Next, a classical pharmacology experiment was designed to validate the effects of Liuweiwuling on different models of fulminant liver failure induced by D-galactosamine/lipopolysaccharide (GalN/LPS) or thioacetamide (TAA). The results indicated that pretreatment with Liuweiwuling restored liver function and reduced lethality induced by GalN/LPS or TAA in a dose-dependent manner, which was partially attributable to the abrogation of hepatocyte apoptosis by multiple synergistic effects. In summary, the integrated strategy discussed in this paper may provide a new approach for the more efficient discovery of new therapeutic uses for TCM formulae.
Collapse
|
8
|
Li D, Lu N, Han J, Chen X, Hao W, Xu W, Liu X, Ye L, Zheng Q. Eriodictyol Attenuates Myocardial Ischemia-Reperfusion Injury through the Activation of JAK2. Front Pharmacol 2018; 9:33. [PMID: 29441020 PMCID: PMC5797583 DOI: 10.3389/fphar.2018.00033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/11/2018] [Indexed: 01/01/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury remains the leading risk factor of disability and mortality worldwide. In this study, the myocardial protective effect of eriodictyol (EDT) and the underlying mechanism in an ex vivo model of global myocardial I/R was investigated. After treatment with different concentrations of EDT, the decreased hemodynamic parameters induced by myocardial I/R injury were significantly attenuated by EDT. The elevated levels of IL-6, CRP, IL-8, and TNF-α were effectively reduced by EDT treatment. EDT also remarkably suppressed the levels of Bax and cleaved Caspase-3, and up-regulated the level of Bcl-2 in cardiac tissues from EDT-treated groups. Further studies showed that EDT could increase the levels of p-JAK2 and p-STAT3 in cardiac tissues. Meanwhile, treatment of AG490, a specific inhibitor of JAK2, abolished the protective effect of EDT on hemodynamic parameters, myocardial inflammation and myocardial cell apoptosis induced by I/R injury. These results demonstrated that EDT could protect against myocardial I/R injury through the activation of JAK2, providing a potential treatment with EDT during myocardial I/R injury.
Collapse
Affiliation(s)
- Defang Li
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Ning Lu
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| | - Jichun Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Xiaoyu Chen
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Wenjin Hao
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Wenjuan Xu
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Xiaona Liu
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Lei Ye
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Qiusheng Zheng
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, China
| |
Collapse
|
9
|
Zeng X, Yu X, Xiao S, Yao H, Zhu J. Effects of 1,25-dihydroxyvitamin D3 on pathological changes in rats with diabetic cardiomyopathy. Lipids Health Dis 2017; 16:109. [PMID: 28595623 PMCID: PMC5465473 DOI: 10.1186/s12944-017-0498-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 05/25/2017] [Indexed: 12/18/2022] Open
Abstract
Background The role of 1,25-dihydroxyvitamin D3 (vitamin D) in the apoptosis of diabetic cardiomyopathy (DCM) is unclear. This study is to investigate the effects of vitamin D on the pathological changes in rats with DCM. Methods Rats were randomly divided into the control, model, and treatment groups. DCM model was established by the high-fat and -sugar diet. Plasma glucose, body weight, heart weight, heart weight index, and serum levels of lactate dehydrogenase (LDH) and creatine kinase (CK) were determined. Heart tissue morphology was detected with histochemical staining. Expression levels of Fas and FasL were detected with RT-PCR and immunohistochemistry. Results Compared with the control group, the body weights and heart weights were significantly declined, while the plasma glucose levels and heart weight indexes were significantly elevated, in the model group (P < 0.05). However, vitamin D significantly reversed the pathological changes in DCM rats (P < 0.05). Moreover, the serum levels of LDH and CK were significantly increased in the models, which were significantly decreased by vitamin D (P < 0.05). HE staining showed that, vitamin D significantly alleviated the histological changes of myocardial cells in DCM rats. In addition, the mRNA and protein expression levels of Fas and FasL were significantly elevated in the models (P < 0.05), which were significantly declined by vitamin D (P < 0.05). Conclusion Vitamin D could alleviate pathological changes, reduce Fas/FasL expression, and attenuate myocardial cell apoptosis in DCM rats, which might be used as a potential effective therapy for the disease.
Collapse
Affiliation(s)
- Xiaoyun Zeng
- Department of Endocrinology, the First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830054, China.,Prevention, Diagnosis and Treatment Center of Diabetes, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Xintian Yu
- Department of Endocrinology, the First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830054, China
| | - Shan Xiao
- Department of Endocrinology, the First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830054, China.,Prevention, Diagnosis and Treatment Center of Diabetes, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Hua Yao
- Prevention, Diagnosis and Treatment Center of Diabetes, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.,Xinjiang Key Laboratory of Metabolic Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Jun Zhu
- Department of Endocrinology, the First Affiliated Hospital of Xinjiang Medical University, No. 137, Liyushan South Road, Urumqi, Xinjiang, 830054, China. .,Prevention, Diagnosis and Treatment Center of Diabetes, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China. .,Xinjiang Key Laboratory of Metabolic Disease, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
10
|
Zhang X, Ding J, Gou C, Wen T, Li L, Wang X, Yang H, Liu D, Lou J, Chen D, Ren F, Li X. Qingchangligan formula attenuates the inflammatory response to protect the liver from acute failure induced by d-galactosamine/lipopolysaccharide in mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 201:108-116. [PMID: 27833028 DOI: 10.1016/j.jep.2016.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/26/2016] [Accepted: 11/04/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Qingchangligan formula, a traditional Chinese medicine comprising five herbs, is useful for treatment of patients with liver failure; however, its protective and regulatory mechanisms remain elusive. AIM OF THE STUDY To test the hypothesis that the Qingchangligan formula protects mice against acute liver failure by inhibiting liver inflammation. MATERIALS AND METHODS Acute liver failure (ALF) was induced by intraperitoneal injection of D-GalN (700mg/kg) plus LPS (10μg/kg). The Qingchangligan formula was administered to mice in three doses of 50mg/kg (on day 1, day 2, and day 3) prior to D-GalN/LPS injection by intragastric administration. The mice in different groups were sacrificed at 6h after D-GalN/LPS injection, and liver samples and blood were collected for analysis. RESULTS Administration of the Qingchangligan formula not only ameliorated liver injury, as evidenced by reduced transaminase levels and well-preserved liver architecture, but also decreased the lethality in ALF mice. Moreover, in the ALF model, pretreatment with the Qingchangligan formula alleviated liver inflammation and decreased hepatocyte apoptosis. Further demonstrating the protective effects of the Qingchangligan formula, we found that pretreatment with the Qingchangligan formula reduced the expression of inflammatory cytokines by decreasing the expression of components of the mitogen-activated protein kinase (MAPK) pathway and promoting autophagy in vitro and in vivo. CONCLUSIONS Our findings demonstrated that the Qingchangligan formula exerts a protective effect against the pathophysiology of ALF, especially in regulating liver inflammation, and provide a rationale for using the Qingchangligan formula as a potential therapeutic strategy to ameliorate ALF.
Collapse
Affiliation(s)
- Xiangying Zhang
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China; Beijing Institute of Hepatology, Beijing 100069, PR China.
| | - Jianbo Ding
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Chunyan Gou
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Tao Wen
- Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100043, PR China.
| | - Li Li
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Xiaojun Wang
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Huasheng Yang
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Dan Liu
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Jinli Lou
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Dexi Chen
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China; Beijing Institute of Hepatology, Beijing 100069, PR China.
| | - Feng Ren
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China; Beijing Institute of Hepatology, Beijing 100069, PR China.
| | - Xiuhui Li
- Beijing You-An Hospital, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
11
|
Tanoi T, Tamura T, Sano N, Nakayama K, Fukunaga K, Zheng YW, Akhter A, Sakurai Y, Hayashi Y, Harashima H, Ohkohchi N. Protecting liver sinusoidal endothelial cells suppresses apoptosis in acute liver damage. Hepatol Res 2016; 46:697-706. [PMID: 26490536 DOI: 10.1111/hepr.12607] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/30/2015] [Accepted: 10/14/2015] [Indexed: 01/01/2023]
Abstract
AIM Apoptosis is associated with various types of hepatic disorders. We have developed a novel cell-transfer drug delivery system (DDS) using a multifunctional envelope-type nano device that targets liver sinusoidal endothelial cells (LSECs). The purpose of this study was to determine the efficacy of the novel DDS containing siRNA at suppressing apoptosis in LSECs. METHODS Bax siRNA was transfected into a sinusoidal endothelial cell line (M1) to suppress apoptosis induced by an anti-Fas antibody and staurosporine. C57BL/6J mice were divided into three groups: (i) a control group, only intravenous saline; (ii) a nonselective group, injections of siRNA sealed in the nonselective DDS; and (iii) an LSEC-transfer efficient group, injections of siRNA sealed in an LSEC-transfer efficient DDS. Hepatic cell apoptosis was induced by an anti-Fas antibody. RESULTS Bax siRNA had an anti-apoptotic effect on M1 cells. Serum alanine aminotransferase was reduced in the LSEC-transfer efficient group, as were cleaved caspase-3 and the number of terminal deoxynucleotidyl transferase dUTP nick end labeling positive hepatocytes. Silver impregnation staining indicated that the sinusoidal space was maintained in the LSEC-transfer efficient group but not in the other groups. Electron microscopy showed that the LSECs were slightly impaired, although the sinusoidal structure was maintained in the LSEC-transfer efficient group. CONCLUSION Hepatocyte apoptosis was reduced by the efficient suppression of LSEC apoptosis with a novel DDS. Protecting the sinusoidal structure by suppressing LSEC damage will be an effective treatment for acute liver failure.
Collapse
Affiliation(s)
- Tomohito Tanoi
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takafumi Tamura
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoki Sano
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Ken Nakayama
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kiyoshi Fukunaga
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yun-Wen Zheng
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Afsana Akhter
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yu Sakurai
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Yasuhiro Hayashi
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hideyoshi Harashima
- Laboratory of Innovative Nanomedicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Nobuhiro Ohkohchi
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
12
|
Komarov AP, Komarova EA, Green K, Novototskaya LR, Baker PS, Eroshkin A, Osterman AL, Chenchick AA, Frangou C, Gudkov AV. Functional genetics-directed identification of novel pharmacological inhibitors of FAS- and TNF-dependent apoptosis that protect mice from acute liver failure. Cell Death Dis 2016; 7:e2145. [PMID: 26986512 PMCID: PMC4823946 DOI: 10.1038/cddis.2016.45] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 01/25/2016] [Accepted: 02/04/2016] [Indexed: 12/12/2022]
Abstract
shRNA-mediated gene-silencing technology paired with cell-based functional readouts reveals potential targets directly, providing an opportunity to identify drugs against the target without knowing the precise role of the target in the pathophysiological processes of interest. By screening a lentiviral shRNA library targeting for major components of human signaling pathways and known drug targets, we identified and validated both canonical as well as 52 novel mediators of FAS and TNF ligand-induced apoptosis. Presence of potential therapeutic targets among these mediators was confirmed by demonstration of in vivo activity of siRNAs against four identified target candidates that protected mice from acute liver failure (ALF), a life-threatening disease with known involvement of death receptor (DR)-mediated apoptosis. Network-based modeling was used to predict small-molecule inhibitors for several candidate apoptosis mediators, including somatostatin receptor 5 (SSTR5) and a regulatory subunit of PP2A phosphatase, PPP2R5A. Remarkably, pharmacological inhibition of either SSTR5 or PPP2R5A reduced apoptosis induced by either FASL or TNF in cultured cells and dramatically improved survival in several mouse models of ALF. These results demonstrate the utility of loss-of-function genetic screens and network-based drug-repositioning methods for expedited identification of targeted drug candidates and revealed pharmacological agents potentially suitable for treatment of DR-mediated pathologies.
Collapse
Affiliation(s)
| | - E A Komarova
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - K Green
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - L R Novototskaya
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - P S Baker
- Buffalo BioLabs, LLC, Buffalo, NY, USA
| | - A Eroshkin
- Infectious and Inflammatory Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - A L Osterman
- Buffalo BioLabs, LLC, Buffalo, NY, USA
- Infectious and Inflammatory Disease Center, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - C Frangou
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - A V Gudkov
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
13
|
Chang A, Chen Y, Shen W, Gao R, Zhou W, Yang S, Liu Y, Luo Y, Chuang TH, Sun P, Liu C, Xiang R. Ifit1 Protects Against Lipopolysaccharide and D-galactosamine-Induced Fatal Hepatitis by Inhibiting Activation of the JNK Pathway. J Infect Dis 2016; 212:1509-20. [PMID: 26459629 DOI: 10.1093/infdis/jiv221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Treatment of mice with lipopolysaccharide (LPS) and the liver-specific transcriptional inhibitor D-(+)-galactosamine (GalN) induces fatal hepatitis, which is mediated by tumor necrosis factor α (TNF-α) and characterized by massive hepatic apoptosis. Previous studies suggest that GalN increases the sensitivity to LPS/TNF-α, probably by blocking the transcription of protective factors, but the identity of most of these factors is still unclear. Here, we report that Ifit1 protects against LPS/GalN-induced fatal hepatitis. Forced expression of Ifit1 in hepatocytes significantly diminished TNF-α-mediated apoptosis. Moreover, targeted expression of Ifit1 in the liver by recombinant adeno-associated virus serotype 8 protected mice from LPS/GalN-induced lethal hepatitis, which was associated with the inhibition of TNF-α-mediated activation of the c-Jun N-terminal kinase (JNK)-Bim cascade. Furthermore, Ifit1 bound to a scaffolding protein Axin and inhibited its function to mediate JNK activation. Together, our data demonstrate that Ifit1 is a novel protective factor that inhibits LPS/GalN-induced (TNF-α-mediated) fatal hepatitis, suggesting that Ifit1 is a potential therapeutic target for treatment of inflammatory liver diseases.
Collapse
Affiliation(s)
- Antao Chang
- School of Medicine, Nankai University, Tianjin Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University
| | - Yanan Chen
- School of Medicine, Nankai University, Tianjin
| | - Wenzhi Shen
- School of Medicine, Nankai University, Tianjin
| | - Ruifang Gao
- School of Medicine, Nankai University, Tianjin
| | - Wei Zhou
- School of Medicine, Nankai University, Tianjin
| | - Shuang Yang
- School of Medicine, Nankai University, Tianjin
| | - Yanhua Liu
- School of Medicine, Nankai University, Tianjin
| | - Yunping Luo
- Department of Immunology, School of Basic Medicine, Peking Union Medical College, Beijing, People's Republic of China
| | - Tsung-Hsien Chuang
- Immunology Research Center, National Health Research Institutes, Miaoli, Taiwan
| | - Peiqing Sun
- Department of Cell and Molecular Biology, Scripps Research Institute, La Jolla, California
| | - Chenghu Liu
- School of Medicine, Nankai University, Tianjin
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin
| |
Collapse
|
14
|
Renna MS, Figueredo CM, Rodríguez-Galán MC, Icely PA, Cejas H, Cano R, Correa SG, Sotomayor CE. Candida albicans up-regulates the Fas-L expression in liver Natural Killer and Natural Killer T cells. Immunobiology 2015; 220:1210-8. [DOI: 10.1016/j.imbio.2015.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/12/2015] [Accepted: 06/05/2015] [Indexed: 01/01/2023]
|
15
|
Luo M, Zhao A, Li J, Chen Y, Tian D, Wang C, Hu Z, Gao J. Acute liver injury attenuation of a novel recombinant sTNFR through blocking hepatic apoptosis. Immunopharmacol Immunotoxicol 2015; 37:295-300. [PMID: 25982795 DOI: 10.3109/08923973.2015.1035390] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
CONTEXT Tumor necrosis factor (TNF) α plays a key role in acute liver injury (ALI) induced by injection of d-galactosamine (D-Gal)/lipopolysaccharide (LPS). A novel recombinant trimeric sTNFRII, sTNFRII-gAD, has been tested to be effective in ameliorating ALI, when administered prior to ALI establishment. This study aims to validate the protective effect of sTNFRII-gAD when given after ALI setup and further explore its effect on hepatic apoptosis. MATERIALS AND METHODS The treatments were carried out concomitantly with ALI establishment with clinically approved sTNFRII-Fc (the dimeric sTNFRII) as a positive control. Lethality, liver weight, and serum alanine transaminase were measured, and histological analysis was performed to evaluate liver injury induced by D-Gal/LPS. Additionally, Terminal-deoxynucleoitidyl transferase-mediated nick end labeling (TUNEL) and Western blot analyses of caspase-3 were used to examine hepatocellular apoptosis. RESULTS sTNFRII-gAD given after D-Gal/LPS injection turned out to attenuate animal mortality significantly (p < 0.01), and had better hepatic protection. In terms of apoptosis, both sTNFRII-gAD and sTNFRII-Fc displayed noticeable improvement of apoptosis evidenced by dramatic decline of active caspase-3 compared to the control group. CONCLUSIONS The results demonstrated that sTNFRII-gAD therapeutically diminished the lethality induced by D-Gal/LPS, possibly through blocking hepatic apoptosis initiated by TNFα. Of note, sTNFRII-gAD was superior to sTNFRII-Fc in some respects, indicating a promising alternative for the therapeutic strategy against the diseases associated with excessive TNFα.
Collapse
Affiliation(s)
- Mansheng Luo
- Department of Microbiology & Immunology, School of Medicine, JingGangShan University , Jian , China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Salvianolic acid A attenuates TNF-α- and d-GalN-induced ER stress-mediated and mitochondrial-dependent apoptosis by modulating Bax/Bcl-2 ratio and calcium release in hepatocyte LO2 cells. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:817-30. [DOI: 10.1007/s00210-015-1116-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 03/13/2015] [Indexed: 02/07/2023]
|
17
|
Kuhla A, Thrum M, Schaeper U, Fehring V, Schulze-Topphoff U, Abshagen K, Vollmar B. Liver-specific Fas silencing prevents galactosamine/lipopolysaccharide-induced liver injury. Apoptosis 2015; 20:500-11. [DOI: 10.1007/s10495-015-1088-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Sayed RH, Khalil WKB, Salem HA, Kenawy SA, El-Sayeh BM. Sulforaphane increases the survival rate in rats with fulminant hepatic failure induced by D-galactosamine and lipopolysaccharide. Nutr Res 2014; 34:982-9. [PMID: 25439027 DOI: 10.1016/j.nutres.2014.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/28/2014] [Accepted: 10/02/2014] [Indexed: 12/14/2022]
Abstract
Fulminant hepatic failure (FHF) is a life-threatening clinical syndrome, with liver transplantation being the only effective therapy. Sulforaphane (SFN) is a natural compound that is extracted from cruciferous vegetables and possesses potent anti-inflammatory, antioxidant, and anticancer activities. This study was designed to test the hypothesis that SFN (3 mg/kg) may protect against FHF induced in rats by administering a combination of D-galactosamine (GalN; 300 mg/kg) and lipopolysaccharide (LPS; 30 μg/kg). The rats were given a single intraperitoneal injection of SFN, 1 hour before the FHF induction. Sulforaphane reduced the mortality and alleviated the pathological liver injury. In addition, SFN significantly reduced the increase in serum aminotransferase activities and lipid peroxidation. The glutathione content decreased in the GalN/LPS group, and this decrease was attenuated by SFN. Increases in serum tumor necrosis factor α, interleukin-6, and interleukin-10, which were observed in GalN/LPS-treated rats, were significantly reduced after using SFN. The GalN/LPS treatment increased the expression of superoxide dismutase-1, glutathione peroxidase 2, catalase, and heme oxygenase-1 genes. Sulforaphane inhibited the induction of reactive oxygen species scavenging proteins. Moreover, SFN inhibited GalN/LPS-induced caspase-3 activation and suppressed FAS and FASL expression. These findings suggest that SFN alleviates GalN/LPS-induced liver injury, possibly by exerting antioxidant, anti-inflammatory, and antiapoptotic effects and modulating certain antioxidant defense enzymes.
Collapse
Affiliation(s)
- Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Wagdy K B Khalil
- Department of Cell Biology, National Research Centre, Cairo, Egypt
| | - Hesham A Salem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Sanaa A Kenawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Bahia M El-Sayeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
19
|
Protective effect of linarin against D-galactosamine and lipopolysaccharide-induced fulminant hepatic failure. Eur J Pharmacol 2014; 738:66-73. [PMID: 24877692 DOI: 10.1016/j.ejphar.2014.05.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 05/01/2014] [Accepted: 05/10/2014] [Indexed: 11/20/2022]
Abstract
Linarin was isolated from Chrysanthemum indicum L. Fulminant hepatic failure is a serious clinical syndrome that results in massive inflammation and hepatocyte death. Apoptosis is an important cellular pathological process in d-galactosamine (GalN)/lipopolysaccharide (LPS)-induced liver injury, and regulation of liver apoptosis might be an effective therapeutic method for fulminant hepatic failure. This study examined the cytoprotective mechanisms of linarin against GalN/LPS-induced hepatic failure. Mice were given an oral administration of linarin (12.5, 25 and 50mg/kg) 1h before receiving GalN (800 mg/kg)/LPS (40 μg/kg). Linarin treatment reversed the lethality induced by GalN/LPS. After 6h of GalN/LPS injection, the serum levels of alanine aminotransferase, aspartate aminotransferase, tumor necrosis factor (TNF)-α, interleukin-6 and interferon-γ were significantly elevated. GalN/LPS increased toll-like receptor 4 and interleukin-1 receptor-associated kinase protein expression. These increases were attenuated by linarin. Linarin attenuated the increased expression of Fas-associated death domain and caspase-8 induced by GalN/LPS, reduced the cytosolic release of cytochrome c and caspase-3 cleavage induced by GalN/LPS, and reduced the pro-apoptotic Bim phosphorylation induced by GalN/LPS. However, linarin increased the level of anti-apoptotic Bcl-xL and phosphorylation of STAT3. Our results suggest that linarin alleviates GalN/LPS-induced liver injury by suppressing TNF-α-mediated apoptotic pathways.
Collapse
|
20
|
Ganai AA, Jahan S, Ahad A, Abdin MZ, Farooqi H. Glycine propionyl l-carnitine attenuates d-Galactosamine induced fulminant hepatic failure in wistar rats. Chem Biol Interact 2014; 214:33-40. [PMID: 24565947 DOI: 10.1016/j.cbi.2014.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/21/2014] [Accepted: 02/13/2014] [Indexed: 10/25/2022]
Abstract
Glycine propionyl l-carnitine (GPLC) is a propionyl ester of carnitine that includes an additional glycine component. The present study evaluated hepatoprotective effect of GPLC in d-Galactosamine (d-GalN) induced fulminant hepatic failure. Rats were intraperitonially administered d-GalN (700mg/kgBW). GPLC was given as a pre-treatment (35mg/kgBW/day) for 1month followed by a single dose of d-GalN on the 31st day. d-GalN administration resulted in increased mortality and serum ALT and AST activities. These increases were significantly attenuated by GPLC. d-GalN treatment increased hepatic lipid peroxidation and a decrease in reduced glutathione content was observed. GPLC pre-treatment significantly decreased lipid peroxidation and augmented the level of GSH. d-GalN increased the circulating level of TNF-α and ATM-Kinase and MAP-Kinase expression. GPLC supplementation inhibited the increase in serum TNF-α and ATM-Kinase and MAP-Kinase expression. d-GalN treatment increased the level of Bax and Caspase-3 m-RNA while as a decline was observed in Bcl2 m-RNA. GPLC prevented the increase in Caspase-3 and Bax m-RNA and at the same time augmented the expression of Bcl2 m-RNA. Our findings suggest that GPLC alleviates d-GalN induced liver injury by strengthening antioxidative defense system and reducing apoptotic signalling pathways.
Collapse
Affiliation(s)
- Ajaz A Ganai
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India.
| | - Sadaf Jahan
- Developmental Toxicology Division, Indian Institute of Toxicology Research, Lucknow 226001, Uttar Pradesh, India.
| | - Amjid Ahad
- Department of Biochemistry, Jamia Hamdard, New Delhi 110062, India.
| | - M Z Abdin
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India.
| | - Humaira Farooqi
- Department of Biotechnology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
21
|
Zong L, Yu QH, Du YX, Deng XM. Edaravone protects endotoxin-induced liver injury by inhibiting apoptosis and reducing proinflammatory cytokines. ACTA ACUST UNITED AC 2014; 47:231-6. [PMID: 24554039 PMCID: PMC3982944 DOI: 10.1590/1414-431x20133186] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 10/22/2013] [Indexed: 11/25/2022]
Abstract
Studies have shown that edaravone may prevent liver injury. This study aimed to investigate the effects of edaravone on the liver injury induced by D-galactosamine (GalN) and lipopolysaccharide (LPS) in female BALB/c mice. Edaravone was injected into mice 30 min before and 4 h after GalN/LPS injection. The survival rate was determined within the first 24 h. Animals were killed 8 h after GalN/LPS injection, and liver injury was biochemically and histologically assessed. Hepatocyte apoptosis was measured by TUNEL staining; proinflammatory cytokines [tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6)] in the liver were assayed by ELISA; expression of caspase-8 and caspase-3 proteins was detected by Western blot assay; and caspase-3 activity was also determined. Results showed that GalN/LPS induced marked elevations in serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT). Edaravone significantly inhibited elevation of serum AST and ALT, accompanied by an improvement in histological findings. Edaravone lowered the levels of TNF-α and IL-6 and reduced the number of TUNEL-positive cells. In addition, 24 h after edaravone treatment, caspase-3 activity and mortality were reduced. Edaravone may effectively ameliorate GalN/LPS-induced liver injury in mice by reducing proinflammatory cytokines and inhibiting apoptosis.
Collapse
Affiliation(s)
- L Zong
- Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Q H Yu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Y X Du
- Department of Anesthesiology, No. 82 Hospital of People's Liberation Army, Jiangsu, China
| | - X M Deng
- Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
22
|
Kuhla A, Hauke M, Sempert K, Vollmar B, Zechner D. Senescence-dependent impact of anti-RAGE antibody on endotoxemic liver failure. AGE (DORDRECHT, NETHERLANDS) 2013; 35:2153-2163. [PMID: 23319363 PMCID: PMC3824992 DOI: 10.1007/s11357-012-9506-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 12/27/2012] [Indexed: 06/01/2023]
Abstract
Aging often restricts the capacity of the immune system. Endotoxemia is characterized by an immune response initiated by a group of pattern recognition receptors including the receptor for advanced glycation end products (RAGE). The aim of this study was to clarify to which extent RAGE and its signaling pathways such as the so called mitogen-activated protein kinase (MAPK) pathways can contribute to the perpetuation of inflammation in the aging organism. We used senescence-accelerated-prone (SAMP8) and senescence-accelerated-resistant (SAMR1) mice and studied them at the age of 2 and 6 months. Livers of SAMP8 mice had significantly higher malondialdehyde concentrations and a modest reduction of glyoxalase-I expression. Consequently, the abundance of highly modified advanced glycation end products was increased in the liver and plasma of these mice. After galactosamine/lipopolysaccharide-induced acute liver injury, significant activation of the MAPK cascade was observed in both mouse strains. Administration of an anti-RAGE antibody diminished p42/44-phosphorylation as well as tissue injury in SAMP8 mice, whereas the identical treatment in SAMR1 mice leads to a significant increase in p42/44-phosphorylation and intensified liver injury. This observation suggests that dependent on the senescence of the organism, anti-RAGE antibody can have differential effects on the progression of endotoxemic liver failure.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| | - Mandy Hauke
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| | - Kai Sempert
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| | - Dietmar Zechner
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, 18057 Rostock, Germany
| |
Collapse
|
23
|
Liang DY, Liu LM, Ye CG, Zhao L, Yu FP, Gao DY, Wang YY, Yang ZW, Wang YY. Inhibition of UII/UTR system relieves acute inflammation of liver through preventing activation of NF-κB pathway in ALF mice. PLoS One 2013; 8:e64895. [PMID: 23755157 PMCID: PMC3670940 DOI: 10.1371/journal.pone.0064895] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/19/2013] [Indexed: 02/06/2023] Open
Abstract
Urotensin II (UII) is implicated in immune inflammatory diseases through its specific high-affinity UT receptor (UTR). Enhanced expression of UII/UTR was recently demonstrated in the liver with acute liver failure (ALF). Here, we analysed the relationship between UII/UTR expression and ALF in lipopolysaccharide (LPS)/D-galactosamine (GalN)-challenged mice. Thereafter, we investigated the effects produced by the inhibition of UII/UTR system using urantide, a special antagonist of UTR, and the potential molecular mechanisms involved in ALF. Urantide was administered to mice treated with LPS/GalN. Expression of UII/UTR, releases of proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β) and interferon-γ (IFN-γ), and activation of nuclear factor κB (NF-κB) signaling pathway were assessed in the lethal ALF with or without urantide pretreatment. We found that LPS/GalN-challenged mice showed high mortality and marked hepatic inflammatory infiltration and cell apoptosis as well as a significant increase of UII/UTR expression. Urantide pretreatment protected against the injury in liver following downregulation of UII/UTR expression. A close relationship between the acutely flamed hepatic injury and UII/UTR expression was observed. In addition, urantide prevented the increases of proinflammatory cytokines such as TNF-α, IL-1β and IFN-γ, and activation of NF-κB signaling pathway induced by LPS/GalN in mice. Thus, we conclude that UII/UTR system plays a role in LPS/GalN-induced ALF. Urantide has a protective effect on the acutely inflamed injury of liver in part through preventing releases of proinflammatory cytokines and activation of NF-κB pathway.
Collapse
Affiliation(s)
- Dong-yu Liang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Liang-ming Liu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- * E-mail:
| | - Chang-gen Ye
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Liang Zhao
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Fang-ping Yu
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - De-yong Gao
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
- Department of Infection, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Ying-ying Wang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Zhi-wen Yang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| | - Yan-yan Wang
- Department of Hepatology, Songjiang Hospital Affiliated to the First People’s Hospital Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
24
|
Gong X, Zhang L, Jiang R, Wang CD, Yin XR, Wan JY. Hepatoprotective effects of syringin on fulminant hepatic failure induced by D-galactosamine and lipopolysaccharide in mice. J Appl Toxicol 2013; 34:265-71. [PMID: 23620140 DOI: 10.1002/jat.2876] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/30/2013] [Accepted: 02/12/2013] [Indexed: 12/27/2022]
Abstract
The prognosis for fulminant hepatic failure (FHF) still remains extremely poor with a high mortality and, therefore, better treatments are urgently needed. Syringin, a main active substance isolated from Eleutherococcus senticosus, has been reported to exhibit immunomodulatory and anti-inflammatory properties. In this study, we investigated the effects and underlying mechanisms of syringin on lipopolysaccharide (LPS) and D-galactosamine (D-GalN)-induced FHF in mice. Mice were administered syringin (10, 30 and 100 mg kg(-1), respectively) intraperitoneally (i.p) 30 min before LPS/D-GalN then mortality and liver injury were evaluated subsequently. We found that syringin dose-dependently attenuated LPS/D-GalN-induced FHF, as indicated by reduced mortality, inhibited aminotransferase and malondialdehyde (MDA) content, an increased glutathione (GSH) concentration and alleviated pathological liver injury. In addition, syringin inhibited LPS/D-GalN-induced hepatic caspase-3 activation and hepatocellular apoptosis, myeloperoxidase (MPO) activity and intercellular adhesion molecule-1 (ICAM-1) expression, as well as hepatic tissues tumor necrosis factor-alpha (TNF-α) production and NF-κB activation in a dose-dependent manner. These experimental data indicate that syringin might alleviate the FHF induced by LPS/D-GalN through inhibiting NF-κB activation to reduce TNF-α production.
Collapse
Affiliation(s)
- Xia Gong
- Department of Anatamy, Chongqing Medical University, Chongqing, 400016, China
| | | | | | | | | | | |
Collapse
|
25
|
Chen L, Ren F, Zhang H, Wen T, Piao Z, Zhou L, Zheng S, Zhang J, Chen Y, Han Y, Duan Z, Ma Y. Inhibition of glycogen synthase kinase 3β ameliorates D-GalN/LPS-induced liver injury by reducing endoplasmic reticulum stress-triggered apoptosis. PLoS One 2012; 7:e45202. [PMID: 23028846 PMCID: PMC3461002 DOI: 10.1371/journal.pone.0045202] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 08/17/2012] [Indexed: 01/22/2023] Open
Abstract
Background Glycogen synthase kinase 3β(GSK3β) is a ubiquitous serine-threonine protein kinase that participates in numerous cellular processes and disease pathophysiology. We aimed to determine therapeutic potential of GSK3β inhibition and its mechanism in a well-characterized model of lipopolysaccharide (LPS)-induced model of acute liver failure (ALF). Methodology In a murine ALF model induced by D-GalN(700 mg/kg)/LPS(10 µg/kg), we analyzed GSK3β mechanisms using a specific chemical inhibitor, SB216763, and detected the role of endoplasmic reticulum stress (ERS). Mice were administered SB216763 at 2 h before or after D-GalN/LPS injection, respectively, and then sacrificed 6 h after D-GalN/LPS treatment to evaluate its prophylactic and therapeutic function. The lethality rate, liver damage, ERS, cytokine expression, MAP kinase, hepatocyte apoptosis and expression of TLR 4 were evaluated, respectively. Whether the inhibition of GSK3β activation protected hepatocyte from ERS-induced apoptosis was investigated in vitro. Principal Findings GSK3β became quickly activated (dephosphorylated) upon D-GalN/LPS exposure. Administration of SB216763 not only ameliorated liver injury, as evidenced by reduced transaminase levels, and well-preserved liver architecture, but also decreased lethality. Moreover, GSK3β inhibition resulted in down-regulation of pro-apoptotic proteins C/EBP–homologous protein(CHOP) and caspase-12, which are related to ERS. To further demonstrate the role of ERS, we found that GSK3β inhibition protected hepatocyte from ERS-induced cell death. GSK3β inhibition down-regulated the MAPK pathways, reduced expression of inflammatory cytokines and decreased expression of TLR4. Conclusions Our findings demonstrate the key function of GSK3β signaling in the pathophysiology of ALF, especially in regulating the ERS, and provide a rationale for targeting GSK3β as a potential therapeutic strategy to ameliorate ALF.
Collapse
Affiliation(s)
- Liyan Chen
- The 2nd Department of Infectious Diseases, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Feng Ren
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Haiyan Zhang
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Tao Wen
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Zhengfu Piao
- Beijing Institute of Liver Diseases, Capital Medical University, Beijing, People’s Republic of China
| | - Li Zhou
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Sujun Zheng
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Jing Zhang
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yu Chen
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yuanping Han
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Zhongping Duan
- Beijing Artificial Liver Treatment & Training Center, Beijing Youan Hospital, Capital Medical University, Beijing, People’s Republic of China
| | - Yingji Ma
- The Department of Infectious Diseases, The 4th Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- * E-mail:
| |
Collapse
|
26
|
Protective effects of a novel trimerized sTNFRII on acute liver injury. Int Immunopharmacol 2012; 13:88-92. [DOI: 10.1016/j.intimp.2012.03.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 03/11/2012] [Accepted: 03/19/2012] [Indexed: 01/03/2023]
|
27
|
Jubendradass R, D’Cruz SC, Judith Amala Rani S, Mathur P. Nonylphenol induces apoptosis via mitochondria- and Fas-l-mediated pathways in the liver of adult male rat. Regul Toxicol Pharmacol 2012; 62:405-11. [DOI: 10.1016/j.yrtph.2012.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/19/2012] [Accepted: 01/19/2012] [Indexed: 11/16/2022]
|
28
|
HIV-1 coinfection and morphine coexposure severely dysregulate hepatitis C virus-induced hepatic proinflammatory cytokine release and free radical production: increased pathogenesis coincides with uncoordinated host defenses. J Virol 2011; 85:11601-14. [PMID: 21900165 DOI: 10.1128/jvi.05239-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Coinfection with human immunodeficiency virus type-1 (HIV-1) and hepatitis C virus (HCV) is a global problem that is more prevalent in injection drug users because they have a higher risk for acquiring both viruses. The roles of inflammatory cytokines and oxidative stress were examined in HIV-1- and HCV-coinfected human hepatic cells. Morphine (the bioactive product of heroin), HIV-1 Tat and the MN strain gp120 (gp120(MN)) proteins, and X4 HIV-1(LAI/IIIB) and R5 HIV-1(SF162) isolates were used to study the mechanisms of disease progression in HCV (JFH1)-infected Huh7.5.1 cell populations. HCV increased tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) release and augmented production of reactive oxygen species (ROS), nitric oxide (NO), and 3-nitrotyrosine (3-NT) in Huh7.5.1 cells. Morphine preferentially affected R5-tropic, but not X4-tropic, HIV-1 interactions with Huh7.5.1 cells. HIV-1 proteins or isolates increased cytokine release in HCV-infected cells, while adding morphine to coinfected cells caused complex imbalances, significantly disrupting cytokine secretion depending on the cytokine, morphine concentration, exposure duration, and particular pathogen involved. Production of ROS, NO, and 3-NT increased significantly in HCV- and HIV-1-coexposed cells while exposure to morphine further increased ROS. The proteasome inhibitor MG132 significantly decreased oxyradicals, cytokine levels, and HCV protein levels. Our findings indicate that hepatic inflammation is increased by combined exposure to HCV and HIV-1, that the ubiquitin-proteasome system and NF-κB contribute to key aspects of the response, and that morphine further exacerbates the disruption of host defenses. The results suggest that opioid abuse and HIV-1 coinfection each further accelerate HCV-mediated liver disease by dysregulating immune defenses.
Collapse
|
29
|
Kuhla A, Trieglaff C, Vollmar B. Role of age and uncoupling protein-2 in oxidative stress, RAGE/AGE interaction and inflammatory liver injury. Exp Gerontol 2011; 46:868-76. [PMID: 21820503 DOI: 10.1016/j.exger.2011.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 05/23/2011] [Accepted: 07/19/2011] [Indexed: 01/12/2023]
Abstract
The objective of this study is to clarify whether age-related oxidative stress enhances hepatic vulnerability via increased interaction of advanced glycation endproducts (AGE) with their receptor RAGE. To further address the role of uncoupling of mitochondrial respiration, mitochondrial uncoupling protein-2 wild-type (UCP2+/+) and knock out (UCP2-/-) mice were used and studied at an age of 8 (young), 38 (adult) and 76 weeks (senescent). First, we could show that UCP2 protein expression increased with age in UCP2+/+ mice. Second, in both mouse strains oxidative stress, as measured by malondialdehyde concentrations and the ratio of glutathione to glutathione disulfide, as well as hepatic RAGE expression and highly modified AGE accumulation significantly increased with age. This, however, was far more pronounced in UCP2-/- mice, in particular at the young age of 8 wk. In addition, the hepatic activity of the AGE precursor detoxifying enzyme glyoxalase-I was significantly decreased in 8 wk old UCP2-/- animals and concomitantly caused 2-fold higher levels of methylglyoxal-modified AGE in these animals. We further showed that the numbers of hepatic cells expressing sRAGE which acts as a decoy for RAGE ligands decreased with age and were markedly lower in the UCP2-/- than the UCP2+/+ mice. As a consequence, young 8 wk old UCP2-/- mice benefited from treatment with recombinant mouse RAGE to block the RAGE/AGE interaction, when challenged with galactosamine/lipopolysaccharide for the induction of acute liver injury. They showed less pronounced tissue damage and slightly lower mortality rate, while older UCP2+/+ and UCP2-/- mice revealed comparably high mortality rates and extent of liver injury, irrespective of their treatment with rRAGE. Taken together, the present study underlines the role of UCP2 in the age-related increase of oxidative stress and the oxidative stress-related RAGE/AGE interaction. In young animals, blockade of the RAGE/AGE interaction is of benefit, while in older animals, this protective effect is lost, supposedly due to the fact that with age other factors than enhanced hepatic glycation products predominantly determine liver injury and injury-related mortality rate.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, University of Rostock, Schillingallee 69a, Rostock, Germany
| | | | | |
Collapse
|
30
|
Bechmann LP, Jochum C, Kocabayoglu P, Sowa JP, Kassalik M, Gieseler RK, Saner F, Paul A, Trautwein C, Gerken G, Canbay A. Cytokeratin 18-based modification of the MELD score improves prediction of spontaneous survival after acute liver injury. J Hepatol 2010; 53:639-47. [PMID: 20630612 DOI: 10.1016/j.jhep.2010.04.029] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 04/28/2010] [Accepted: 04/29/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Predicting the probability of patients with acute liver failure (ALF) to recover spontaneously is of major clinical importance. As apoptotic and necrotic cell death are crucial in the pathogenesis of ALF, we determined whether selected cell-death markers predict outcome of patients with ALF and/or discriminate between etiologies. METHODS In a prospective study (11/2006-06/2009), 68 ALF patients were recruited consecutively. Data were collected over four weeks or until discharge, death or LTx, including CK18/M65 and M30 ELISA and glutathione S-transferase, subtype alpha. Data at date of admission and at the date of peak levels of M65 were individually analyzed and correlated with the patients' prognosis and etiology. RESULTS The predictive sensitivity of total serum M65 for lethal outcome was comparable to the Model for End-Stage Liver Disease (MELD) score at time of admission and at its peak value. In contrast, serum bilirubin levels had no prognostic value, neither at admission nor at later time points. In order to accurately predict the clinical prognosis of ALF patients, we tested a modified MELD score where CK18 M65 substituted bilirubin. This CK18/M65-based MELD score significantly better predicted the prognosis of ALF patients compared with the current MELD score or KCC. A combination of tested parameters contributed to improved discrimination of ALF etiologies by applying cell death and established laboratory parameters. CONCLUSIONS The CK18 M65-based MELD score has superior sensitivity and specifically predicts survival of ALF patients. Further prospective clinical studies could validate its potential role to predict requirement of LTx in ALF patients.
Collapse
Affiliation(s)
- Lars P Bechmann
- Dept. of Gastroenterology and Hepatology, University Hospital, 45122 Essen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lian LH, Jin X, Wu YL, Cai XF, Lee JJ, Nan JX. Hepatoprotective effects of Sedum sarmentosum on D-galactosamine/lipopolysaccharide-induced murine fulminant hepatic failure. J Pharmacol Sci 2010; 114:147-57. [PMID: 20838028 DOI: 10.1254/jphs.10045fp] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The hepatoprotective effects of sarmentosin-containing extracts of Sedum sarmentosum (SS) in D-galactosamine (D-GalN) / lipopolysaccharide (LPS)-induced fulminant hepatic failure mouse model. Pretreatment with SS markedly protected mice from lethal liver injury, which has known to be associated with an abrupt elevation of serum tumor necrosis factor (TNF)-α level. Indeed, SS significantly blocked the elevation of TNF-α and alanine aminotransferase and aspartate aminotransferase as well. SS also remarkably reduced number of apoptotic hepatocytes and DNA fragmentation in the liver, which correlated with blockade of caspase-3 activation. In addition, SS suppressed the increased expression of toll-like receptor 4 (TLR4). The activation of c-Jun NH(2)-terminal kinase, extracellular signal-regulated kinase, and p38 induced by D-GalN/LPS was also significantly suppressed by SS treatment. Furthermore, SS significantly inhibited the activation of nuclear factor-κB. In RAW 264.7 cells stimulated with LPS, TNF-α release and TLR4 expression was suppressed by SS pretreatment, which was in line with in vivo results. These findings suggested that SS prevents D-GalN/LPS-induced fulminant hepatic failure, and this protection is likely associated with its anti-apoptotic activity and the down-regulation of mitogen activated protein kinase activity associated at least in part with suppressing the transcription of LPS receptors.
Collapse
Affiliation(s)
- Li-Hua Lian
- Key Laboratory for Natural Resource of Changbai Mountain & Functional Molecules, Ministry of Education, College of Pharmacy, Yanbian University, China
| | | | | | | | | | | |
Collapse
|
32
|
Kupffer cell depletion reduces hepatic inflammation and apoptosis but decreases survival in abdominal sepsis. Eur J Gastroenterol Hepatol 2010; 22:1039-49. [PMID: 20300005 DOI: 10.1097/meg.0b013e32833847db] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVE During abdominal sepsis, the activation of hepatic Kupffer cells (KC) and its consequences are of central interest. This study evaluates the impact of selective KC depletion on hepatic microcirculation, cytokine release, and systemic alterations in the colon ascendens stent peritonitis (CASP), a model of polymicrobial abdominal sepsis. METHODS For KC depletion clodronate liposomes were injected 24 h before CASP surgery in female C57BL/6N mice. Three and 12 h after CASP, in-vivo fluorescence microscopy of the liver was performed. Analysis of hepatocellular apoptosis was conducted by immunohistochemistry. In addition, levels of tumor necrosis factor (TNF), IL-6, and IL-10 in the liver, lungs, spleen, and plasma were determined, and bacteriology and survival analysis were performed. RESULTS CASP led to significant sinusoidal perfusion failure, increased leukocyte recruitment, hepatocellular apoptosis and increased levels of TNF, IL-6, and IL-10 in the liver and plasma. KC depletion before CASP significantly reduced leukocyte recruitment to the liver and hepatocellular apoptosis. IL-10 secretion decreased dramatically in the liver and plasma of KC-depleted septic mice. In contrast, TNF levels were clearly elevated after clodronate treatment. In the lung and spleen, a compensatory upregulation of IL-10 could be detected after KC depletion. Clodronate treatment resulted in a significant reduction in survival. CONCLUSION The results indicate that KC depletion is locally protective in polymicrobial abdominal sepsis, as it reduces hepatic inflammation and apoptosis. These effects could be observed in the presence of clearly elevated TNF levels. However, the lack of IL-10 in KC-depleted mice resulted in a detrimental systemic proinflammation.
Collapse
|
33
|
Kuhla A, Hettwer C, Menger MD, Vollmar B. Oxidative stress-associated rise of hepatic protein glycation increases inflammatory liver injury in uncoupling protein-2 deficient mice. J Transl Med 2010; 90:1189-98. [PMID: 20368701 DOI: 10.1038/labinvest.2010.84] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dysfunction seems to be intrinsically involved in the pathogenesis of multiple organ failure because of enhanced production of reactive oxygen species and induction of oxidative damage. Chronic oxidative stress in turn causes an accumulation of advanced glycation end products (AGEs). To investigate whether mitochondrial dysfunction-associated oxidative stress leads to increased formation and accumulation of AGE, we studied hepatic glycation in uncoupling protein-2 (UCP2-/-) knockout mice. Using the galactosamine/lipopolysaccharide (G/L)-induced liver injury model, we further tested the hypothesis that a mitochondrial dysfunction-associated increase of hepatic glycation is causative for increased liver injury. Under baseline conditions, UCP2-/- mice showed higher malondialdehyde levels and reduced glutathione/glutathione disulfide ratios as well as significantly higher hepatic levels of AGE and hepatic expression of receptor for AGE (RAGE) when compared with UCP2+/+ mice, indicative for increased oxidative stress and hepatic glycation. Further, livers of G/L-challenged UCP2-/- mice revealed significantly more pronounced tissue injury and were found to express higher levels of AGE and RAGE compared with wild-type mice. Functional blockade of RAGE by application of recombinant RAGE significantly diminished liver damage particularly in UCP2-/- mice. This in turn increased survival from 30% in UCP2+/+ mice to 50% in UCP2-/- mice. In summary, we show for the first time that mitochondrial dysfunction-associated oxidative stress enhances hepatic protein glycation, which aggravates inflammation-induced liver injury. Targeting the AGE/RAGE interaction by the blockade of RAGE might be of therapeutic value for the oxidative stress-exposed liver.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, University of Rostock, Rostock, Germany
| | | | | | | |
Collapse
|
34
|
Manuelpillai U, Tchongue J, Lourensz D, Vaghjiani V, Samuel CS, Liu A, Williams ED, Sievert W. Transplantation of human amnion epithelial cells reduces hepatic fibrosis in immunocompetent CCl₄-treated mice. Cell Transplant 2010; 19:1157-68. [PMID: 20447339 DOI: 10.3727/096368910x504496] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chronic liver injury and inflammation lead to hepatic fibrosis, cirrhosis, and liver failure. Embryonic and mesenchymal stem cells have been shown to reduce experimental liver fibrosis but have potential limitations, including the formation of dysplastic precursors, tumors, and profibrogenic cells. Other stem-like cells may reduce hepatic inflammation and fibrosis without tumor and profibrogenic cell formation. To test this hypothesis we transplanted human amnion epithelial cells (hAEC), isolated from term delivered placenta, into immunocompetent C57/BL6 mice at week 2 of a 4-week regimen of carbon tetrachloride (CCl₄) exposure to induce liver fibrosis. Two weeks following hAEC infusion, intact cells expressing the human-specific markers inner mitochondrial membrane protein and human leukocyte antigen-G were found in mouse liver without evidence of host rejection of the transplanted cells. Human albumin, known to be produced by hAEC, was detected in sera of hAEC-treated mice. Human DNA was detected in mouse liver and also spleen, lungs, and heart of some animals. Following hAEC transplantation, CCl₄-treated animals showed decreased serum ALT levels and reduced hepatocyte apoptosis, compared to controls. hAEC-treated mouse liver had lower TNF-α and IL-6 protein levels and higher IL-10 compared to animals given CCl₄ alone. Compared to CCl₄ controls, hAEC-treated mice showed fewer activated collagen-producing hepatic stellate cells and less fibrosis area and collagen content. Reduced hepatic TGF-β levels in conjunction with a twofold increase in the active form of the collagen-degrading enzyme matrix metalloproteinase-2 in hAEC-treated mice compared to CCl₄ controls may account for the reduction in fibrosis. hAEC transplantation into immunocompetent mice leads to cell engraftment, reduced hepatocyte apoptosis, and decreased hepatic inflammation and fibrosis.
Collapse
Affiliation(s)
- Ursula Manuelpillai
- Centre for Reproduction & Development, Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Palmatine attenuates d-galactosamine/lipopolysaccharide-induced fulminant hepatic failure in mice. Food Chem Toxicol 2010; 48:222-8. [DOI: 10.1016/j.fct.2009.10.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Revised: 09/08/2009] [Accepted: 10/01/2009] [Indexed: 11/20/2022]
|
36
|
Michalopoulos GK. Liver regeneration after partial hepatectomy: critical analysis of mechanistic dilemmas. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 176:2-13. [PMID: 20019184 DOI: 10.2353/ajpath.2010.090675] [Citation(s) in RCA: 548] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver regeneration after partial hepatectomy is one of the most studied models of cell, organ, and tissue regeneration. The complexity of the signaling pathways initiating and terminating this process have provided paradigms for regenerative medicine. Many aspects of the signaling mechanisms involved in hepatic regeneration are under active investigation. The purpose of this review is to focus on the areas still not well understood. The review also aims to provide insights into the ways by which current concepts of liver regeneration can provide understanding regarding malfunction of the regenerative process in liver diseases, such as acute liver failure.
Collapse
Affiliation(s)
- George K Michalopoulos
- University of Pittsburgh, Department of Pathology, School of Medicine, S-410 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| |
Collapse
|
37
|
Tuñón MJ, Alvarez M, Culebras JM, González-Gallego J. An overview of animal models for investigating the pathogenesis and therapeutic strategies in acute hepatic failure. World J Gastroenterol 2009; 15:3086-98. [PMID: 19575487 PMCID: PMC2705730 DOI: 10.3748/wjg.15.3086] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Revised: 05/23/2009] [Accepted: 05/30/2009] [Indexed: 02/06/2023] Open
Abstract
Acute hepatic failure (AHF) is a severe liver injury accompanied by hepatic encephalopathy which causes multiorgan failure with an extremely high mortality rate, even if intensive care is provided. Management of severe AHF continues to be one of the most challenging problems in clinical medicine. Liver transplantation has been shown to be the most effective therapy, but the procedure is limited by shortage of donor organs. Although a number of clinical trials testing different liver assist devices are under way, these systems alone have no significant effect on patient survival and are only regarded as a useful approach to bridge patients with AHF to liver transplantation. As a result, reproducible experimental animal models resembling the clinical conditions are still needed. The three main approaches used to create an animal model for AHF are: surgical procedures, toxic liver injury and infective procedures. Most common models are based on surgical techniques (total/partial hepatectomy, complete/transient devascularization) or the use of hepatotoxic drugs (acetaminophen, galactosamine, thioacetamide, and others), and very few satisfactory viral models are available. We have recently developed a viral model of AHF by means of the inoculation of rabbits with the virus of rabbit hemorrhagic disease. This model displays biochemical and histological characteristics, and clinical features that resemble those in human AHF. In the present article an overview is given of the most widely used animal models of AHF, and their main advantages and disadvantages are reviewed.
Collapse
|
38
|
Kuhla A, Eipel C, Abshagen K, Siebert N, Menger MD, Vollmar B. Role of the perforin/granzyme cell death pathway in D-Gal/LPS-induced inflammatory liver injury. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1069-76. [PMID: 19264954 DOI: 10.1152/ajpgi.90689.2008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cytotoxic T lymphocytes and their granule components, such as perforin and granzyme, play an important role in the defense of hepatic infections caused by different pathogens. Moreover, it has been shown in vitro that hepatocytes can initiate cell death via a perforin-dependent mechanism. Although it is well known that hepatocellular apoptosis in D-galactosamine/lipopolysaccharide (D-Gal/LPS)-associated liver failure is mediated by TNF-alpha-dependent Fas/FasL cytotoxicity, there is no information on the role of perforin-mediated mechanisms in vivo. Therefore, we studied whether the cytolytic perforin/granzyme pathway contributes to the D-Gal/LPS-associated hepatotoxicity. Perforin knockout (Pko) mice showed significantly higher hepatic TNF-alpha and IL-6 mRNA expression as well as plasma TNF-alpha and IL-6 concentrations within the first hour upon D-Gal/LPS challenge compared with perforin wild-type (Pwt) mice. At 6 h upon D-Gal/LPS challenge, Pko mice further presented with higher transaminase release and onconecrotic tissue damage, whereas hepatocellular apoptosis and caspase-3 cleavage remained unaffected by the perforin deficiency. Pretreatment with a recombinant human TNF-alpha receptor fusion protein attenuated necrotic and apoptotic tissue damage and reduced plasma transaminase activities as well as cytokine release, thereby preventing acute liver failure in Pko mice as effectively as in Pwt mice. These data do not only confirm the significance of TNF-alpha as distal mediator of hepatic injury in this model but simultaneously reveal a contribution of a perforin-dependent immunoregulation, limiting the D-Gal/LPS-induced overwhelming cytokine release and onconecrotic tissue injury.
Collapse
Affiliation(s)
- Angela Kuhla
- Institute for Experimental Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | |
Collapse
|
39
|
Both tacrolimus and sirolimus decrease Th1/Th2 ratio, and increase regulatory T lymphocytes in the liver after ischemia/reperfusion. J Transl Med 2009; 89:433-45. [PMID: 19188906 DOI: 10.1038/labinvest.2009.3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The protective effects of immunosuppressants against ischemia/reperfusion (I/R) injury have been attributed to their non-specific anti-inflammatory effect. However, these effects may also depend on their effect on T lymphocytes, which are increasingly considered to be key players in I/R. Here, we studied the effects of tacrolimus and sirolimus on lymphocyte subpopulations in an I/R rat model. The animals were treated with tacrolimus, sirolimus or vehicle, before undergoing a 60-min ischemia event of the right hepatic lobe, followed by excision of the remaining liver. After 2 h, I/R rats showed increased mortality, plasma lactate dehydrogenase (LDH) levels, hepatocyte apoptosis, liver histological injury and parenchymal infiltration by neutrophils, macrophages, NK cells and T lymphocytes. Most of the changes were antagonized by both immunosuppressants. Tacrolimus augmented the proportion of cycling cells in I/R rats, whereas sirolimus showed the opposite effect. The increased Th1/Th2 ratio found in I/R livers after 2 h was reverted by immunosuppressants, which also amplified the proportion of CD4(+)CD25(+)Foxp3(+) regulatory T lymphocytes at 24 h. The protective effects of both tacrolimus and sirolimus correlated well with a decreased ratio of proinflammatory to anti-inflammatory T lymphocytes, and with an increase in the Treg proportion. This suggests a new mechanism to explain the known beneficial effect shown by immunosuppressants early after I/R.
Collapse
|
40
|
Yan BZ, Wang W, Chen LY, Bi MR, Lu YJ, Li BX, Yang BS. Role of cathepsin B-mediated apoptosis in fulminant hepatic failure in mice. World J Gastroenterol 2009; 15:1231-6. [PMID: 19291823 PMCID: PMC2658851 DOI: 10.3748/wjg.15.1231] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the pathogenic role of cathepsin B and the protective effect of a cathepsin B inhibitor (CA-074Me) in fulminant hepatic failure in mice.
METHODS: LPS/D-Gal N was injected into mice of the model group to induce fulminant hepatic failure; the protected group was administered CA-074me for 30 min before LPS/D-Gal N treatment; the normal group was given isochoric physiologic saline. Liver tissue histopathology was determined with HE at 2, 4, 6 and 8 h after Lps/D-Gal injection. Hepatocyte apoptosis was examined by TUNEL method. The expression of cathepsin B in liver tissues was investigated by immunohistochemistry, Western blot and RT-PCR.
RESULTS: Compared with the normal group, massive typical hepatocyte apoptosis occurred in the model group; the number of apoptotic cells reached a maximum 6 h after injection. The apoptosis index (AI) in the protected group was clearly reduced (30.4 ± 2.8 vs 18.1 ± 2.0, P < 0.01 ). Cathepsin B activity was markedly increased in drug-treated mice compared with the normal group (P < 0.01). Incubation with LPS/D-Gal N at selected time points resulted in a time-dependent increase in cathepsin B activity, and reached a maximum by 8 h. The expression of cathepsin B was significantly decreased in the protected group (P < 0.01).
CONCLUSION: Cathepsin B plays an essential role in the pathogenesis of fulminant hepatic failure, and the cathepsin B inhibitor CA-074me can attenuate apoptosis and liver injury.
Collapse
|