1
|
Hu P, Shan X, Dong H, Yu S, Wang B, Xiong H, Ji Z, Jing W, Cui Y, Li Z, Zhou Y, Wang Z, Wang J, Tang J, Wang T, Xie K, Yu Q. Macrophage-specific PHGDH protects against MAFLD by suppressing TAK1. Cell Rep 2025; 44:115426. [PMID: 40096087 DOI: 10.1016/j.celrep.2025.115426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/26/2025] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a progressive disease with only one approved treatment currently available. Hepatic phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme of the serine biosynthesis pathway, regulates MAFLD development. However, the role of macrophage PHGDH in MAFLD progression remains unclear. Here, we demonstrate that the lipotoxicity inducer palmitic acid (PA) significantly increases macrophage PHGDH expression and that PHGDH deficiency in macrophages promotes PA-induced inflammatory responses. Myeloid-specific PHGDH deficiency exacerbates MAFLD in mice. Mechanistically, tetrameric PHGDH binds to transforming growth factor-β-activated kinase 1 (TAK1) to inhibit its interaction with TAK1 binding protein 1 (TAB1), sequentially suppressing the activation of TAK1 and downstream NF-κB and MAPK signaling. Inhibition of TAK1 activation slows the development of metabolic dysfunction-associated steatohepatitis (MASH) caused by myeloid PHGDH knockout. Importantly, adeno-associated virus-mediated PHGDH overexpression in liver macrophages alleviates MAFLD in mice. Collectively, these results identify macrophage PHGDH as a promising therapeutic agent for MAFLD.
Collapse
Affiliation(s)
- Penghui Hu
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiao Shan
- Department of Health Management Center and Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Hongyuan Dong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Sujun Yu
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Baochen Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hui Xiong
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zemin Ji
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Weijia Jing
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yan Cui
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zihan Li
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yanzhao Zhou
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China; Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou 450008, China
| | - Zhe Wang
- Department of Health Management Center and Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Jinrong Wang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jiuzhou Tang
- Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Ting Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin 300070, China
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Qiujing Yu
- Department of Health Management Center and Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610000, China; Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
2
|
Liu X, Shi J, Wu M, Gao J, Zhang Y, Guo W, Zhang S. Betaine-homocysteine methyltransferase attenuates liver ischemia-reperfusion injury by targeting TAK1. FASEB J 2025; 39:e70349. [PMID: 39854060 DOI: 10.1096/fj.202402239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/26/2025]
Abstract
Liver ischemia-reperfusion (IR) injury is a common complication following liver surgery, significantly impacting the prognosis of liver transplantation and other liver surgeries. Betaine-homocysteine methyltransferase (BHMT), a crucial enzyme in the methionine cycle, has been previously confirmed the pivotal role in hepatocellular carcinoma, and it has also been demonstrated that BHMT inhibits inflammation, apoptosis, but its role in liver IR injury remains unknow. Following I/R injury, we found that BHMT expression was significantly upregulated in human liver transplant specimens, mice and hepatocytes. Utilizing BHMT knockout mice, we established an in vivo model of liver IR injury, and with BHMT knockout and overexpression AML12 cell lines, we created an in vitro hypoxia-reoxygenation model. Our findings reveal that BHMT deficiency exacerbates liver IR injury, leading to increased reactive oxygen species, apoptosis and inflammation, whereas BHMT overexpression mitigates these effects. We observed that BHMT inhibits the c-Jun N-terminal kinase (JNK)/p38 signaling pathway in liver IR injury by interacting with TAK1 and inhibiting its activity. The application of 5z-7-ox, a TAK1 inhibitor, reversed the worsening of liver IR injury and the activation of the JNK/p38 pathway associated with BHMT deficiency. These results demonstrate that BHMT protects against liver IR injury by targeting TAK1 and inhibiting the JNK/p38 signaling pathway. Our findings suggest that BHMT may be a promising therapeutic target for preventing liver IR injury.
Collapse
Affiliation(s)
- Xudong Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| | - Jihua Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| | - Min Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| | - Jie Gao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| | - Yi Zhang
- Department of Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Province, Zhengzhou, China
| |
Collapse
|
3
|
Shan H, Wang Z, Chen Y, Ma TF, Zhang J, Zhang J, Cheng Z, Jia L. Etomidate Inhibits Hepatic Ischemia-Reperfusion Injury Depending on the Activation of Nrf2-HO-1 Signaling Pathway. DNA Cell Biol 2025; 44:13-24. [PMID: 39470379 DOI: 10.1089/dna.2024.0125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024] Open
Abstract
Hepatic ischemia-reperfusion (I/R) injury (HIRI) is recognized as a local aseptic inflammatory response driven by innate immunity and is considered a leading cause of early organ dysfunction and failure following liver transplantation. Etomidate (Eto), an anesthetic drug known for its ability to inhibit inflammatory response and apoptosis, was the focus of our investigation. In this study, we conducted hepatic I/R surgery in vivo on C57 mice, analyzing liver damage through histopathology. Additionally, primary hepatocytes isolated from mice were cultured and subjected to hypoxia/reoxygenation (H/R) insult in vitro, with cell activity assessed using the CCK8 assay and immunofluorescence staining employed to analyze liver inflammatory cell infiltration and apoptosis. Results showed that Eto effectively inhibited liver injury, inflammatory response, and apoptosis induced by HIRI surgery, with the greatest effect observed at an Eto concentration of 10 mg/kg. Furthermore, Eto also showed the ability to inhibit H/R-induced cell damage, inflammatory activation, and apoptosis in primary hepatocytes. Further mechanistic studies revealed that Eto could promote the activation of the Nrf2-HO-1 signaling pathway, and the protective effect of Eto on HIRI was nullified when the Nrf2 inhibitor ML385 was utilized. This study highlights the potential of Eto to protect against HIRI by promoting the Nrf2-HO-1 signaling axis.
Collapse
Affiliation(s)
- Huajing Shan
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China
| | - Zhifang Wang
- Department of Nephrology, Huanggang Central Hospital, Huanggang, China
| | - Yun Chen
- Clinical Trial Centers, Huanggang Central Hospital, Huanggang, China
| | - Teng-Fei Ma
- Clinical Trial Centers, Huanggang Central Hospital, Huanggang, China
- Huanggang Institute of Translational Medicine, Huanggang, China
| | - Jianqing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinpeng Zhang
- Department of Critical Care Medicine, Huanggang Central Hospital, Huanggang, China
| | - Zhonghua Cheng
- Department of Orthopedics, Huanggang Central Hospital, Huanggang, China
| | - Liping Jia
- Respiratory and Critical Care Medicine, Huanggang Central Hospital, Huanggang, China
| |
Collapse
|
4
|
de Farias JB, de Lima Vitorino ML, Martins Esteves FA, da Fonseca Lima EJ, da Silva RA, de Lima Filho JL. Label-Free Proteomics of Severe Acute Hepatitis of Unknown Origin in Children by High-Resolution Mass Spectrometry. ACS OMEGA 2024; 9:50685-50694. [PMID: 39741806 PMCID: PMC11684528 DOI: 10.1021/acsomega.4c08745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/17/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
Acute hepatitis of unknown etiology (non-HepA-E hepatitis) emerged affecting children in 2021 and in parallel with the COVID-19 pandemic. In the present article, we performed an analysis between two plasma samples from pediatric patients, one with non-HepA-E hepatitis and the other healthy, to evaluate possible proteomic alterations associated with viral targets as possible causative agents and pathophysiological processes using the high-resolution and label-free LC-MS/MS technique. We identified 72 altered differentially expressed proteins, 45 upregulated and 27 downregulated. Gremlin-1, a protein associated with tissue fibrosis, was detected exclusively in the positive sample. Proteins involved in immunological processes, coagulation cascade, complement cascade, lipid transport, oxidative stress, acute inflammatory response, and those related to extracellular matrix deposition were also identified. In addition, some proteins of viral origin were detected, mainly from respiratory viruses. Proteomic studies of diseases such as hepatitis and other hepatopathologies have become essential for understanding pathophysiological processes and detecting molecular triggers.
Collapse
Affiliation(s)
- Josivan Barbosa de Farias
- Universidade
Federal de Pernambuco—Instituto Keizo Asami iLIKA. Av. Prof.
Moraes Rego, 1235-Cidade
Universitária, 50670-901 Recife, Pernambuco, Brazil
| | - Maria Luiza de Lima Vitorino
- Universidade
Federal de Pernambuco—Instituto Keizo Asami iLIKA. Av. Prof.
Moraes Rego, 1235-Cidade
Universitária, 50670-901 Recife, Pernambuco, Brazil
| | | | - Eduardo Jorge da Fonseca Lima
- IMIP
Hospital—Instituto de Medicina Integral Professor Fernando
Figueira. Rua dos Coelhos, 300-Boa Vista, 50070-902 Recife, Pernambuco, Brazil
| | - Roberto Afonso da Silva
- Universidade
Federal de Pernambuco—Instituto Keizo Asami iLIKA. Av. Prof.
Moraes Rego, 1235-Cidade
Universitária, 50670-901 Recife, Pernambuco, Brazil
| | - José Luiz de Lima Filho
- Universidade
Federal de Pernambuco—Instituto Keizo Asami iLIKA. Av. Prof.
Moraes Rego, 1235-Cidade
Universitária, 50670-901 Recife, Pernambuco, Brazil
| |
Collapse
|
5
|
Wang P, Xiang M, Zhu L, Zhang R, Zheng X, Zheng Z, Li K. ALKBH5 Protects Against Hepatic Ischemia-Reperfusion Injury by Regulating YTHDF1-Mediated YAP Expression. Int J Mol Sci 2024; 25:11537. [PMID: 39519091 PMCID: PMC11546256 DOI: 10.3390/ijms252111537] [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: 08/01/2024] [Revised: 10/17/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemia/reperfusion (I/R) injury with severe cell death is a major complication involved in liver transplantation and resection. The identification of key regulators improving hepatocyte activity may provide potential strategies to clinically resolve I/R-induced injury. N6-methyladenosine (m6A) RNA modification is essential for tissue homeostasis and pathogenesis. However, the potential involvement of m6A in the regulation of hepatocyte activity and liver injury has not been fully explored. In the present study, we found that hepatocyte AlkB homolog H5 (ALKBH5) levels were decreased both in vivo and in vitro I/R models. Hepatocyte-specific ALKBH5 overexpression effectively attenuated I/R-induced liver necrosis and improved cell proliferation in mice. Mechanistically, ALKBH5-mediated m6A demethylation improved the mRNA stability of YTH N6-methyladenosine RNA-binding protein 1 (YTHDF1), thereby increasing its expression, which consequently promoted the translation of Yes-associated protein (YAP). In conclusion, ALKBH5 is a regulator of hepatic I/R injury that improves hepatocyte repair and proliferation by maintaining YTHDF1 stability and YAP content. The ALKBH5-m6A-YTHDF1-YAP axis represents promising therapeutic targets for hepatic I/R injury to improve the prognosis of liver surgery.
Collapse
Affiliation(s)
- Pixiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Mei Xiang
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China;
| | - Ling Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Rixin Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Xiaolin Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Zhi Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Kai Li
- Department of Hepatobiliary and Pancreatic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| |
Collapse
|
6
|
Xie L, Xue F, Cheng C, Sui W, Zhang J, Meng L, Lu Y, Xiong W, Bu P, Xu F, Yu X, Xi B, Zhong L, Yang J, Zhang C, Zhang Y. Cardiomyocyte-specific knockout of ADAM17 alleviates doxorubicin-induced cardiomyopathy via inhibiting TNFα-TRAF3-TAK1-MAPK axis. Signal Transduct Target Ther 2024; 9:273. [PMID: 39406701 PMCID: PMC11480360 DOI: 10.1038/s41392-024-01977-z] [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: 01/20/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
The pathogenesis of doxorubicin-induced cardiomyopathy remains unclear. This study was carried out to test our hypothesis that ADAM17 aggravates cardiomyocyte apoptosis induced by doxorubicin and inhibition of ADAM17 may ameliorate doxorubicin-induced cardiomyopathy. C57BL/6J mice were intraperitoneally injected with a cumulative dose of doxorubicin to induce cardiomyopathy. Cardiomyocyte-specific ADAM17-knockout (A17α-MHCKO) and ADAM17-overexpressing (AAV9-oeA17) mice were generated. In addition, RNA sequencing of the heart tissues in different mouse groups and in vitro experiments in neonatal rat cardiomyocytes (NRCMs) receiving different treatment were performed. Mouse tumor models were constructed in A17fl/fl and A17α-MHCKO mice. In addition, cardiomyocyte-specific TRAF3-knockdown and TRAF3-overexpressing mice were generated. ADAM17 expression and activity were markedly upregulated in doxorubicin-treated mouse hearts and NRCMs. A17α-MHCKO mice showed less cardiomyocyte apoptosis induced by doxorubicin than A17fl/fl mice, and cardiomyocyte ADAM17 deficiency did not affect the anti-tumor effect of doxorubicin. In contrast, AAV9-oeA17 mice exhibited markedly aggravated cardiomyocyte apoptosis relative to AAV9-oeNC mice after doxorubicin treatment. Mechanistically, doxorubicin enhanced the expression of transcription factor C/EBPβ, leading to increased expression and activity of ADAM17 in cardiomyocyte, which enhanced TNF-α shedding and upregulated the expression of TRAF3. Increased TRAF3 promoted TAK1 autophosphorylation, resulting in activated MAPKs pathway and cardiomyocyte apoptosis. ADAM17 acted as a positive regulator of cardiomyocyte apoptosis and cardiac remodeling and dysfunction induced by doxorubicin by upregulating TRAF3/TAK1/MAPKs signaling. Thus, targeting ADAM17/TRAF3/TAK1/MAPKs signaling holds a promising potential for treating doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Lin Xie
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fei Xue
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wenhai Sui
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Linlin Meng
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenjing Xiong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Peili Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng Xu
- Department of Emergency Medicine, Chest Pain Center, Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Bo Xi
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Zhong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jianmin Yang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Cheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Yun Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
7
|
Xia K, Wang T, Chen Z, Guo J, Yu B, Chen Q, Qiu T, Zhou J, Zheng S. Hepatocellular SETDB1 Regulates Hepatic Ischemia-Reperfusion Injury through Targeting Lysine Methylation of ASK1 Signal. RESEARCH (WASHINGTON, D.C.) 2023; 6:0256. [PMID: 37915765 PMCID: PMC10616969 DOI: 10.34133/research.0256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 10/07/2023] [Indexed: 11/03/2023]
Abstract
Background: Hepatic ischemia-reperfusion injury (HIRI) stands as an unavoidable complication arising from liver surgery, profoundly intertwined with its prognosis. The role of lysine methyltransferase SET domain bifurcated 1 (SETDB1) in HIRI remains elusive, despite its confirmation as a potential therapeutic target for diverse diseases. Here, we investigated the mechanism by which SETDB1 regulated HIRI. Methods: RNA sequencing data were used to identify the expression and potential targets of SETDB1 through bioinformatics analysis. To elucidate the impact of SETDB1 on HIRI, both an in vivo model of HIRI in mice and an in vitro model of hepatocyte hypoxia/reoxygenation were established. Biochemical and histological analyses were used to investigate the influence of SETDB1 on liver damage mediated by HIRI. Chromatin immunoprecipitation and coimmunoprecipitation were implemented to explore the in-depth mechanism of SETDB1 regulating HIRI. Results: We confirmed that hepatocellular SETDB1 was up-regulated during HIRI and had a close correlation with HIRI-related inflammation and apoptosis. Moreover, inhibition of SETDB1 could mitigate HIRI-induced liver damage, inflammation, and apoptosis. Through our comprehensive mechanistic investigation, we revealed that SETDB1 interacts with apoptosis-signal-regulating kinase 1 (ASK1) and facilitates the methylation of its lysine residues. Inhibition of SETDB1 resulted in reduced phosphorylation of ASK1, leading to a marked suppression of downstream c-Jun N-terminal kinase (JNK)/p38 signaling pathway activation. The therapeutic effect on inflammation and apoptosis achieved through SETDB1 inhibition was nullified by the restoration of JNK/p38 signaling activation through ASK1 overexpression. Conclusions: The findings from our study indicate that SETDB1 mediates lysine methylation of ASK1 and modulates the activation of the ASK1-JNK/p38 pathway, thus involved in HIRI-induced inflammation and apoptosis. These results suggest that SETDB1 holds promise as a potential therapeutic target for mitigating HIRI.
Collapse
Affiliation(s)
- Kang Xia
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of general surgery,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianyu Wang
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of general surgery,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongbao Chen
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiayu Guo
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Yu
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of general surgery,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi Chen
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Qiu
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Department of Urology,
Renmin Hospital of Wuhan University, Wuhan, China
| | - Shusen Zheng
- Department of Organ Transplantation,
Renmin Hospital of Wuhan University, Wuhan, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine,
Zhejiang University, Hangzhou, China
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Hangzhou, China
- Key Laboratory of Organ Transplantation, Hangzhou, Zhejiang Province, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| |
Collapse
|
8
|
Wang Y, Ke W, Gan J, Zhu H, Xie X, He G, Liu S, Huang Y, Tang H. MicroRNA-29b-3p promotes intestinal permeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway. PLoS One 2023; 18:e0287597. [PMID: 37428806 PMCID: PMC10332595 DOI: 10.1371/journal.pone.0287597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
Irritable bowel syndrome with predominant diarrhea (IBS-D) is characterized by increased intestinal permeability. Previous studies have shown that the microRNA-29 gene is involved in the regulation of intestinal permeability in patients with IBS-D. NF-κB was proved to play a key role in inflammatory response of intestine and resultant disruption of tight junction integrity, whose activity could be inhibited by TNF Receptor-Associated Factor 3 (TRAF3). However, the exact mechanism that induces increased intestinal permeability in IBS-D patients has not been clarified. In this study, we found that microRNA-29b‑3p (miR-29b-3p) was significantly upregulated, while TRAF3 was decreased and the NF-κB-MLCK pathway was activated within the colonic tissue of IBS-D patients. Subsequently, we confirmed the targeting relationship between miR-29b-3p and TRAF3 through a double-luciferase reporter assay. Lentivirus transfection of NCM460 cells with miR-29b-3p-overexpressing and -silencing vectors demonstrated that the expression of TRAF3 was negatively correlated with the level of miR-29b-3p. The NF-κB/MLCK pathway was activated in the miR-29b-3p-overexpressing group and inhibited to some extent in the miR-29b-3p-silencing group. Results in WT and miR-29 knockout mice showed that miR-29b-3p levels were increased, TRAF3 levels were decreased, and the NF-κB/MLCK signaling was activated in the WT IBS-D group as compared with the WT control group. The protein levels of TRAF3 and TJs in the miR-29b-/- IBS-D group were partially recovered and NF-κB/MLCK pathway indicators were, to a certain extent, decreased as compared with the WT IBS-D group. These results suggested that miR-29b-3p deletion enhances the TRAF3 level in IBS-D mice and alleviates the high intestinal permeability. In brief, through the analysis of intestinal tissue samples from IBS-D patients and miR-29b-/- IBS-D mice, we showed that miR-29b-3p is involved in the pathogenesis of intestinal hyperpermeability in IBS-D via targeting TRAF3 to regulate the NF-κB-MLCK signaling pathway.
Collapse
Affiliation(s)
- Yongfu Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Ke
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianfeng Gan
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - He Zhu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiangyu Xie
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guodong He
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shan Liu
- The First Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yusheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Hongmei Tang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Chen Z, Hu F, Zhang Y, Zhang L, Wang T, Kong C, Hu H, Guo J, Chen Q, Yu B, Liu Y, Zou J, Zhou J, Qiu T. Ubiquitin-specific protease 29 attenuates hepatic ischemia-reperfusion injury by mediating TGF-β-activated kinase 1 deubiquitination. Front Immunol 2023; 14:1167667. [PMID: 37304282 PMCID: PMC10250730 DOI: 10.3389/fimmu.2023.1167667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Background and aims In the course of clinical practice, hepatic ischemia/reperfusion (I/R) injury is a prevalent pathophysiological event and is caused by a combination of complex factors that involve multiple signaling pathways such as MAPK and NF-κB. USP29 is a deubiquitinating enzyme important during the development of tumors, neurological diseases, and viral immunity. However, it is unknown how USP29 contributes to hepatic I/R injury. Methods and results We systematically investigated the role of the USP29/TAK1-JNK/p38 signaling pathway in hepatic I/R injury. We first found reduced USP29 expression in both mouse hepatic I/R injury and the primary hepatocyte hypoxia-reoxygenation (H/R) models. We established USP29 full knockout mice (USP29-KO) and hepatocyte-specific USP29 transgenic mice (USP29-HTG), and we found that USP29 knockout significantly exacerbates the inflammatory infiltration and injury processes during hepatic I/R injury, whereas USP29 overexpression alleviates liver injury by decreasing the inflammatory response and inhibiting apoptosis. Mechanistically, RNA sequencing results showed the effects of USP29 on the MAPK pathway, and further studies revealed that USP29 interacts with TAK1 and inhibits its k63-linked polyubiquitination, thereby preventing the activation of TAK1 and its downstream signaling pathways. Consistently, 5z-7-Oxozeaneol, an inhibitor of TAK1, blocked the detrimental effects of USP29 knockout on H/R-induced hepatocyte injury, further confirming that USP29 plays a regulatory role in hepatic I/R injury by targeting TAK1. Conclusion Our findings imply that USP29 is a therapeutic target with promise for the management of hepatic I/R injury via TAK1-JNK/p38 pathway-dependent processes.
Collapse
Affiliation(s)
- Zhongbao Chen
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Fengjiao Hu
- Medical Science Research Centre, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yalong Zhang
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Long Zhang
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Tianyu Wang
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Chenyang Kong
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Haochong Hu
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Qi Chen
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Bo Yu
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Yiting Liu
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jilin Zou
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital, Wuhan University, Wuhan, China
- Department of Urology, Renmin Hospital, Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Liu L, Xiao F, Sun J, Wang Q, Wang A, Zhang F, Li Z, Wang X, Fang Z, Qiao Y. Hepatocyte-derived extracellular vesicles miR-122-5p promotes hepatic ischemia reperfusion injury by regulating Kupffer cell polarization. Int Immunopharmacol 2023; 119:110060. [PMID: 37044034 DOI: 10.1016/j.intimp.2023.110060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Ischemia reperfusion injury remains a major barrier to liver transplantation, especially using grafts from donation after circulatory death, and it is also a pressing issue to be solved in clinical practice. Kupffer cell polarization toward a proinflammatory M1 phenotype is an early trigger of liver ischemia-reperfusion injury. However, the molecular mechanism regulating Kupffer cell polarization has not yet been fully elucidated. We induced liver ischemia reperfusion injury in mice and obtained samples from patients undergoing liver transplantation, serum and hepatocytes-derived extracellular vesicles were isolated by differential ultracentrifugation. Kupffer cell polarization was examined by flow cytometry and immunofluorescence histochemistry. RNA-seq was conducted to detect the differentially expressed miRNAs in extracellular vesicles. The role and mechanism of exosomal miR-122-5p in liver ischemia-reperfusion injury were determined both in vitro and in vivo. We identified ischemia reperfusion induced extracellular vesicles as a major cause of hepatic inflammation and tissue damage using adoptive transfer and release inhibition. The study also demonstrated that hepatocyte-derived exosomal miR-122-5p mediates liver ischemia reperfusion injury by polarizing Kupffer cell via PPARδ down-regulation and NF-κB pathway activation using profiling and functional analysis. Moreover, inhibiting miR-122-5p with antagomir suppressed Kupffer cell M1 polarization and attenuated liver ischemia reperfusion injury. Overall, our study demonstrated that hepatocyte-derived exosomal miR-122-5p played a critical role in promoting hepatic ischemia reperfusion injury through modulating PPARδ signaling and NF-κB pathway to introduce M1 polarization of Kupffer cell. Inhibition of miR-122-5p exhibited a protective effect against liver ischemia reperfusion injury, suggesting a potential therapeutic target for liver transplantation.
Collapse
Affiliation(s)
- Long Liu
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, Zhejiang 317000, China
| | - Fei Xiao
- Department of Organ Transplantation, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Jie Sun
- Medical Records Department, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Qi Wang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Aidong Wang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| | - Fabiao Zhang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Zhu Li
- Department of Organ Transplantation, Liaocheng People's Hospital, Liaocheng, Shandong 252000, China
| | - Xuequan Wang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China
| | - Zheping Fang
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province, Zhejiang University, Linhai, Zhejiang 317000, China; Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China.
| | - Yingli Qiao
- Department of Hepatobiliary Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang 317000, China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, China.
| |
Collapse
|
11
|
Motwani H, Patel M, Nanavaty V, Dixit N, Rawal RM, Patel SK, Solanki HA. Small RNA sequencing and identification of Andrographis paniculata miRNAs with potential cross‑kingdom human gene targets. Funct Integr Genomics 2023; 23:55. [PMID: 36725761 DOI: 10.1007/s10142-023-00976-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023]
Abstract
Cross-species post-transcriptional regulatory potential of plant derived small non-coding microRNAs (miRNAs) has been well documented by plenteous studies. MicroRNAs are transferred to host cells via oral ingestion wherein they play a decisive role in regulation of host genes; thus, miRNAs have evolved as the nascent bioactive molecules imparting pharmacological values to traditionally used medicinal plants. The present study aims to investigate small RNA profiling in order to uncover the potential regulatory role of miRNAs derived from Andrographis paniculata, one of the most widely used herb by tribal communities for liver disorders and document the pharmacological properties of A. paniculata miRNAs. In this study, high-throughput sequencing method was used to generate raw data, ~ 60 million sequences were generated from A. paniculata leaves. Using computational tools and bioinformatics approach, analyses of 3,480,097 clean reads resulted in identification of 3440 known and 51 putative novel miRNAs regulating 1365 and 192 human genes respectively. Remarkably, the identified plausible novel miRNAs apa-miR-5, apa-miR-1, apa-miR-26, and apa-miR-30 are projected to target significant host genes including CDK6, IKBKB, TRAF3, CHD4, MECP2, and ADIPOQ. Subsequent annotations revealed probable involvement of the target genes in various pathways for instance p38-MAPK, AKT, AMPK, NF-Kβ, ERK, WNT signalling, MYD88 dependant cascade, and pathways in cancer. Various diseases such as human papilloma virus infection, Alzheimer's, Non-alcoholic Fatty Liver, Alcoholic liver diseases, HepatoCellular Carcinoma (HCC), and numerous other cancers were predominantly found to be linked with target genes. Our findings postulate novel interpretations regarding modulation of human transcripts by A. paniculata miRNAs and exhibit the regulation of human diseases by plant-derived miRNAs. Though our study elucidates miRNAs as novel therapeutic agents, however, experimental validations for assessment of therapeutic potential of these miRNAs are still warranted.
Collapse
Affiliation(s)
- Harsha Motwani
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad-380009, Gujarat, India
| | - Maulikkumar Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad-380009, Gujarat, India
| | - Vishal Nanavaty
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad-380009, Gujarat, India
- Neuberg Centre for Genomic Medicine, Neuberg Supratech Reference Laboratory, Ahmedabad, Gujarat, India
| | - Nandan Dixit
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad-380009, Gujarat, India
| | - Rakesh M Rawal
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad-380009, Gujarat, India
| | - Saumya K Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad-380009, Gujarat, India.
| | - Hitesh A Solanki
- Department of Botany, Bioinformatics and Climate Change Impacts Management, Gujarat University, Ahmedabad-380009, Gujarat, India.
| |
Collapse
|
12
|
Wang J, Wang X, Cao M, Zhang L, Lin J. CircUSP39/miR-362-3p/TRAF3 Axis Mediates Hypoxia/Reoxygenation-Induced Cardiomyocyte Oxidative Stress, Inflammation, and Apoptosis. Int Heart J 2023; 64:263-273. [PMID: 37005320 DOI: 10.1536/ihj.22-232] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
Accumulating evidence suggested that aberrantly regulated circular RNA (circRNA) is a critical contributor to cardiovascular diseases, including acute myocardial infarction (AMI). However, the role and molecular mechanism of circUSP39 in AMI development remain unclear.Candidate circRNAs were screened from the Gene Expression Omnibus (GEO) database (GSE160717) and analyzed using the GEO2R tool. Hypoxia/reoxygenation (H/R) -induced AC16 cells were used to investigate the function of circUSP39 in H/R injury of cardiomyocytes. Quantitative real-time PCR (qRT-PCR) was employed to test RNA levels in H/R-induced AC16 cells. Cell Counting Kit-8, enzyme-linked immunosorbent assay, flow cytometry, and western blot (WB) assay were used to determine cell viability, oxidative stress, inflammatory factor levels, and cell apoptosis. RNA immunoprecipitation, RNA pull-down, and dual-luciferase reporter assay were conducted to validate the interactions between circRNA ubiquitin-specific peptidase 39 (circUSP39), miR-362-3p, and tumor necrosis factor receptor-associated factor 3 (TRAF3).In H/R-induced AC16 cells, the expression levels of circUSP39 and TRAF3 were upregulated whereas miR-362-3p expression was downregulated. CircUSP39 silencing markedly enhanced cell viability and superoxide dismutase activity but mitigated malondialdehyde level, secretion of inflammatory factors (IL-6, TNF-α, IL-1β, and MCP-1), and cell apoptosis in H/R-induced AC16 cells. CircUSP39 expedited H/R-induced AC16 cell injury by sponging miR-362-3p to increase the expression of TRAF3.CircUSP39 could facilitate H/R-induced cardiomyocyte oxidative stress, inflammation, and apoptosis by the miR-362-3p/TRAF3 axis, elucidating that it might be a therapeutic target for AMI.
Collapse
Affiliation(s)
| | - Xuan Wang
- Department of International Medical Center, Tianjin Hospital
| | - Mingying Cao
- Department of Cardiology, Tianjin Union Medical Center
| | - Lingli Zhang
- Department of Cardiology, Tianjin Union Medical Center
| | - Jingna Lin
- Department of Endocrinology, Tianjin Union Medical Center
| |
Collapse
|
13
|
Guo J, Song Z, Yu J, Li C, Jin C, Duan W, Liu X, Liu Y, Huang S, Tuo Y, Pei F, Jian Z, Zhou P, Zheng S, Zou Z, Zhang F, Gong Q, Liang S. Hepatocyte-specific TMEM16A deficiency alleviates hepatic ischemia/reperfusion injury via suppressing GPX4-mediated ferroptosis. Cell Death Dis 2022; 13:1072. [PMID: 36572666 PMCID: PMC9792590 DOI: 10.1038/s41419-022-05518-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022]
Abstract
Ischemia/reperfusion (I/R)-induced liver injury with severe cell death is a major complication of liver transplantation. Transmembrane member 16A (TMEM16A), a component of hepatocyte Ca2+-activated chloride channel, has been implicated in a variety of liver diseases. However, its role in hepatic I/R injury remains unknown. Here, mice with hepatocyte-specific TMEM16A knockout or overexpression were generated to examine the effect of TMEM16A on hepatic I/R injury. TMEM16A expression increased in liver samples from patients and mice with I/R injury, which was correlated with liver damage progression. Hepatocyte-specific TMEM16A knockout alleviated I/R-induced liver damage in mice, ameliorating inflammation and ferroptotic cell death. However, mice with hepatic TMEM16A overexpression showed the opposite phenotype. In addition, TMEM16A ablation decreased inflammatory responses and ferroptosis in hepatocytes upon hypoxia/reoxygenation insult in vitro, whereas TMEM16A overexpression promoted the opposite effects. The ameliorating effects of TMEM16A knockout on hepatocyte inflammation and cell death were abolished by chemically induced ferroptosis, whereas chemical inhibition of ferroptosis reversed the potentiated role of TMEM16A in hepatocyte injury. Mechanistically, TMEM16A interacted with glutathione peroxidase 4 (GPX4) to induce its ubiquitination and degradation, thereby enhancing ferroptosis. Disruption of TMEM16A-GPX4 interaction abrogated the effects of TMEM16A on GPX4 ubiquitination, ferroptosis, and hepatic I/R injury. Our results demonstrate that TMEM16A exacerbates hepatic I/R injury by promoting GPX4-dependent ferroptosis. TMEM16A-GPX4 interaction and GPX4 ubiquitination are therefore indispensable for TMEM16A-regulated hepatic I/R injury, suggesting that blockades of TMEM16A-GPX4 interaction or TMEM16A inhibition in hepatocytes may represent promising therapeutic strategies for acute liver injury.
Collapse
Affiliation(s)
- Jiawei Guo
- grid.410654.20000 0000 8880 6009Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou, China
| | - Zihao Song
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jie Yu
- grid.284723.80000 0000 8877 7471Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chengyi Li
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Chenchen Jin
- grid.508040.90000 0004 9415 435XCenter for Neuro-Metabolism and Regeneration Research, The Bioland Laboratory, Guangzhou, China
| | - Wei Duan
- grid.410654.20000 0000 8880 6009Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Xiu Liu
- grid.416466.70000 0004 1757 959XDepartment of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingying Liu
- grid.413428.80000 0004 1757 8466Guangzhou Women and Children’s Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Shuai Huang
- grid.412534.5Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yonghua Tuo
- grid.412534.5Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Fei Pei
- grid.12981.330000 0001 2360 039XDepartment of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China ,Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, China
| | - Zhengyang Jian
- Center For Drug Inspection of Guizhou Medical Products Administration, Guiyang, China
| | - Pengyu Zhou
- grid.416466.70000 0004 1757 959XDepartment of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shaoyi Zheng
- grid.416466.70000 0004 1757 959XDepartment of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaowei Zou
- grid.284723.80000 0000 8877 7471Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Zhang
- grid.34477.330000000122986657Department of Radiology, University of Washington School of Medicine, Seattle, WA USA
| | - Quan Gong
- grid.410654.20000 0000 8880 6009Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Sijia Liang
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
14
|
Jiao J, Jiang Y, Qian Y, Liu G, Xu M, Wang F, Sun X, Gao Y, Su L, Shi Y, Kong X. Expression of STING Is Increased in Monocyte-Derived Macrophages and Contributes to Liver Inflammation in Hepatic Ischemia-Reperfusion Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1745-1762. [PMID: 36174680 DOI: 10.1016/j.ajpath.2022.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/25/2022] [Accepted: 09/07/2022] [Indexed: 10/14/2022]
Abstract
Ischemia/reperfusion (I/R) injury, aggravated by innate immune cell-mediated inflammatory response, is a major problem in liver transplantation. Stimulator of interferon gene (STING) is a crucial regulatory signaling molecule in the DNA-sensing pathway, and its activation can produce strong innate immunity. However, the STING-mediated innate immune pathway in hepatic I/R injury has not been fully elucidated. In this study, we first examined the STING expression changes in the liver tissues of mice after hepatic I/R injury by using quantitative polymerase chain reaction and Western blot assays. We then investigated the role of STING in I/R injury by using a murine hepatic I/R model. STING up-regulation in mouse liver tissues in response to I/R injury and STING deficiency in myeloid cells was found to significantly ameliorate I/R-induced liver injury and inflammatory responses. STING inhibitors were also able to ameliorate hepatic I/R injury. Mechanically, STING may have a protective effect on hepatic I/R injury by the inhibition of hypoxia-inducible factor-1 alpha and enhancement of phosphorylated AMP-activated protein kinase to reduce macrophage activation. These findings show the potential regulatory effects of STING in hepatic I/R and suggest a new method for clinical protection of hepatic I/R injury.
Collapse
Affiliation(s)
- Junzhe Jiao
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yiya Jiang
- Department of General Practice, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yihan Qian
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Guanjie Liu
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Min Xu
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Fang Wang
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Xuehua Sun
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Yueqiu Gao
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China
| | - Li Su
- School of Translational Medicine, Shanghai University, Shanghai, China
| | - Yanjun Shi
- Department of General Practice, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaoni Kong
- Central Laboratory, Department of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China.
| |
Collapse
|
15
|
Conde de la Rosa L, Goicoechea L, Torres S, Garcia-Ruiz C, Fernandez-Checa JC. Role of Oxidative Stress in Liver Disorders. LIVERS 2022; 2:283-314. [DOI: 10.3390/livers2040023] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Oxygen is vital for life as it is required for many different enzymatic reactions involved in intermediate metabolism and xenobiotic biotransformation. Moreover, oxygen consumption in the electron transport chain of mitochondria is used to drive the synthesis of ATP to meet the energetic demands of cells. However, toxic free radicals are generated as byproducts of molecular oxygen consumption. Oxidative stress ensues not only when the production of reactive oxygen species (ROS) exceeds the endogenous antioxidant defense mechanism of cells, but it can also occur as a consequence of an unbalance between antioxidant strategies. Given the important role of hepatocytes in the biotransformation and metabolism of xenobiotics, ROS production represents a critical event in liver physiology, and increasing evidence suggests that oxidative stress contributes to the development of many liver diseases. The present review, which is part of the special issue “Oxidant stress in Liver Diseases”, aims to provide an overview of the sources and targets of ROS in different liver diseases and highlights the pivotal role of oxidative stress in cell death. In addition, current antioxidant therapies as treatment options for such disorders and their limitations for future trial design are discussed.
Collapse
Affiliation(s)
- Laura Conde de la Rosa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Leire Goicoechea
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Sandra Torres
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
| | - Carmen Garcia-Ruiz
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - José C. Fernandez-Checa
- Department of Cell Death and Proliferation, Institute of Biomedical Research of Barcelona (IIBB), CSIC, 08036 Barcelona, Spain
- Liver Unit, Hospital Clinic i Provincial de Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBEREHD), 08036 Barcelona, Spain
- Research Center for ALPD, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
16
|
Salidroside alleviates hepatic ischemia-reperfusion injury during liver transplant in rat through regulating TLR-4/NF-κB/NLRP3 inflammatory pathway. Sci Rep 2022; 12:13973. [PMID: 35978104 PMCID: PMC9385636 DOI: 10.1038/s41598-022-18369-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/10/2022] [Indexed: 11/09/2022] Open
Abstract
Salidroside has anti-inflammatory, antioxidant and hepatoprotective properties. However, its effect on hepatic ischemia–reperfusion injury (IRI), an unavoidable side effect associated with liver transplantation, remains undefined. Here, we aimed to determine whether salidroside alleviates hepatic IRI and elucidate its potential mechanisms. We used both in vivo and in vitro assays to assess the effect and mechanisms of salidroside on hepatic IRI. Hepatic IRI rat models were pretreated with salidroside (5, 10 or 20 mg/kg/day) for 7 days following liver transplantation while hypoxia/reoxygenation (H/R) model of RAW 264.7 macrophages were pretreated with salidroside (1, 10 or 50 μM). The effect of salidroside on hepatic IRI was assessed using hematoxylin–eosin staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, qRT-PCR, immunosorbent assay and western blotting. Our in vivo assays showed that salidroside significantly reduced pathological liver damage, serum aminotransferase levels and serum levels of IL-1, IL-18 and TNF-α. Besides, salidroside reduced the expression of TLR-4/NF-κB/NLRP3 inflammatory pathway associated proteins (TLR-4, MyD88, p-IKKα, p-IKKβ, p-IKK, p-IκBα, p-P65, NLRP3, ASC, Cleaved caspase-1, IL-1β, IL-18, TNF-α and IL-6) in rats after liver transplantation. On the other hand, data from the in vitro analysis demonstrated that salidroside blocks expression of TLR-4/NF-κB/NLRP3 inflammatory pathway related proteins in the RAW264.7 cells treated with H/R. The salidroside-specific anti-inflammatory effects were partially inhibited by the TLR-4 agonist lipopolysaccharide. Taken together, our study showed that salidroside inhibits hepatic IRI following liver transplantation by modulating the TLR-4/NF-κB/NLRP3 inflammatory pathway.
Collapse
|
17
|
Tao L, Ren X, Zhai W, Chen Z. Progress and Prospects of Non-Canonical NF-κB Signaling Pathway in the Regulation of Liver Diseases. Molecules 2022; 27:molecules27134275. [PMID: 35807520 PMCID: PMC9268066 DOI: 10.3390/molecules27134275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Non-canonical nuclear factor kappa B (NF-κB) signaling pathway regulates many physiological and pathological processes, including liver homeostasis and diseases. Recent studies demonstrate that non-canonical NF-κB signaling pathway plays an essential role in hyperglycemia, non-alcoholic fatty liver disease, alcoholic liver disease, liver regeneration, liver injury, autoimmune liver disease, viral hepatitis, and hepatocellular carcinoma. Small-molecule inhibitors targeting to non-canonical NF-κB signaling pathway have been developed and shown promising results in the treatment of liver injuries. Here, the recent advances and future prospects in understanding the roles of the non-canonical NF-κB signaling pathways in the regulation of liver diseases are discussed.
Collapse
Affiliation(s)
- Li Tao
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Xiaomeng Ren
- College of Pharmaceutical and Biology Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| | - Wenhui Zhai
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| |
Collapse
|
18
|
Zhou J, Guo L, Ma T, Qiu T, Wang S, Tian S, Zhang L, Hu F, Li W, Liu Z, Hu Y, Wang T, Kong C, Yang J, Zhou J, Li H. N-acetylgalactosaminyltransferase-4 protects against hepatic ischemia/reperfusion injury by blocking apoptosis signal-regulating kinase 1 N-terminal dimerization. Hepatology 2022; 75:1446-1460. [PMID: 34662438 DOI: 10.1002/hep.32202] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND AIMS Ischemia-reperfusion (I/R) injury is an inevitable complication of liver transplantation (LT) and compromises its prognosis. Glycosyltransferases have been recognized as promising targets for disease therapy, but their roles remain open for study in hepatic I/R (HIR) injury. Here, we aim to demonstrate the exact function and molecular mechanism of a glycosyltransferase, N-acetylgalactosaminyltransferase-4 (GALNT4), in HIR injury. APPROACH AND RESULTS By an RNA-sequencing data-based correlation analysis, we found a close correlation between GALNT4 expression and HIR-related molecular events in a murine model. mRNA and protein expression of GALNT4 were markedly up-regulated upon reperfusion surgery in both clinical samples from subjects who underwent LT and in a mouse model. We found that GALNT4 deficiency significantly exacerbated I/R-induced liver damage, inflammation, and cell death, whereas GALNT4 overexpression led to the opposite phenotypes. Our in-depth mechanistic exploration clarified that GALNT4 directly binds to apoptosis signal-regulating kinase 1 (ASK1) to inhibit its N-terminal dimerization and subsequent phosphorylation, leading to a robust inactivation of downstream c-Jun N-terminal kinase (JNK)/p38 and NF-κB signaling. Intriguingly, the inhibitory capacity of GALNT4 on ASK1 activation is independent of its glycosyltransferase activity. CONCLUSIONS GALNT4 represents a promising therapeutic target for liver I/R injury and improves liver surgery prognosis by inactivating the ASK1-JNK/p38 signaling pathway.
Collapse
Affiliation(s)
- Jiangqiao Zhou
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Lina Guo
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
- Institute of Model AnimalWuhan UniversityWuhanChina
| | - Tengfei Ma
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of NeurologyHuanggang Central HospitalHuanggangChina
- Huanggang Institute of Translational MedicineHuanggangChina
| | - Tao Qiu
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Sichen Wang
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
- Institute of Model AnimalWuhan UniversityWuhanChina
| | - Song Tian
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Li Zhang
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Fengjiao Hu
- Institute of Model AnimalWuhan UniversityWuhanChina
- Medical Science Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wei Li
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhen Liu
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Yufeng Hu
- Medical Science Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Tianyu Wang
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Chenyang Kong
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
| | - Juan Yang
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Junjie Zhou
- Institute of Model AnimalWuhan UniversityWuhanChina
- Medical Science Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Hongliang Li
- Department of Organ TransplantationRenmin HospitalSchool of Basic Medical SciencesWuhan UniversityWuhanChina
- Institute of Model AnimalWuhan UniversityWuhanChina
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
- Medical Science Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| |
Collapse
|
19
|
Wang YL, Zhang Y, Cai DS. Hepatoprotective effects of sevoflurane against hepatic ischemia-reperfusion injury by regulating microRNA-124-3p-mediated TRAF3/CREB axis. Cell Death Dis 2022; 8:105. [PMID: 35260558 PMCID: PMC8904859 DOI: 10.1038/s41420-021-00784-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 11/24/2022]
Abstract
The purpose of the present study is to define the role of sevoflurane (SEV) in hepatic ischemia-reperfusion (I/R) injury as well as its underlying mechanism. Initially, hepatic I/R animal models and I/R hepatocyte models were established in C57BL/6 mice and normal mouse hepatocytes (BNL CL.2) after SEV preconditioning, respectively, followed by detection of microRNA-124-3p (miR-124-3p), TRAF3, and CREB expression by RT-qPCR and Western blot analysis. In addition, miR-124-3p, TRAF3 and CREB expression in hepatocytes was altered to identify their roles in modulating the levels of glutathione transferase (GST), aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and inflammation-related factors and hepatocyte apoptosis by ELISA and flow cytometry respectively. The effects of SEV on the miR-124-3p/TRAF3/CREB axis were also verified in vitro and in vivo. IP assay was performed to verify the effect of TRAF3 on CREB ubiquitination in BNL CL.2 cells, and the cycloheximide (CHX) intervention experiment to detect the stability of CREB protein. SEV augmented the miR-124-3p expression in I/R animal and cell models. Moreover, SEV was observed to suppress I/R-induced liver damage, GST, ALT, and AST levels, hepatocyte apoptosis and inflammation. Overexpression of miR-124-3p resulted in alleviation of hepatic I/R injury, which was countered by TRAF3 overexpression. miR-124-3p targeted TRAF3, while TRAF3 promoted CREB ubiquitination and reduced protein stability of CREB. SEV could impede I/R-induced liver damage, GST, ALT, and AST levels, hepatocyte apoptosis and inflammation via mediation of the miR-124-3p/TRAF3/CREB axis in vitro and in vivo. Collectively, SEV may upregulate miR-124-3p to inhibit TRAF3 expression, thereby reducing the ubiquitination and degradation of CREB, alleviating hepatic I/R injury.
Collapse
Affiliation(s)
- Yi-Liang Wang
- Department of Anaesthesiology, The First Hospital of China Medical University, Shenyang, 110001, PR China
| | - Ying Zhang
- Department of Thyroid and Breast Surgery, Liaoning Provincial People's Hospital, Shenyang, 110001, PR China
| | - Da-Sheng Cai
- Department of Anaesthesiology, The First Hospital of China Medical University, Shenyang, 110001, PR China.
| |
Collapse
|
20
|
Wei Q, Zhou J, Wang K, Zhang X, Chen J, Lu D, Wei X, Zheng S, Xu X. Combination of Early Allograft Dysfunction and Protein Expression Patterns Predicts Outcome of Liver Transplantation From Donation After Cardiac Death. Front Med (Lausanne) 2021; 8:775212. [PMID: 34957150 PMCID: PMC8692269 DOI: 10.3389/fmed.2021.775212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
Early allograft dysfunction (EAD) after liver transplantation (LT) accompanies poor prognosis. This study aims to explore the relationship between pretransplant intrahepatic proteins and the incidence of EAD, and the value of combined EAD and protein profiles for predicting recipient and graft survival prognosis. Liver biopsy specimens of 105 pretransplant grafts used for LT were collected and used for immunohistochemistry analysis of 5 proteins. And matched clinical data of donor, recipient, transplantation, and prognosis were analyzed. The incidence of EAD was 41.9% (44/105) in this cohort. Macrovesicular steatosis (P = 0.016), donor body mass index (P = 0.013), recipients' pretransplant serum creatinine (P = 0.036), and intrahepatic expression of heme oxygenase 1 (HO1) (P = 0.015) and tumor necrosis factor α (TNF-α) (P = 0.039) were independent predictors of EAD. Inferior graft and recipient prognosis were observed in patients who experienced EAD (P = 0.028 and 0.031) or received grafts with higher expression of sirtuin 1 (P = 0.005 and 0.013). The graft and recipient survival were worst in patients with both EAD and high expression of sirtuin 1 (P = 0.001 and 0.004). In conclusion, pretransplant intrahepatic expression of HO1 and TNF-α are associated with the incidence of EAD. The combination of EAD and EAD-unrelated proteins showed superiority in distinguishing recipients with worse prognosis.
Collapse
Affiliation(s)
- Qiang Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Junbin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Kun Wang
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuanyu Zhang
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junli Chen
- China Liver Transplant Registery, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Shusen Zheng
- Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The Center for Integrated Oncology and Precision Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Institute of Organ Transplantation, Zhejiang University, Hangzhou, China.,National Health Commission Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China.,Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Zhou J, Hu M, He M, Wang X, Sun D, Huang Y, Cheng X, Fu J, Cai J, Ma T, Tian S, Hu Y, Hu F, Liu D, He Y, Yan L, She ZG, Zhang XJ, Ji YX, Liu H, Li H, Yang H, Zhang P. TNFAIP3 Interacting Protein 3 Is an Activator of Hippo-YAP Signaling Protecting Against Hepatic Ischemia/Reperfusion Injury. Hepatology 2021; 74:2133-2153. [PMID: 34133792 DOI: 10.1002/hep.32015] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia/reperfusion (I/R) injury, a common clinical problem that occurs during liver surgical procedures, causes a large proportion of early graft failure and organ rejection cases. The identification of key regulators of hepatic I/R injury may provide potential strategies to clinically improve the prognosis of liver surgery. Here, we aimed to identify the role of tumor necrosis factor alpha-induced protein 3-interacting protein 3 (TNIP3) in hepatic I/R injury and further reveal its immanent mechanisms. APPROACH AND RESULTS In the present study, we found that hepatocyte TNIP3 was markedly up-regulated in livers of both persons and mice subjected to I/R surgery. Hepatocyte-specific Tnip3 overexpression effectively attenuated I/R-induced liver necrosis and inflammation, but improved cell proliferation in mice, whereas TNIP3 ablation largely aggravated liver injury. This inhibitory effect of TNIP3 on hepatic I/R injury was found to be dependent on significant activation of the Hippo-YAP signaling pathway. Mechanistically, TNIP3 was found to directly interact with large tumor suppressor 2 (LATS2) and promote neuronal precursor cell-expressed developmentally down-regulated 4-mediated LATS2 ubiquitination, leading to decreased Yes-associated protein (YAP) phosphorylation at serine 112 and the activated transcription of factors downstream of YAP. Notably, adeno-associated virus delivered TNIP3 expression in the liver substantially blocked I/R injury in mice. CONCLUSIONS TNIP3 is a regulator of hepatic I/R injury that alleviates cell death and inflammation by assisting ubiquitination and degradation of LATS2 and the resultant YAP activation.TNIP3 represents a promising therapeutic target for hepatic I/R injury to improve the prognosis of liver surgery.
Collapse
Affiliation(s)
- Junjie Zhou
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Manli Hu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Meiling He
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Xiaoming Wang
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Dating Sun
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yongping Huang
- Institute of Model Animal, Wuhan University, Wuhan, China
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Xu Cheng
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiajun Fu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Jie Cai
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tengfei Ma
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Song Tian
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yufeng Hu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fengjiao Hu
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Dan Liu
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yanqi He
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Lanlan Yan
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| | - Hui Liu
- Institute of Model Animal, Wuhan University, Wuhan, China
- Tongren Hospital of Wuhan University and Wuhan Third Hospital, Wuhan, China
| | - Hongliang Li
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hailong Yang
- Institute of Model Animal, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peng Zhang
- Medical Science Research Center, Zhongnan Hospital, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animal, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Gurley JM, Gmyrek GB, Hargis EA, Bishop GA, Carr DJJ, Elliott MH. The Chx10-Traf3 Knockout Mouse as a Viable Model to Study Neuronal Immune Regulation. Cells 2021; 10:cells10082068. [PMID: 34440839 PMCID: PMC8391412 DOI: 10.3390/cells10082068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022] Open
Abstract
Uncontrolled inflammation is associated with neurodegenerative conditions in central nervous system tissues, including the retina and brain. We previously found that the neural retina (NR) plays an important role in retinal immunity. Tumor necrosis factor Receptor-Associated Factor 3 (TRAF3) is a known immune regulator expressed in the retina; however, whether TRAF3 regulates retinal immunity is unknown. We have generated the first conditional NR-Traf3 knockout mouse model (Chx10-Cre/Traf3f/f) to enable studies of neuronal TRAF3 function. Here, we evaluated NR-Traf3 depletion effects on whole retinal TRAF3 protein expression, visual acuity, and retinal structure and function. Additionally, to determine if NR-Traf3 plays a role in retinal immune regulation, we used flow cytometry to assess immune cell infiltration following acute local lipopolysaccharide (LPS) administration. Our results show that TRAF3 protein is highly expressed in the NR and establish that NR-Traf3 depletion does not affect basal retinal structure or function. Importantly, NR-Traf3 promoted LPS-stimulated retinal immune infiltration. Thus, our findings propose NR-Traf3 as a positive regulator of retinal immunity. Further, the NR-Traf3 mouse provides a tool for investigations of neuronal TRAF3 as a novel potential target for therapeutic interventions aimed at suppressing retinal inflammatory disease and may also inform treatment approaches for inflammatory neurodegenerative brain conditions.
Collapse
Affiliation(s)
- Jami M. Gurley
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (G.B.G.); (E.A.H.); (D.J.J.C.); (M.H.E.)
- Correspondence:
| | - Grzegorz B. Gmyrek
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (G.B.G.); (E.A.H.); (D.J.J.C.); (M.H.E.)
| | - Elizabeth A. Hargis
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (G.B.G.); (E.A.H.); (D.J.J.C.); (M.H.E.)
| | - Gail A. Bishop
- Department of Microbiology and Immunology, University of Iowa and VAMC, Iowa City, IA 52242, USA;
| | - Daniel J. J. Carr
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (G.B.G.); (E.A.H.); (D.J.J.C.); (M.H.E.)
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Michael H. Elliott
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center (OUHSC), 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (G.B.G.); (E.A.H.); (D.J.J.C.); (M.H.E.)
| |
Collapse
|
23
|
Pu JL, Huang ZT, Luo YH, Mou T, Li TT, Li ZT, Wei XF, Wu ZJ. Fisetin mitigates hepatic ischemia-reperfusion injury by regulating GSK3β/AMPK/NLRP3 inflammasome pathway. Hepatobiliary Pancreat Dis Int 2021; 20:352-360. [PMID: 34024736 DOI: 10.1016/j.hbpd.2021.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 04/27/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND Hepatic ischemia-reperfusion (I/R) injury (IRI) represents a crucial challenge in liver transplantation. Fisetin has anti-inflammatory, anti-aging and anti-oxidative properties. This study aimed to examine whether fisetin mitigates hepatic IRI and examine its underlying mechanisms. METHODS Sham or warm hepatic I/R operated mice were pretreated with fisetin (5, 10 or 20 mg/kg). Hepatic histological assessments, TUNEL assays and serum aminotransferase measurements were performed. An in vitro hypoxia/reoxygenation (H/R) model using RAW264.7 macrophages pretreated with fisetin (2.5, 5 or 10 µmol/L) was also used. Serum and cell supernatant concentrations of interleukin-1β (IL-1β), IL-18 and tumor necrosis factor-α (TNF-α) were determined by enzyme-linked immunosorbent assay (ELISA). Protein levels of p-GSK3β, p-AMPK and NLR family pyrin domain-containing 3 (NLRP3)-associated proteins were detected by Western blotting. RESULTS Compared with the I/R group, fisetin pretreatment reduced pathological liver damage, serum aminotransferase levels, serum concentrations of IL-1β, IL-18 and TNF-α in the murine IRI model. Fisetin also reduced the expression of NLRP3 inflammasome-associated proteins (NLRP3, cleaved caspase-1, IL-1β and IL-18) in I/R-operated liver. The experiments in vitro showed that fisetin decreased the release of IL-1β, IL-18 and TNF-α, and reduced the expression of NLRP3 inflammasome-associated proteins in H/R-treated RAW264.7 cells. Moreover, fisetin increased the expressions of p-GSK3β and p-AMPK in both models, indicating that its anti-inflammatory effects were dependent on GSK3β/AMPK signaling. The anti-inflammatory effects of fisetin were partially inhibited by the AMPK specific inhibitor compound C. CONCLUSIONS Fisetin showed protective effects against hepatic IRI, countering inflammatory responses through mediating the GSK3β/AMPK/NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Jun-Liang Pu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zuo-Tian Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yun-Hai Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Tong Mou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Ting-Ting Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhong-Tang Li
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing 400016, China
| | - Xu-Fu Wei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhong-Jun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
24
|
MicroRNA MiR-27a-5p Alleviates the Cerulein-Induced Cell Apoptosis and Inflammatory Injury of AR42J Cells by Targeting Traf3 in Acute Pancreatitis. Inflammation 2021; 43:1988-1998. [PMID: 32647955 DOI: 10.1007/s10753-020-01272-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Acute pancreatitis (AP), a sudden inflammatory process of pancreas, is painful and may contribute to death. The aberrant expression of miR-27a-5p has been reported in many types of cancers and diseases including AP. Thus, it is urgent to manifest the functions and mechanism of miR-27a-5p in AP. The levels of miR-27a-5p, tumor necrosis factor (TNF) receptor-associated factor 3 (Traf3) in serum of AP patient, or cerulein-treated AR42J cells were detected by qRT-PCR. Functionally, the apoptotic rate, the protein levels of Bcl-2 and Bax, the caspase-3 activity, and the levels of IL-1β, IL-6, and TNF-α in cerulein-treated AR42J cells were measured by flow cytometry, Western blot, caspase-3 activity assay, and qRT-PCR and ELISA assay, respectively. In addition, the putative target of miR-27a-5p was predicted by TargetScan online database, and the dual luciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to verify this interaction. Cerulein-treated mouse AP model was established to explore the role of miR-27a-5p in AP in vivo. The level of miR-27a-5p was notably downregulated in AP patients and cerulein-treated AR42J cells. The functional experiments indicated that miR-27a-5p mimics attenuated the promotion effects on cell apoptosis and the inflammatory response in AR42J cells caused by cerulein. The interaction between miR-27a-5p and Traf3 was predicted by TargetScan online database and validated by dual luciferase reporter assay and RIP assay. Following qRT-PCR results exhibited that Traf3 was apparently enhanced in cerulein-treated AR42J cells. The further functional experiments disclosed that Traf3 overexpression relieved the inhibitory effects on cell apoptosis and the inflammatory response induced by miR-27a-5p mimics in cerulein-treated AR42J cells. Moreover, miR-27a-5p alleviated cerulein-induced injury in vivo. In this study, we established the cerulein-treated AR42J cells as AP model in vitro. We validated that miR-27a-5p was significantly downregulated, and Traf3 was strikingly upregulated in AP patient and/or cerulein-treated AR42J cells. The further mechanistical and functional experiments unraveled that miR-27a-5p regulated Traf3 to relieve the cerulein-induced cell apoptosis and inflammatory injury of AR42J cells. Therefore, this novel regulatory network may provide therapeutic target for AP patients.
Collapse
|
25
|
LncRNA Gm4419 Regulates Myocardial Ischemia/Reperfusion Injury Through Targeting the miR-682/TRAF3 Axis. J Cardiovasc Pharmacol 2021; 76:305-312. [PMID: 32590403 DOI: 10.1097/fjc.0000000000000867] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myocardial cell death during acute myocardial infarction occurs because of acute ischemia, persistent ischemia, reperfusion-associated injury, and the inflammatory infiltrate as a response to cell necrosis. In the present study, quantitative real-time PCR showed that lncRNA Gm4419 was highly upregulated in ischemia/reperfusion myocardial tissues and hypoxia/reoxygenation H9C2 cells, whereas miR-682 was downregulated. Knocking down Gm4419 with sh-Gm4419 resulted in the rescue of myocardial infarction and apoptosis induced by ischemia/reperfusion or hypoxia/reoxygenation. Our study further demonstrated that Gm4419 may bind with miR-682 directly. Moreover, in vitro experiments further demonstrated that miR-682 could bind to tumor necrosis factor receptor-associated factor 3 (TRAF3) directly. Most importantly, TRAF3 overexpression could counteract the effect of sh-Gm4419. Taken together, our study indicated that Gm4419 may target miR-682 via sponging to increase TRAF3 expression, thereby contributing to myocardial I/R injury. Therefore, the Gm4419/miR-682/TRAF3 axis may be an important regulatory mechanism in myocardial ischemia/reperfusion injury.
Collapse
|
26
|
Chen M, Wu HL, Wong TS, Chen B, Gong RH, Wong HLX, Xiao H, Bian Z, Kwan HY. Combination of Wogonin and Artesunate Exhibits Synergistic anti-Hepatocellular Carcinoma Effect by Increasing DNA-Damage-Inducible Alpha, Tumor Necrosis Factor α and Tumor Necrosis Factor Receptor-Associated Factor 3-mediated Apoptosis. Front Pharmacol 2021; 12:657080. [PMID: 34025421 PMCID: PMC8131852 DOI: 10.3389/fphar.2021.657080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/29/2021] [Indexed: 02/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is difficult to treat, and is the second leading cause of cancer-related death worldwide. This study aimed to examine whether combination of wogonin and artesunate exhibits synergistic anti-HCC effect. Our data show that the combination treatment exhibits synergistic effect in reducing HCC cell viability by increasing apoptosis as indicated by the elevated cleavage of caspase 8, 3 and PARP. Interestingly, PCR array and the subsequent studies indicate that the combination treatment significantly increases the expression of DNA-damage-inducible, alpha (GADD45A), tumor necrosis factor (TNFα) and TNF receptor-associated factor 3 (TRAF3). Knockdown of GADD45A, TNFα or TRAF3 abolishes the combination treatment-enhanced apoptosis and the synergistic effect in reducing HCC cell viability. In the HCC-bearing xenograft mouse models, although the combination treatment increases the activity of NFκB in the tumor tissues, it exhibits a more potent anti-HCC effect than the mono-treatment, which may due to the enhanced apoptosis as indicated by the increased expression of GADD45A, TNFα, TRAF3 and apoptotic markers. Our study clearly demonstrates that the combination of artesunate and wogonin exhibits synergistic anti-HCC effect, and support the further development of this combination as alternative therapeutics for HCC management.
Collapse
Affiliation(s)
- Minting Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hsin Ling Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Tsz Sin Wong
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Baisen Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Rui-Hong Gong
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hoi Leong Xavier Wong
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhaoxiang Bian
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hiu Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
27
|
Anti-CD321 antibody immunotherapy protects liver against ischemia and reperfusion-induced injury. Sci Rep 2021; 11:6312. [PMID: 33737554 PMCID: PMC7973783 DOI: 10.1038/s41598-021-85001-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/23/2021] [Indexed: 12/18/2022] Open
Abstract
The prognosis of the liver transplant patients was frequently deteriorated by ischemia and reperfusion injury (IRI) in the liver. Infiltration of inflammatory cells is reported to play critical roles in the pathogenesis of hepatic IRI. Although T lymphocytes, neutrophils and monocytes infiltrated into the liver underwent IRI, we found that neutrophil depletion significantly attenuated the injury and serum liver enzyme levels in a murine model. Interestingly, the expression of CD321/JAM-A/F11R, one of essential molecules for transmigration of circulating leukocytes into inflammatory tissues, was significantly augmented on hepatic sinusoid endothelium at 1 h after ischemia and maintained until 45 min after reperfusion. The intraportal administration of anti-CD321 monoclonal antibody (90G4) significantly inhibited the leukocytes infiltration after reperfusion and diminished the damage responses by hepatic IRI (serum liver enzymes, inflammatory cytokines and hepatocyte cell death). Taken together, presented results demonstrated that blockade of CD321 by 90G4 antibody significantly attenuated hepatic IRI accompanied with substantial inhibition of leukocytes infiltration, particularly inhibition of neutrophil infiltration in the early phase of reperfusion. Thus, our work offers a potent therapeutic target, CD321, for preventing liver IRI.
Collapse
|
28
|
Zhou W, Zhong Z, Lin D, Liu Z, Zhang Q, Xia H, Peng S, Liu A, Lu Z, Wang Y, Ye S, Ye Q. Hypothermic oxygenated perfusion inhibits HECTD3-mediated TRAF3 polyubiquitination to alleviate DCD liver ischemia-reperfusion injury. Cell Death Dis 2021; 12:211. [PMID: 33627626 PMCID: PMC7904838 DOI: 10.1038/s41419-021-03493-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 01/31/2023]
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable and serious clinical problem in donations after heart death (DCD) liver transplantation. Excessive sterile inflammation plays a fateful role in liver IRI. Hypothermic oxygenated perfusion (HOPE), as an emerging organ preservation technology, has a better preservation effect than cold storage (CS) for reducing liver IRI, in which regulating inflammation is one of the main mechanisms. HECTD3, a new E3 ubiquitin ligase, and TRAF3 have an essential role in inflammation. However, little is known about HECTD3 and TRAF3 in HOPE-regulated liver IRI. Here, we aimed to investigate the effects of HOPE on liver IRI in a DCD rat model and explore the roles of HECTD3 and TRAF3 in its pathogenesis. We found that HOPE significantly improved liver damage, including hepatocyte and liver sinusoidal endothelial cell injury, and reduced DCD liver inflammation. Mechanistically, both the DOC and HECT domains of HECTD3 directly interacted with TRAF3, and the catalytic Cys (C832) in the HECT domain promoted the K63-linked polyubiquitination of TRAF3 at Lys138. Further, the ubiquitinated TRAF3 at Lys138 increased oxidative stress and activated the NF-κB inflammation pathway to induce liver IRI in BRL-3A cells under hypoxia/reoxygenation conditions. Finally, we confirmed that the expression of HECTD3 and TRAF3 was obviously increased in human DCD liver transplantation specimens. Overall, these findings demonstrated that HOPE can protect against DCD liver transplantation-induced-liver IRI by reducing inflammation via HECTD3-mediated TRAF3 K63-linked polyubiquitination. Therefore, HOPE regulating the HECTD3/TRAF3 pathway is a novel target for improving IRI in DCD liver transplantation.
Collapse
Affiliation(s)
- Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Danni Lin
- The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Innovation Center for the Study of Pancreatic Diseases, Hangzhou, China
| | - Zhongzhong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qiuyan Zhang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Haoyang Xia
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Sheng Peng
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Anxiong Liu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Zhongshan Lu
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Yanfeng Wang
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Shaojun Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.
- The 3rd Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China.
| |
Collapse
|
29
|
Zhou Y, Tao T, Liu G, Gao X, Gao Y, Zhuang Z, Lu Y, Wang H, Li W, Wu L, Zhang D, Hang C. TRAF3 mediates neuronal apoptosis in early brain injury following subarachnoid hemorrhage via targeting TAK1-dependent MAPKs and NF-κB pathways. Cell Death Dis 2021; 12:10. [PMID: 33414375 PMCID: PMC7790824 DOI: 10.1038/s41419-020-03278-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
Neuronal apoptosis has an important role in early brain injury (EBI) following subarachnoid hemorrhage (SAH). TRAF3 was reported as a promising therapeutic target for stroke management, which covered several neuronal apoptosis signaling cascades. Hence, the present study is aimed to determine whether downregulation of TRAF3 could be neuroprotective in SAH-induced EBI. An in vivo SAH model in mice was established by endovascular perforation. Meanwhile, primary cultured cortical neurons of mice treated with oxygen hemoglobin were applied to mimic SAH in vitro. Our results demonstrated that TRAF3 protein expression increased and expressed in neurons both in vivo and in vitro SAH models. TRAF3 siRNA reversed neuronal loss and improved neurological deficits in SAH mice, and reduced cell death in SAH primary neurons. Mechanistically, we found that TRAF3 directly binds to TAK1 and potentiates phosphorylation and activation of TAK1, which further enhances the activation of NF-κB and MAPKs pathways to induce neuronal apoptosis. Importantly, TRAF3 expression was elevated following SAH in human brain tissue and was mainly expressed in neurons. Taken together, our study demonstrates that TRAF3 is an upstream regulator of MAPKs and NF-κB pathways in SAH-induced EBI via its interaction with and activation of TAK1. Furthermore, the TRAF3 may serve as a novel therapeutic target in SAH-induced EBI.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Guangjie Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Xuan Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Yongyue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Han Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Clinical Medical College of Southern Medical University (Guangzhou), 210008, Nanjing, Jiangsu, People's Republic of China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Lingyun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China
| | - Dingding Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China.
| | - Chunhua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Zhongshan Road 321, 210008, Nanjing, Jiangsu, People's Republic of China.
| |
Collapse
|
30
|
Immunity as Cornerstone of Non-Alcoholic Fatty Liver Disease: The Contribution of Oxidative Stress in the Disease Progression. Int J Mol Sci 2021; 22:ijms22010436. [PMID: 33406763 PMCID: PMC7795122 DOI: 10.3390/ijms22010436] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/18/2020] [Accepted: 12/30/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is considered the hepatic manifestation of metabolic syndrome and has become the major cause of chronic liver disease, especially in western countries. NAFLD encompasses a wide spectrum of hepatic histological alterations, from simple steatosis to steatohepatitis and cirrhosis with a potential development of hepatocellular carcinoma. Non-alcoholic steatohepatitis (NASH) is characterized by lobular inflammation and fibrosis. Several studies reported that insulin resistance, redox unbalance, inflammation, and lipid metabolism dysregulation are involved in NAFLD progression. However, the mechanisms beyond the evolution of simple steatosis to NASH are not clearly understood yet. Recent findings suggest that different oxidized products, such as lipids, cholesterol, aldehydes and other macromolecules could drive the inflammation onset. On the other hand, new evidence indicates innate and adaptive immunity activation as the driving force in establishing liver inflammation and fibrosis. In this review, we discuss how immunity, triggered by oxidative products and promoting in turn oxidative stress in a vicious cycle, fuels NAFLD progression. Furthermore, we explored the emerging importance of immune cell metabolism in determining inflammation, describing the potential application of trained immune discoveries in the NASH pathological context.
Collapse
|
31
|
Pan W, Wang H, Zhang X, Xu P, Wang G, Li Y, Huang K, Zhang Y, Zhao H, Du R, Huang H, Zhang X, Zhang J. miR-210 Participates in Hepatic Ischemia Reperfusion Injury by Forming a Negative Feedback Loop With SMAD4. Hepatology 2020; 72:2134-2148. [PMID: 32155285 PMCID: PMC7818437 DOI: 10.1002/hep.31221] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/21/2020] [Accepted: 03/03/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion (IR) injury is a major complication of liver transplantation, resection, and hemorrhagic shock. Hypoxia is a key pathological event associated with IR injury. MicroRNA-210 (miR-210) has been characterized as a micromanager of hypoxia pathway. However, its function and mechanism in hepatic IR injury is unknown. APPROACH AND RESULTS In this study, we found miR-210 was induced in liver tissues from patients subjected to IR-related surgeries. In a murine model of hepatic IR, the level of miR-210 was increased in hepatocytes but not in nonparenchymal cells. miR-210 deficiency remarkably alleviated liver injury, cell inflammatory responses, and cell death in a mouse hepatic IR model. In vitro, inhibition of miR-210 decreased hypoxia/reoxygenation (HR)-induced cell apoptosis of primary hepatocytes and LO2 cells, whereas overexpression of miR-210 increased cells apoptosis during HR. Mechanistically, miR-210 directly suppressed mothers against decapentaplegic homolog 4 (SMAD4) expression under normoxia and hypoxia condition by directly binding to the 3' UTR of SMAD4. The pro-apoptotic effect of miR-210 was alleviated by SMAD4, whereas short hairpin SMAD4 abrogated the anti-apoptotic role of miR-210 inhibition in primary hepatocytes. Further studies demonstrated that hypoxia-induced SMAD4 transported into nucleus, in which SMAD4 directly bound to the promoter of miR-210 and transcriptionally induced miR-210, thus forming a negative feedback loop with miR-210. CONCLUSIONS Our study implicates a crucial role of miR-210-SMAD4 interaction in hepatic IR-induced cell death and provides a promising therapeutic approach for liver IR injury.
Collapse
Affiliation(s)
- Wen‐Ming Pan
- Department of Emergency SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hui Wang
- Department of Emergency SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Medical GeneticsBasic School of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Fei Zhang
- Center for Translational MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Peng Xu
- Department of Emergency SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Guo‐Liang Wang
- Department of Hepatobiliary SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yi‐Jing Li
- Department of Medical GeneticsBasic School of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kun‐Peng Huang
- Department of Emergency SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yun‐Wei Zhang
- Department of EmergencyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huan Zhao
- College of Life SciencesWuhan UniversityWuhanChina
| | - Run‐Lei Du
- College of Life SciencesWuhan UniversityWuhanChina
| | - Hai Huang
- Department of SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Dong Zhang
- College of Life SciencesWuhan UniversityWuhanChina
- Department of SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jin‐Xiang Zhang
- Department of Emergency SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
32
|
Zhou W, Lin D, Zhong Z, Ye Q. Roles of TRAFs in Ischemia-Reperfusion Injury. Front Cell Dev Biol 2020; 8:586487. [PMID: 33224951 PMCID: PMC7674171 DOI: 10.3389/fcell.2020.586487] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor (TRAF) proteins are a family of signaling molecules that function downstream of multiple receptor signaling pathways, and they play a pivotal role in the regulation of intracellular biological progresses. These TRAF-dependent signaling pathways and physiological functions have been involved in the occurrence and progression of ischemia-reperfusion injury (IRI), which is a common pathophysiological process that occurs in a wide variety of clinical events, including ischemic shock, organ transplantation, and thrombolytic therapy, resulting in a poor prognosis and high mortality. IRI occurs in multiple organs, including liver, kidney, heart, lung, brain, intestine, and retina. In recent years, mounting compelling evidence has confirmed that the genetic alterations of TRAFs can cause subversive phenotype changes during IRI of those organs. In this review, based on current knowledge, we summarized and analyzed the regulatory effect of TRAFs on the IRI of various organs, providing clear direction and a firm theoretical basis for the development of treatment strategies to manipulate TRAF proteins or TRAF-dependent signaling pathways in IRI-related diseases.
Collapse
Affiliation(s)
- Wei Zhou
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Danni Lin
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The First Affiliated Hospital, Zhejiang University School of Medicine, Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial Key Laboratory of Pancreatic Disease, Innovation Center for the Study of Pancreatic Diseases, Hangzhou, China
| | - Zibiao Zhong
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China
| | - Qifa Ye
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Engineering Research Center of Natural Polymer-based Medical Materials in Hubei Province, Wuhan, China.,The Third Xiangya Hospital of Central South University, Research Center of National Health Ministry on Transplantation Medicine Engineering and Technology, Changsha, China
| |
Collapse
|
33
|
Jiangqiao Z, Tianyu W, Zhongbao C, Long Z, Jilin Z, Xiaoxiong M, Tao Q. Ubiquitin-Specific Peptidase 10 Protects Against Hepatic Ischaemic/Reperfusion Injury via TAK1 Signalling. Front Immunol 2020; 11:506275. [PMID: 33133065 PMCID: PMC7550542 DOI: 10.3389/fimmu.2020.506275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Ubiquitin-specific peptidase 10 (USP10) protein is a deubiquitination enzyme involved in many important biological processes. However, the function of USP10 in hepatic ischaemic/reperfusion (I/R) injury remains unknown. The aim of this study was to explore the role of USP10 in hepatic I/R injury. USP10 Heterozygote mice and primary hepatocytes were used to construct hepatic I/R models. The effect of USP10 on hepatic I/R injury was examined via pathological and molecular analyses. Our results indicated that USP10 was significantly downregulated in the livers of mice after hepatic I/R injury and in hepatocytes subjected to hypoxia/reoxygenation stimulation. USP10 Heterozygote mice exhibited exacerbated hepatic I/R injury, as evidenced by enhanced liver inflammation via the NF-κB signalling pathway and increased hepatocyte apoptosis. Additionally, USP10 overexpression inhibited hepatocyte inflammation and apoptosis in hepatic I/R injury in vitro and in vivo. Mechanistically, our study demonstrated that USP10 knockdown exerted its detrimental effects on hepatic I/R injury by inducing activation of the transforming growth factor β-activated kinase 1 (TAK1)-JNK/p38 signalling pathways. TAK1 was required for USP10 function in hepatic I/R injury as TAK1 inhibition abolished USP10 function in vitro. In conclusion, our study demonstrated that USP10 plays a protective role in hepatic I/R injury by inhibiting the activation of the TAK1-JNK/p38 signalling pathways. Modulation of USP10/TAK1 might be a promising strategy to prevent this pathological process.
Collapse
Affiliation(s)
- Zhou Jiangqiao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Wang Tianyu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Chen Zhongbao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zhang Long
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zou Jilin
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ma Xiaoxiong
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Qiu Tao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Liu E, Sun H, Wu J, Kuang Y. MiR-92b-3p regulates oxygen and glucose deprivation-reperfusion-mediated apoptosis and inflammation by targeting TRAF3 in PC12 cells. Exp Physiol 2020; 105:1792-1801. [PMID: 32818322 DOI: 10.1113/ep088708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the central question of this study? MiR-92b-3p was found to be reduced in a rat model of middle cerebral artery occlusion: what are the functions of miR-92b-3p in oxygen and glucose deprivation-reperfusion (OGD/R)? What is the main finding and its importance? MiR-92b-3p abated apoptosis, mitochondrial dysfunction and inflammation caused by OGD/R via targeting TRAF3, suggesting that miR-92b-3p may serve as a potential therapeutic target in ischaemic stroke treatment. ABSTRACT Stroke is the most common cause of human neurological disability. MiR-92b-3p has been shown to be decreased in a rat model of middle cerebral artery occlusion, but its effects in cerebral ischaemic insult are unknown. In this study, PC12 cells were exposed to oxygen and glucose deprivation-reperfusion (OGD/R) to establish cerebral ischaemic injury in vitro. Quantitative real time-PCR analysis demonstrated that OGD/R exposure led to down-regulation of miR-92b-3p and increased mRNA and protein levels of tumour necrosis factor receptor-associated factor 3 (TRAF3). Gain of miR-92b-3p expression facilitated cell survival; attenuated lactate dehydrogenase leakage, cell apoptosis, caspase 3 activity and cleaved-caspase 3 (c-caspase 3) expression; and decreased the Bax/Bcl-2 ratio. Furthermore, miR-92b-3p repressed mitochondrial membrane potential depolarization, reactive oxygen species production, cytochrome c protein expression, inflammatory cytokine production and the reduction of ATP content. MiR-92b-3p directly targeted the 3'-untranslated region of TRAF3 and decreased TRAF3 expression. Reinforced expression of TRAF3 partly abrogated the biological activity of miR-92b-3p during OGD/R. Hence, miR-92b-3p abated apoptosis, mitochondrial dysfunction and inflammatory responses induced by OGD/R by targeting TRAF3.
Collapse
Affiliation(s)
- Enyu Liu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Haodong Sun
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Jianping Wu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Yongqin Kuang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
35
|
Zhou J, Chen J, Wei Q, Saeb-Parsy K, Xu X. The Role of Ischemia/Reperfusion Injury in Early Hepatic Allograft Dysfunction. Liver Transpl 2020; 26:1034-1048. [PMID: 32294292 DOI: 10.1002/lt.25779] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/15/2020] [Accepted: 04/06/2020] [Indexed: 12/13/2022]
Abstract
Liver transplantation (LT) is the only available curative treatment for patients with end-stage liver disease. Early allograft dysfunction (EAD) is a life-threatening complication of LT and is thought to be mediated in large part through ischemia/reperfusion injury (IRI). However, the underlying mechanisms linking IRI and EAD after LT are poorly understood. Most previous studies focused on the clinical features of EAD, but basic research on the underlying mechanisms is insufficient, due, in part, to a lack of suitable animal models of EAD. There is still no consensus on definition of EAD, which hampers comparative analysis of data from different LT centers. IRI is considered as an important risk factor of EAD, which can induce both damage and adaptive responses in liver grafts. IRI and EAD are closely linked and share several common pathways. However, the underlying mechanisms remain largely unclear. Therapeutic interventions against EAD through the amelioration of IRI is a promising strategy, but most approaches are still in preclinical stages. To further study the mechanisms of EAD and promote collaborations between LT centers, optimized animal models and unified definitions of EAD are urgently needed. Because IRI and EAD are closely linked, more attention should be paid to the underlying mechanisms and the fundamental relationship between them. Ischemia/reperfusion-induced adaptive responses may play a crucial role in the prevention of EAD, and more preclinical studies and clinical trials are urgently needed to address the current limitation of available therapeutic interventions.
Collapse
Affiliation(s)
- Junbin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Health and Family Planning Commission (NHFPC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Jian Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Health and Family Planning Commission (NHFPC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Qiang Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Health and Family Planning Commission (NHFPC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, United Kingdom.,Cambridge National Institute of Health Research Biomedical research Centre, Cambridge, United Kingdom
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,National Health and Family Planning Commission (NHFPC) Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| |
Collapse
|
36
|
Li S, Zhang M, Zhang B. MTMR14 protects against hepatic ischemia-reperfusion injury through interacting with AKT signaling in vivo and in vitro. Biomed Pharmacother 2020; 129:110455. [PMID: 32768948 DOI: 10.1016/j.biopha.2020.110455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/07/2020] [Accepted: 06/23/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatic ischemia-reperfusion (IR) injury is characterized by severe inflammation and cell death. However, very few effective therapies are presently available for hepatic IR injury treatment. Here, we reported a protective function and the underlying mechanism of myotubularin-related protein 14 (MTMR14) during hepatic IR injury. Hepatocyte-specific MTMR14 knockout (HKO) and transgenic (TG) mice were subjected to hepatic IR operation to explore MTMR14 function in vivo. Primary hepatocytes isolated from MTMR14-HKO and MTMR14-TG mice were subjected to hypoxia/reoxygenation (HR) insult in vitro. We found that MTMR14 expression in liver tissues from individuals with hepatic IR was markedly decreased, and similar results were detected in mice with hepatic IR surgery. MTMR14-TG mice following hepatic IR operation had obviously ameliorated liver pathological changes, along with improved hepatic dysfunction, which was proved by the decreased serum alanine amino transferase (ALT) and aspartate amino transferase (AST) levels. MTMR14-HKO and MTMR14-TG animal models indicated that MTMR14 alleviated cell death and inflammatory response. In addition, MTMR14 inhibited nuclear transcription factor κB (NF-κB) signaling. Of note, promoting MTMR14 expression improved phosphatidylinositol 3-kinase/protein kinase-B (PI3K/AKT) pathway through a physical interaction with AKT, subsequently reducing cell death and inflammation. Therefore, MTMR14 is a protective factor during hepatic IR injury, and the MTMR14/AKT signaling is involved the pathogenesis hepatic IR injury. Improvement of this axis might be a novel therapeutic strategy for the prevention of this pathological process.
Collapse
Affiliation(s)
- Shufang Li
- Liver Department, Xi'an Hospital of Traditional Chinese Medicine, Xi'an 710021, China
| | - Meng Zhang
- Department of Vascular Surgery, Yidu Central Hospital of Weifang, Weifang 262500, China
| | - Bei Zhang
- Department of Intervention Radiology (Department of Pain), Tangdu Hospital, the Forth Military Medical University, Xi'an 710038, China.
| |
Collapse
|
37
|
Rayego-Mateos S, Morgado-Pascual JL, Valdivielso JM, Sanz AB, Bosch-Panadero E, Rodrigues-Díez RR, Egido J, Ortiz A, González-Parra E, Ruiz-Ortega M. TRAF3 Modulation: Novel Mechanism for the Anti-inflammatory Effects of the Vitamin D Receptor Agonist Paricalcitol in Renal Disease. J Am Soc Nephrol 2020; 31:2026-2042. [PMID: 32631974 DOI: 10.1681/asn.2019111206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND CKD leads to vitamin D deficiency. Treatment with vitamin D receptor agonists (VDRAs) may have nephroprotective and anti-inflammatory actions, but their mechanisms of action are poorly understood. METHODS Modulation of the noncanonical NF-κB2 pathway and its component TNF receptor-associated factor 3 (TRAF3) by the VDRA paricalcitol was studied in PBMCs from patients with ESKD, cytokine-stimulated cells, and preclinical kidney injury models. RESULTS In PBMCs isolated from patients with ESKD, TRAF3 protein levels were lower than in healthy controls. This finding was associated with evidence of noncanonical NF-κB2 activation and a proinflammatory state. However, PBMCs from patients with ESKD treated with paricalcitol did not exhibit these features. Experiments in cultured cells confirmed the link between TRAF3 and NF-κB2/inflammation. Decreased TRAF3 ubiquitination in K48-linked chains and cIAP1-TRAF3 interaction mediated the mechanisms of paricalcitol action.TRAF3 overexpression by CRISPR/Cas9 technology mimicked VDRA's effects. In a preclinical model of kidney injury, paricalcitol inhibited renal NF-κB2 activation and decreased renal inflammation. In VDR knockout mice with renal injury, paricalcitol prevented TRAF3 downregulation and NF-κB2-dependent gene upregulation, suggesting a VDR-independent anti-inflammatory effect of paricalcitol. CONCLUSIONS These data suggest the anti-inflammatory actions of paricalcitol depend on TRAF3 modulation and subsequent inhibition of the noncanonical NF-κB2 pathway, identifying a novel mechanism for VDRA's effects. Circulating TRAF3 levels could be a biomarker of renal damage associated with the inflammatory state.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain
| | - Jose Luis Morgado-Pascual
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - José Manuel Valdivielso
- Vascular and Renal Translational Research Group. Institut de Receca Biomedica de Lleida (IRBLleida), Lleida, Spain.,REDinREN (Red de Investigación Renal), Madrid, Spain
| | - Ana Belén Sanz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Enrique Bosch-Panadero
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Raúl R Rodrigues-Díez
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz.Universidad Autónoma. 28040 Madrid, Spain; Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM). 28029 Madrid, Spain
| | - Alberto Ortiz
- REDinREN (Red de Investigación Renal), Madrid, Spain.,Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Emilio González-Parra
- Laboratory of Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Molecular and Cellular Biology in Renal and Vascular Pathology, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz,Universidad autonoma de madrid, Madrid, Spain .,REDinREN (Red de Investigación Renal), Madrid, Spain
| |
Collapse
|
38
|
Xu X, Zhang Z, Lu Y, Sun Q, Liu Y, Liu Q, Tian W, Yin Y, Yu H, Sun B. ARRB1 ameliorates liver ischaemia/reperfusion injury via antagonizing TRAF6-mediated Lysine 6-linked polyubiquitination of ASK1 in hepatocytes. J Cell Mol Med 2020; 24:7814-7828. [PMID: 32445435 PMCID: PMC7348167 DOI: 10.1111/jcmm.15412] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/14/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022] Open
Abstract
Hepatic ischaemia/reperfusion (I/R) injury is a major clinical problem during liver surgical procedures, which usually lead to early transplantation failure and higher organ rejection rate, and current effective therapeutic strategies are still limited. Therefore, in‐depth exploring of the molecular mechanisms underlying liver I/R injury is key to the development of new therapeutic methods. β‐arrestins are multifunctional proteins serving as important signalling scaffolds in numerous physiopathological processes, including liver‐specific diseases. However, the role and underlying mechanism of β‐arrestins in hepatic I/R injury remain largely unknown. Here, we showed that only ARRB1, but not ARRB2, was down‐regulated during liver I/R injury. Hepatocyte‐specific overexpression of ARRB1 significantly ameliorated liver damage, as demonstrated by decreases in serum aminotransferases, hepatocellular necrosis and apoptosis, infiltrating inflammatory cells and secretion of pro‐inflammatory cytokines relative to control mice, whereas experiments with ARRB1 knockout mice gotten opposite effects. Mechanistically, ARRB1 directly interacts with ASK1 in hepatocytes and inhibits its TRAF6‐mediated Lysine 6‐linked polyubiquitination, which then prevents the activation of ASK1 and its downstream signalling pathway during hepatic I/R injury. In addition, inhibition of ASK1 remarkably abolished the disruptive effect result from ARRB1 deficiency in liver I/R injury in vivo, indicating that ASK1 was required for ARRB1 function in hepatic I/R injury. In conclusion, we proposed that ARRB1 is a novel protective regulator during liver I/R injury, and modulation of the regulatory axis between ARRB1 and ASK1 could be a novel therapeutic strategy to prevent this pathological process.
Collapse
Affiliation(s)
- Xiaoliang Xu
- School of Medicine, Southeast University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zechuan Zhang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yijun Lu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qikai Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yang Liu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiaoyu Liu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenfang Tian
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Yin
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Hailong Yu
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Beicheng Sun
- School of Medicine, Southeast University, Nanjing, China.,Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
39
|
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med 2020; 152:116-141. [PMID: 32156524 DOI: 10.1016/j.freeradbiomed.2020.02.025] [Citation(s) in RCA: 768] [Impact Index Per Article: 153.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide and is strongly associated with the presence of oxidative stress. Disturbances in lipid metabolism lead to hepatic lipid accumulation, which affects different reactive oxygen species (ROS) generators, including mitochondria, endoplasmic reticulum, and NADPH oxidase. Mitochondrial function adapts to NAFLD mainly through the downregulation of the electron transport chain (ETC) and the preserved or enhanced capacity of mitochondrial fatty acid oxidation, which stimulates ROS overproduction within different ETC components upstream of cytochrome c oxidase. However, non-ETC sources of ROS, in particular, fatty acid β-oxidation, appear to produce more ROS in hepatic metabolic diseases. Endoplasmic reticulum stress and NADPH oxidase alterations are also associated with NAFLD, but the degree of their contribution to oxidative stress in NAFLD remains unclear. Increased ROS generation induces changes in insulin sensitivity and in the expression and activity of key enzymes involved in lipid metabolism. Moreover, the interaction between redox signaling and innate immune signaling forms a complex network that regulates inflammatory responses. Based on the mechanistic view described above, this review summarizes the mechanisms that may account for the excessive production of ROS, the potential mechanistic roles of ROS that drive NAFLD progression, and therapeutic interventions that are related to oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Ruifeng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
40
|
Wu YK, Hu LF, Lou DS, Wang BC, Tan J. Targeting DUSP16/TAK1 signaling alleviates hepatic dyslipidemia and inflammation in high fat diet (HFD)-challenged mice through suppressing JNK MAPK. Biochem Biophys Res Commun 2020; 524:142-149. [PMID: 31982140 DOI: 10.1016/j.bbrc.2020.01.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/03/2020] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is featured by hepatic steatosis, insulin resistance, lipid deposition and inflammation. However, the pathogenic mechanism of NAFLD is still poorly understood. Dual-specificity phosphatase 16 (DUSP16), a c-Jun N-terminal kinase-specific phosphatase, has been reported to negatively modulate the mitogen-activated protein kinases (MAPKs) signaling, and it has never been investigated in NAFLD progression. In the study, we identified that DUSP16 could directly interact with TAK1 in human hepatocytes. DUSP16 knockdown in the isolated primary hepatocytes stimulated by palmitate (PA) showed accelerated lipid deposition and inflammatory response, along with the exacerbated activation of c-Jun NH2-terminal kinase (JNK), Transforming growth factor β (TGF-β)-activated kinase (TAK1) and nuclear factor-κB (NF-κB) signaling pathways; however, the opposite results were detected in PA-treated hepatocytes with DUSP16 over-expression. The in vivo experiments confirmed that DUSP16 knockout significantly aggravated the metabolic disorder and insulin resistance in high fat diet (HFD)-challenged mice. In addition, HFD-provoked hepatic lipid accumulation and inflammation were further promoted in mice with DUSP16 knockout through the same molecular mechanism as detected in vitro. Herein, these findings demonstrated that DUSP16 could directly interact with TAK1 and negatively regulate JNK signaling to alleviate metabolic stress-induced hepatic steatosis, and thus could be considered as a promising new molecular target for NAFLD treatment.
Collapse
Affiliation(s)
- Ye-Kuan Wu
- Postdoctoral Research Station of Biology, Chongqing University, Chongqing, 400030, China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China; Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Lin-Feng Hu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China; Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - De-Shuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China
| | - Bo-Chu Wang
- Postdoctoral Research Station of Biology, Chongqing University, Chongqing, 400030, China; Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, China.
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, PR China; Research Center of Brain Intellectual Promotion and Development for Children Aged 0-6 Years, Chongqing University of Education, Chongqing, 400067, PR China.
| |
Collapse
|
41
|
Guo W, Fang H, Cao S, Chen S, Li J, Shi J, Tang H, Zhang Y, Wen P, Zhang J, Wang Z, Shi X, Pang C, Yang H, Hu B, Zhang S. Six-Transmembrane Epithelial Antigen of the Prostate 3 Deficiency in Hepatocytes Protects the Liver Against Ischemia-Reperfusion Injury by Suppressing Transforming Growth Factor-β-Activated Kinase 1. Hepatology 2020; 71:1037-1054. [PMID: 31393024 PMCID: PMC7155030 DOI: 10.1002/hep.30882] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Hepatic ischemia-reperfusion (I/R) injury remains a major challenge affecting the morbidity and mortality of liver transplantation. Effective strategies to improve liver function after hepatic I/R injury are limited. Six-transmembrane epithelial antigen of the prostate 3 (Steap3), a key regulator of iron uptake, was reported to be involved in immunity and apoptotic processes in various cell types. However, the role of Steap3 in hepatic I/R-induced liver damage remains largely unclear. APPROACH AND RESULTS In the present study, we found that Steap3 expression was significantly up-regulated in liver tissue from mice subjected to hepatic I/R surgery and primary hepatocytes challenged with hypoxia/reoxygenation insult. Subsequently, global Steap3 knockout (Steap3-KO) mice, hepatocyte-specific Steap3 transgenic (Steap3-HTG) mice, and their corresponding controls were subjected to partial hepatic warm I/R injury. Hepatic histology, the inflammatory response, and apoptosis were monitored to assess liver damage. The molecular mechanisms of Steap3 function were explored in vivo and in vitro. The results demonstrated that, compared with control mice, Steap3-KO mice exhibited alleviated liver damage after hepatic I/R injury, as shown by smaller necrotic areas, lower serum transaminase levels, decreased apoptosis rates, and reduced inflammatory cell infiltration, whereas Steap3-HTG mice had the opposite phenotype. Further molecular experiments showed that Steap3 deficiency could inhibit transforming growth factor-β-activated kinase 1 (TAK1) activation and downstream c-Jun N-terminal kinase (JNK) and p38 signaling during hepatic I/R injury. CONCLUSIONS Steap3 is a mediator of hepatic I/R injury that functions by regulating inflammatory responses as well as apoptosis through TAK1-dependent activation of the JNK/p38 pathways. Targeting hepatocytes, Steap3 may be a promising approach to protect the liver against I/R injury.
Collapse
Affiliation(s)
- Wen‐Zhi Guo
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Hong‐Bo Fang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Sheng‐Li Cao
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - San‐Yang Chen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Jie Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Ji‐Hua Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Hong‐Wei Tang
- Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Yi Zhang
- Department of SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina
| | - Pei‐Hao Wen
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Jia‐Kai Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Zhi‐Hui Wang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Xiao‐Yi Shi
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Chun Pang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Han Yang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Bo‐Wen Hu
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| | - Shui‐Jun Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan ProvinceChina,Henan Key Laboratory of Digestive Organ TransplantationZhengzhouHenan ProvinceChina,Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan UniversitiesZhengzhouHenan ProvinceChina
| |
Collapse
|
42
|
Hepatocellular HO-1 mediated iNOS-induced hepatoprotection against liver ischemia reperfusion injury. Biochem Biophys Res Commun 2020; 521:1095-1100. [PMID: 31733834 DOI: 10.1016/j.bbrc.2019.11.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 02/06/2023]
Abstract
Hepatocyte-derived inducible nitric oxide synthase (iNOS) was proved to impart protection against liver ischemia reperfusion (I/R) injury in our prior analysis. However, the mechanism for this hepatoprotection remains incompletely understood. Bone marrow chimeric mice were generated using expression of iNOS in a hepatocyte-selective manner against an iNOS-knockout background. The function of heme oxygenase 1 (HO-1) in iNOS-stimulated hepatoprotection and the molecular mechanisms were explored in vitro and in vivo, respectively. Hepatocyte-derived iNOS conferred protection from I/R injury and anoxia/reoxygenation stimulation. Mechanistically, iNOS activated nuclear factor erythroid 2-related factor 2 (Nrf-2) and subsequently, stimulated the transcription of HO-1. Results from our study led to the conclusion that HO-1 is another potent mediator of iNOS-mediated protection after liver I/R.
Collapse
|
43
|
Zabala V, Boylan JM, Thevenot P, Frank A, Senthoor D, Iyengar V, Kim H, Cohen A, Gruppuso PA, Sanders JA. Transcriptional changes during hepatic ischemia-reperfusion in the rat. PLoS One 2019; 14:e0227038. [PMID: 31891620 PMCID: PMC6938360 DOI: 10.1371/journal.pone.0227038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
There are few effective targeted strategies to reduce hepatic ischemia-reperfusion (IR) injury, a contributor to poor outcomes in liver transplantation recipients. It has been proposed that IR injury is driven by the generation of reactive oxygen species (ROS). However, recent studies implicate other mediators of the injury response, including mitochondrial metabolic dysfunction. We examined changes in global gene expression after transient hepatic ischemia and at several early reperfusion times to identify potential targets that could be used to protect against IR injury. Male Wistar rats were subjected to 30 minutes of 70% partial warm ischemia followed by 0, 0.5, 2, or 6 hours of reperfusion. RNA was extracted from the reperfused and non-ischemic lobes at each time point for microarray analysis. Identification of differentially expressed genes and pathway analysis were used to characterize IR-induced changes in the hepatic transcriptome. Changes in the reperfused lobes were specific to the various reperfusion times. We made the unexpected observation that many of these changes were also present in tissue from the paired non-ischemic lobes. However, the earliest reperfusion time, 30 minutes, showed a marked increase in the expression of a set of immediate-early genes (c-Fos, c-Jun, Atf3, Egr1) that was exclusive to the reperfused lobe. We interpreted these results as indicating that this early response represented a tissue autonomous response to reperfusion. In contrast, the changes that occurred in both the reperfused and non-ischemic lobes were interpreted as indicating a non-autonomous response resulting from hemodynamic changes and/or circulating factors. These tissue autonomous and non-autonomous responses may serve as targets to ameliorate IR injury.
Collapse
Affiliation(s)
- Valerie Zabala
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, United States of America
- Division of Biology and Medicine, Brown University, Providence, RI, United States of America
| | - Joan M. Boylan
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, United States of America
- Division of Biology and Medicine, Brown University, Providence, RI, United States of America
| | - Paul Thevenot
- Institute of Translational Research, Ochsner Health Systems, New Orleans LA, United States of America
| | - Anderson Frank
- Institute of Translational Research, Ochsner Health Systems, New Orleans LA, United States of America
| | - Dewahar Senthoor
- Warren Alpert Medical School, Providence, RI, United States of America
| | - Varun Iyengar
- Warren Alpert Medical School, Providence, RI, United States of America
| | - Hannah Kim
- Division of Biology and Medicine, Brown University, Providence, RI, United States of America
| | - Ari Cohen
- Institute of Translational Research, Ochsner Health Systems, New Orleans LA, United States of America
| | - Philip A. Gruppuso
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, United States of America
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, United States of America
| | - Jennifer A. Sanders
- Department of Pediatrics, Rhode Island Hospital and Brown University, Providence, RI, United States of America
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States of America
| |
Collapse
|
44
|
Chen Z, Yu Y, Cai J, Li H. Emerging Molecular Targets for Treatment of Nonalcoholic Fatty Liver Disease. Trends Endocrinol Metab 2019; 30:903-914. [PMID: 31597607 DOI: 10.1016/j.tem.2019.08.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/14/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
In parallel with the obesity epidemic, nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide. Disequilibrium of lipid metabolism and the subsequent metabolic-stress-induced inflammation are believed to be central in the pathogenesis of NAFLD. Of note, metabolic inflammation is primarily mediated by innate immune signaling, which is increasingly recognized as a driving force in NAFLD progression. Currently, a series of agents targeting one or more of these pathomechanisms have shown encouraging results in preclinical models and clinical trials. This review summarizes the emerging molecular targets involved in signaling in the lipid metabolism and innate immunity aspects of NAFLD, focusing on their mechanistic roles and translational potentials.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Yao Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Model Animals of Wuhan University, Wuhan 430072, China; Basic Medical School, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
45
|
Aloin Preconditioning Attenuates Hepatic Ischemia/Reperfusion Injury via Inhibiting TLR4/MyD88/NF- κB Signal Pathway In Vivo and In Vitro. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3765898. [PMID: 31827674 PMCID: PMC6886335 DOI: 10.1155/2019/3765898] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/22/2019] [Accepted: 11/02/2019] [Indexed: 12/11/2022]
Abstract
Background Aloin exerts considerable protective effects in various disease models, and its effect on hepatic ischemia-reperfusion (HIR) injury remains unknown. This research is aimed at conducting an in-depth investigation of the antioxidant, anti-inflammatory, and antiapoptosis effects of aloin in HIR injury and explain the underlying molecular mechanisms. Methods In vivo, different concentrations of aloin were intraperitoneally injected 1 h before the establishment of the HIR model in male mice. The hepatic function, pathological status, oxidative stress, and inflammatory and apoptosis markers were measured. In vitro, aloin (AL, C21H22O9) or lipopolysaccharide (LPS) was added to a culture of mouse primary hepatocytes before it underwent hypoxia/reoxygenation (H/R), and the apoptosis in the mouse primary hepatocytes was analyzed. Results We found that 20 mg/kg was the optimum concentration of aloin for mitigating I/R-induced liver tissue damage, characterized by decreased serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST). Aloin pretreatment substantially suppressed the generation of hepatic malondialdehyde (MDA), tumor necrosis factor alpha (TNF-α), and IL-6 and enhanced the hepatic superoxide dismutase (SOD) activities as well as glutathione (GSH) and IL-10 levels in the liver tissue of I/R mice; this indicated that aloin ameliorated I/R-induced liver damage by reducing the oxidative stress and inflammatory response. Moreover, aloin inhibited hepatocyte apoptosis and inflammatory response that was caused by the upregulated expression of Bcl-2, the downregulated expression of cleaved caspase3(C-caspase3), Bax, Toll-like receptor 4 (TLR4), FADD, MyD88, TRAF6, phosphorylated IKKα/β (p-IKKα/β), and phosphorylated nuclear factor κB p65 (p-NF-κB p65).
Collapse
|
46
|
Yan ZZ, Huang YP, Wang X, Wang HP, Ren F, Tian RF, Cheng X, Cai J, Zhang Y, Zhu XY, She ZG, Zhang XJ, Huang Z, Li H. Integrated Omics Reveals Tollip as an Regulator and Therapeutic Target for Hepatic Ischemia-Reperfusion Injury in Mice. Hepatology 2019; 70:1750-1769. [PMID: 31077413 DOI: 10.1002/hep.30705] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/04/2019] [Indexed: 12/24/2022]
Abstract
Hepatic ischemia-reperfusion (IR) injury is the leading cause of liver dysfunction and failure after liver resection or transplantation and lacks effective therapeutic strategies. Here, we applied a systematic proteomic analysis to identify the prominent contributors to IR-induced liver damage and promising therapeutic targets for this condition. Based on an unbiased proteomic analysis, we found that toll-interacting protein (Tollip) expression was closely correlated with the hepatic IR process. RNA sequencing analysis and phenotypic examination showed a dramatically alleviated hepatic IR injury by Tollip deficiency both in vivo and in hepatocytes. Mechanistically, Tollip interacts with apoptosis signal-regulating kinase 1 (ASK1) and facilitates the recruitment of tumor necrosis factor receptor-associated factor 6 (TRAF6) to ASK1, leading to enhanced ASK1 N-terminal dimerization and the subsequent activation of downstream mitogen-activated protein kinase (MAPK) signaling. Furthermore, the Tollip methionine and phenylalanine motif and TRAF6 ubiquitinating activity are required for Tollip-regulated ASK1-MAPK axis activation. Conclusion: Tollip is a regulator of hepatic IR injury by facilitating ASK1 N-terminal dimerization and the resultant c-Jun N-terminal kinase/p38 signaling activation. Inhibiting Tollip or its interaction with ASK1 might be promising therapeutic strategies for hepatic IR injury.
Collapse
Affiliation(s)
- Zhen-Zhen Yan
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yong-Ping Huang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Xin Wang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hai-Ping Wang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
| | - Fei Ren
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Rui-Feng Tian
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xu Cheng
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Cai
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Yan Zhang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Zhi-Gang She
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Zan Huang
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hongliang Li
- College of Life Sciences, Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
- Institute of Model Animal of Wuhan University, Wuhan, China
- Basic Medical School, Wuhan University, Wuhan, China
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
47
|
Dai Y, Mao Z, Han X, Xu Y, Xu L, Yin L, Qi Y, Peng J. MicroRNA-29b-3p reduces intestinal ischaemia/reperfusion injury via targeting of TNF receptor-associated factor 3. Br J Pharmacol 2019; 176:3264-3278. [PMID: 31167039 PMCID: PMC6692574 DOI: 10.1111/bph.14759] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 05/07/2019] [Accepted: 05/25/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The microRNA miR-29b-3p shows important roles in regulating apoptosis and inflammation. However, its effects on intestinal ischaemia/reperfusion (II/R) injury have not been reported. Here we have investigated the functions of miR-29b-3p on II/R injury on order to find drug targets to treat the injury. EXPERIMENTAL APPROACH Two models - in vitro hypoxia/reoxygenation (H/R) of IEC-6 cells; in vivo, II/R injury in C57BL/6 mice were used. Western blotting and dual-luciferase reporter assays were used and mimic and siRNA transfection tests were applied to assess the effects of miR-29b-3p on II/R injury via targeting TNF receptor-associated factor 3 (TRAF3). KEY RESULTS The H/R procedure decreased cell viability and promoted inflammation and apoptosis in IEC-6 cells, and the II/R procedure also promoted intestinal inflammation and apoptosis in mice. Expression levels of miR-29b-3p were decreased in H/R-induced cells and II/R-induced intestinal tissues of mice compared with control group or sham group, which directly targeted TRAF3. Decreased miR-29b-3p level markedly increased TRAF3 expression via activating TGF-α-activated kinase 1 phosphorylation, increasing NF-κB (p65) levels to promote inflammation, up-regulating Bcl2-associated X expression, and down-regulating Bcl-2 expression to trigger apoptosis. In addition, the miR-29b-3p mimetic and TRAF3 siRNA in IEC-6 cells markedly suppressed apoptosis and inflammation to alleviate II/R injury via inhibiting TRAF3 signallimg. CONCLUSIONS AND IMPLICATIONS The miR-29b-3p played a critical role in II/R injury, via targeting TRAF3, which should be considered as a significant drug target to treat the disease.
Collapse
Affiliation(s)
- Yan Dai
- College of PharmacyDalian Medical UniversityDalianChina
| | - Zhang Mao
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xu Han
- College of PharmacyDalian Medical UniversityDalianChina
| | - Youwei Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lina Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lianhong Yin
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yan Qi
- College of PharmacyDalian Medical UniversityDalianChina
| | - Jinyong Peng
- College of PharmacyDalian Medical UniversityDalianChina
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning ProvinceDalian Medical UniversityDalianChina
- National‐Local Joint Engineering Research Center for Drug Development (R&D) of Neurodegenerative DiseasesDalian Medical UniversityDalianChina
| |
Collapse
|
48
|
Hu W, Guo G, Chi Y, Li F. Construction of Traf3 knockout liver cancer cell line using CRISPR/Cas9 system. J Cell Biochem 2019; 120:14908-14915. [PMID: 31016787 DOI: 10.1002/jcb.28753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE The gene editing technology in CRISPR/Cas9 system was used to construct the Traf3 knockout HepG2 cell line to explore the role of Traf3 in the development of liver cancer. METHODS Five sgRNA sites were designed for the exons of Traf3. The recombinant plasmid of Lentiviral vector2-Traf3-sgRNA was constructed and transformed into Stbl3 competent cells. The recombinants were screened and sequenced, and the effectiveness of the designed gRNA was verified by sequencing. The constructed vector was transfected into HepG2 cells by lentiviral, and the monoclonal antibody was selected to detect the knockout effect of Traf3 gene in HepG2 cells by Western blot. PCR amplification and gene sequencing were performed to obtain the cell line, which the Traf3 gene was knocked out. MTT and Transwell assays were used to detect the effect of Traf3-knockout on HepG2 cell proliferation and cell invasion, respectively. RESULTS The Lentiviral vector2-sgRNA expression vector was successfully constructed. PCR amplification electrophoresis and gene sequencing showed that the Trep3-knockdown HepG2 cells were successfully constructed. Compared with the wild HepG2 cells group, the proliferation and invasion ability of HepG2 cells were enhanced in the Traf3 knockout group. CONCLUSION Knockout Traf3 gene by CRISPR/Cas9 system enhanced the proliferation, migration, and invasion of HepG2 cells, and provided an effective tool for studying the function and mechanism of Traf3.
Collapse
Affiliation(s)
- Wenbo Hu
- Class 12, Grade 2015, Faculty of Clinical Medicine, Jilin University, Changchun, Jilin, China
| | - Guiying Guo
- Department of Library, The People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Yuhua Chi
- Department of Oncology, The People's Hospital of Rizhao City, Rizhao, Shandong, China
| | - Fei Li
- Department of Respiratory Medicine, The People's Hospital of Rizhao City, Rizhao, Shandong, China
| |
Collapse
|
49
|
Tao Q, Tianyu W, Jiangqiao Z, Zhongbao C, Xiaoxiong M, Long Z, Jilin Z. Tripartite Motif 8 Deficiency Relieves Hepatic Ischaemia/reperfusion Injury via TAK1-dependent Signalling Pathways. Int J Biol Sci 2019; 15:1618-1629. [PMID: 31360105 PMCID: PMC6643225 DOI: 10.7150/ijbs.33323] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023] Open
Abstract
Tripartite motif (Trim) 8 is an E3 ubiquitin ligase, interacting with and ubiquitinating diverse substrates, and is closely involved in innate immunity. However, the function of Trim8 in hepatic ischaemia/reperfusion (I/R) injury remains largely unknown. The aim of this study is to explore the role of Trim8 in hepatic I/R injury. Trim8 gene knockout mice and primary hepatocytes were used to construct hepatic I/R models. The effect of Trim8 on hepatic I/R injury was analysed via pathological and molecular analyses. The results indicated that Trim8 was significantly upregulated in liver of mice subjected to hepatic I/R injury. Trim8 knockout relieved hepatocyte injury triggered by I/R. Silencing of Trim8 expression alleviated hepatic inflammation responses and inhibited apoptosis in vitro and in vivo. Mechanistically, our study suggests that Trim8 deficiency may elicit hepatic protective effects by inhibiting the activation of transforming growth factor β-activated kinase 1 (TAK1)-p38/JNK signalling pathways. TAK1 was required for Trim8 function in hepatic I/R injury as TAK1 activation abolished Trim8 function in vitro. In conclusion, our study demonstrates that Trim8 deficiency plays a protective role in hepatic I/R injury by inhibiting the activation of TAK1-dependent signalling pathways.
Collapse
Affiliation(s)
- Qiu Tao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Wang Tianyu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Zhou Jiangqiao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Chen Zhongbao
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Ma Xiaoxiong
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Zhang Long
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Zou Jilin
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| |
Collapse
|
50
|
Hung KY, Liao WI, Pao HP, Wu SY, Huang KL, Chu SJ. Targeting F-Box Protein Fbxo3 Attenuates Lung Injury Induced by Ischemia-Reperfusion in Rats. Front Pharmacol 2019; 10:583. [PMID: 31178737 PMCID: PMC6544082 DOI: 10.3389/fphar.2019.00583] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/06/2019] [Indexed: 01/12/2023] Open
Abstract
Background: Increasing evidence suggests that Fbxo3 signaling has an important impact on the pathophysiology of the inflammatory process. Fbxo3 protein inhibition has reduced cytokine-driven inflammation and improved disease severity in animal model of Pseudomonas-induced lung injury. However, it remains unclear whether inhibition of Fbxo3 protein provides protection in acute lung injury induced by ischemia-reperfusion (I/R). In this study, we investigated the protective effects of BC-1215 administration, a Fbxo3 inhibitor, on acute lung injury induced by I/R in rats. Methods: Lung I/R injury was induced by ischemia (40 min) followed by reperfusion (60 min). The rats were randomly assigned into one of six experimental groups (n = 6 rats/group): the control group, control + BC-1215 (Fbxo3 inhibitor, 0.5 mg/kg) group, I/R group, or I/R + BC-1215 (0.1, 0.25, 0.5 mg/kg) groups. The effects of BC-1215 on human alveolar epithelial cells subjected to hypoxia-reoxygenation (H/R) were also examined. Results: BC-1215 significantly attenuated I/R-induced lung edema, indicated by a reduced vascular filtration coefficient, wet/dry weight ratio, lung injury scores, and protein levels in bronchoalveolar lavage fluid (BALF). Oxidative stress and the level of inflammatory cytokines in BALF were also significantly reduced following administration of BC-1215. Additionally, BC-1215 mitigated I/R-stimulated apoptosis, NF-κB, and mitogen-activated protein kinase activation in the injured lung tissue. BC-1215 increased Fbxl2 protein expression and suppressed Fbxo3 and TNFR associated factor (TRAF)1–6 protein expression. BC-1215 also inhibited IL-8 production and NF-κB activation in vitro in experiments with alveolar epithelial cells exposed to H/R. Conclusions: Our findings demonstrated that Fbxo3 inhibition may represent a novel therapeutic approach for I/R-induced lung injury, with beneficial effects due to destabilizing TRAF proteins.
Collapse
Affiliation(s)
- Kuei-Yi Hung
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan.,Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|