1
|
Liu Y, Yuan W, Fang M, Guo H, Zhang X, Mei X, Zhang Y, Ji L, Gao Y, Wang J, Qian Z, Li M, Gao Y. Determination of HMGB1 in hepatitis B virus-related acute-on-chronic liver failure patients with acute kidney injury: Early prediction and prognostic implications. Front Pharmacol 2023; 13:1031790. [PMID: 36712653 PMCID: PMC9880762 DOI: 10.3389/fphar.2022.1031790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/13/2022] [Indexed: 01/15/2023] Open
Abstract
Background: Acute kidney injury (AKI) is a frequent complication in patients with hepatitis B virus-related acute-on-chronic liver failure (HBV-ACLF) and is associated with high rates of mortality. We aimed to estimate serum high mobility group protein 1 (HMGB1) levels in hepatitis B virus-related acute-on-chronic liver failure patients and analyze their clinical value in the development and outcomes of Acute kidney injury. Methods: A total of 251 consecutive patients with hepatitis B virus-related acute-on-chronic liver failure were enrolled in this retrospective study. Using the International Club of Ascites staging criteria of Acute kidney injury, 153 patients developed Acute kidney injury. The clinical data of patients were collected and serum levels of high mobility group protein 1 were measured by ELISA. All patients were followed up until death or for a minimum of 3 months. Early prediction and prognostic implications of high mobility group protein 1 in Hepatitis B Virus-Related Acute-on-Chronic Liver Failure Patients with Acute Kidney Injury were investigated in different cohorts, including a propensity score-matched ACLF cohort. Results: Among all individuals with hepatitis B virus-related acute-on-chronic liver failure, the incidence of Acute kidney injury was 61.0% (153/251). The patients who developed stage 2/3 Acute kidney injury showed the highest high mobility group protein 1 levels, followed by those who developed stage 1 Acute kidney injury, and those without Acute kidney injury showed the lowest high mobility group protein 1 levels. Moreover, high mobility group protein 1 levels were significantly higher in non-survivors than in survivors among hepatitis B virus-related acute-on-chronic liver failure patients with Acute kidney injury. Furthermore, analysis of the area under the receiver operating characteristic curve (AUROC) indicated that serum high mobility group protein 1 levels (pre-matching: AUC = 0.740; post-matching: AUC = 0.661) may be a potential predictive factor for Acute kidney injury development and that high mobility group protein 1 (AUC = 0.727) might be a reliable biomarker for prognosis in patients with Acute kidney injury. Conclusion: In patients with hepatitis B virus-related acute-on-chronic liver failure, Acute kidney injury is universal. Acute kidney injury and its stages negatively influence the 90-day transplant-free mortality rate. Serum high mobility group protein 1 levels can serve as a positive predictor of Acute kidney injury development, and high mobility group protein 1 might also be a prognostic biomarker for Acute kidney injury among hepatitis B virus-related acute-on-chronic liver failure patients.
Collapse
Affiliation(s)
- Yu Liu
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei Yuan
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Miao Fang
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongying Guo
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xin Zhang
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xue Mei
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yuyi Zhang
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Longshan Ji
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yating Gao
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiefei Wang
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhiping Qian
- Department of Liver Intensive Care Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China,*Correspondence: Zhiping Qian, ; Man Li, ; Yueqiu Gao,
| | - Man Li
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Zhiping Qian, ; Man Li, ; Yueqiu Gao,
| | - Yueqiu Gao
- Laboratory of Cellular Immunity, Institute of Clinical Immunology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China,Institute of Infectious Diseases of Integrated Traditional Chinese and Western Medicine, Shanghai, China,*Correspondence: Zhiping Qian, ; Man Li, ; Yueqiu Gao,
| |
Collapse
|
2
|
The E3 ubiquitin ligase NEDD4-1 protects against acetaminophen-induced liver injury by targeting VDAC1 for degradation. Acta Pharm Sin B 2023; 13:1616-1630. [PMID: 37139424 PMCID: PMC10150139 DOI: 10.1016/j.apsb.2023.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/27/2022] [Accepted: 12/15/2022] [Indexed: 01/30/2023] Open
Abstract
Acetaminophen (APAP) overdose is a major cause of liver injury. Neural precursor cell expressed developmentally downregulated 4-1 (NEDD4-1) is an E3 ubiquitin ligase that has been implicated in the pathogenesis of numerous liver diseases; however, its role in APAP-induced liver injury (AILI) is unclear. Thus, this study aimed to investigate the role of NEDD4-1 in the pathogenesis of AILI. We found that NEDD4-1 was dramatically downregulated in response to APAP treatment in mouse livers and isolated mouse hepatocytes. Hepatocyte-specific NEDD4-1 knockout exacerbated APAP-induced mitochondrial damage and the resultant hepatocyte necrosis and liver injury, while hepatocyte-specific NEDD4-1 overexpression mitigated these pathological events both in vivo and in vitro. Additionally, hepatocyte NEDD4-1 deficiency led to marked accumulation of voltage-dependent anion channel 1 (VDAC1) and increased VDAC1 oligomerization. Furthermore, VDAC1 knockdown alleviated AILI and weakened the exacerbation of AILI caused by hepatocyte NEDD4-1 deficiency. Mechanistically, NEDD4-1 was found to interact with the PPTY motif of VDAC1 through its WW domain and regulate K48-linked ubiquitination and degradation of VDAC1. Our present study indicates that NEDD4-1 is a suppressor of AILI and functions by regulating the degradation of VDAC1.
Collapse
|
3
|
Tian T, Yao D, Zheng L, Zhou Z, Duan Y, Liu B, Wang P, Li Y. Sphingosine kinase 1 regulates HMGB1 translocation by directly interacting with calcium/calmodulin protein kinase II-δ in sepsis-associated liver injury. Cell Death Dis 2020; 11:1037. [PMID: 33281190 PMCID: PMC7719708 DOI: 10.1038/s41419-020-03255-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 01/22/2023]
Abstract
Previously, we confirmed that sphingosine kinase 1 (SphK1) inhibition improves sepsis-associated liver injury. High-mobility group box 1 (HMGB1) translocation participates in the development of acute liver failure. However, little information is available on the association between SphK1 and HMGB1 translocation during sepsis-associated liver injury. In the present study, we aimed to explore the effect of SphK1 inhibition on HMGB1 translocation and the underlying mechanism during sepsis-associated liver injury. Primary Kupffer cells and hepatocytes were isolated from SD rats. The rat model of sepsis-associated liver damage was induced by intraperitoneal injection with lipopolysaccharide (LPS). We confirmed that Kupffer cells were the cells primarily secreting HMGB1 in the liver after LPS stimulation. LPS-mediated HMGB1 expression, intracellular translocation, and acetylation were dramatically decreased by SphK1 inhibition. Nuclear histone deacetyltransferase 4 (HDAC4) translocation and E1A-associated protein p300 (p300) expression regulating the acetylation of HMGB1 were also suppressed by SphK1 inhibition. HDAC4 intracellular translocation has been reported to be controlled by the phosphorylation of HDAC4. The phosphorylation of HDAC4 is modulated by CaMKII-δ. However, these changes were completely blocked by SphK1 inhibition. Additionally, by performing coimmunoprecipitation and pull-down assays, we revealed that SphK1 can directly interact with CaMKII-δ. The colocalization of SphK1 and CaMKII-δ was verified in human liver tissues with sepsis-associated liver injury. In conclusion, SphK1 inhibition diminishes HMGB1 intracellular translocation in sepsis-associated liver injury. The mechanism is associated with the direct interaction of SphK1 and CaMKII-δ.
Collapse
Affiliation(s)
- Tao Tian
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Danhua Yao
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Lei Zheng
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Zhiyuan Zhou
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Yantao Duan
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Bin Liu
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China
| | - Pengfei Wang
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China.
| | - Yousheng Li
- Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
4
|
Cardiopulmonary Bypass Induces Acute Lung Injury via the High-Mobility Group Box 1/Toll-Like Receptor 4 Pathway. DISEASE MARKERS 2020; 2020:8854700. [PMID: 33062073 PMCID: PMC7532999 DOI: 10.1155/2020/8854700] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/23/2020] [Accepted: 08/31/2020] [Indexed: 11/25/2022]
Abstract
During cardiopulmonary bypass (CPB), pulmonary ischemia/reperfusion (I/R) injury can cause acute lung injury (ALI). Our previous research confirmed that abnormal high-mobility group box 1 (HMGB1) release after CPB was closely related to ALI. However, the mechanism underlying the HMGB1-mediated induction of ALI after CPB is unclear. Our previous study found that HMGB1 binds Toll-like receptor 4 (TLR4), leading to lung injury, but direct evidence of a role for these proteins in the mechanism of CPB-induced lung injury has not been shown. We examined the effects of inhibiting HMGB1 or reducing TLR4 expression on CPB-induced lung injury in rats administered anti-HMBG1 antibody or TLR4 short-hairpin RNA (shTLR4), respectively. In these rat lungs, we studied the histologic changes and levels of interleukin- (IL-) 1β, tumour necrosis factor- (TNF-) α, HMGB1, and TLR4 after CPB. After CPB, the lung tissues from untreated rats showed histologic features of injury and significantly elevated levels of IL-1β, TNF-α, HMGB1, and TLR4. Treatment with anti-HMGB1 attenuated the CPB-induced morphological inflammatory response and protein levels of IL-1β, TNF-α, HMGB1, and TLR4 in the lung tissues and eventually alleviated the ALI after CPB. Treatment with shTLR4 attenuated the CPB-induced morphological inflammatory response and protein levels of IL-1β, TNF-α, and TLR4 in the lung tissues and eventually alleviated the ALI after CPB, but could not alleviate the HMGB1 protein levels induced by CPB. In summary, the present study demonstrated that the HMGB1/TLR4 pathway mediated the development of ALI induced by CPB.
Collapse
|
5
|
Minsart C, Liefferinckx C, Lemmers A, Dressen C, Quertinmont E, Leclercq I, Devière J, Moreau R, Gustot T. New insights in acetaminophen toxicity: HMGB1 contributes by itself to amplify hepatocyte necrosis in vitro through the TLR4-TRIF-RIPK3 axis. Sci Rep 2020; 10:5557. [PMID: 32221312 PMCID: PMC7101425 DOI: 10.1038/s41598-020-61270-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/20/2020] [Indexed: 12/26/2022] Open
Abstract
Extracellular release of HMGB1 contributes to acetaminophen-induced liver injury. HMGB1 acts as a danger-associated molecular patterns during this toxic process but the mechanisms of action and targeted cells are incompletely defined. Here we studied, in vitro, the role of HMGB1 in amplifying the acetaminophen-induced hepatocyte necrosis process. Using cultured HepaRG cells, primary human hepatocytes and selective chemical inhibitors we evaluated acetaminophen-induced toxicity. We confirmed that addition of acetaminophen induced HepaRG cell death and HMGB1 release. We showed that inhibition of HMGB1 decreased acetaminophen-induced HepaRG cell death, suggesting a feedforward effect. We provide the first evidence that exposure of HepaRG cells to recombinant human HMGB1 (rhHMGB1) also resulted in cell death. Moreover, we found that both acetaminophen and rhHMGB1 induced programmed HepaRG cell necrosis through a RIPK3-dependent mechanism. By using TLR4 blocking antibody, we demonstrated the reduction of the HepaRG cell death induced by acetaminophen and rhHMGB1. Furthermore, inhibition of TRIF, known to induce a RIPK3-dependent cell death, reduced rhHMGB1-induced HepaRG cell death. Our data support that released HMGB1 from acetaminophen-stressed hepatocytes induced necrosis of neighboring hepatocytes by TLR4-TRIF-RIPK3- pathway. This in vitro study gives new insights in the role of HMGB1 in the amplification of acetaminophen-induced toxicity.
Collapse
Affiliation(s)
- Charlotte Minsart
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium.
| | - Claire Liefferinckx
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Arnaud Lemmers
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, HepatoPancreatology and Digestive Oncology, C.U.B. Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Cindy Dressen
- Laboratory of Physiology and Pharmacology, Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Quertinmont
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Leclercq
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jacques Devière
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, HepatoPancreatology and Digestive Oncology, C.U.B. Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Richard Moreau
- Inserm Unité 1149, Centre de Recherche sur l'inflammation [CRI], Paris, France
- UMR S_1149, Université Paris Diderot, Paris, France
- DHU UNITY, Service d'Hépatologie, Hôpital Beaujon, APHP, Clichy, France
| | - Thierry Gustot
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium.
- Department of Gastroenterology, HepatoPancreatology and Digestive Oncology, C.U.B. Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium.
- UMR S_1149, Université Paris Diderot, Paris, France.
- DHU UNITY, Service d'Hépatologie, Hôpital Beaujon, APHP, Clichy, France.
| |
Collapse
|
6
|
Gaskell H, Ge X, Nieto N. High-Mobility Group Box-1 and Liver Disease. Hepatol Commun 2018; 2:1005-1020. [PMID: 30202816 PMCID: PMC6128227 DOI: 10.1002/hep4.1223] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/03/2018] [Indexed: 12/12/2022] Open
Abstract
High‐mobility group box‐1 (HMGB1) is a ubiquitous protein. While initially thought to be simply an architectural protein due to its DNA‐binding ability, evidence from the last decade suggests that HMGB1 is a key protein participating in the pathogenesis of acute liver injury and chronic liver disease. When it is passively released or actively secreted after injury, HMGB1 acts as a damage‐associated molecular pattern that communicates injury and inflammation to neighboring cells by the receptor for advanced glycation end products or toll‐like receptor 4, among others. In the setting of acute liver injury, HMGB1 participates in ischemia/reperfusion, sepsis, and drug‐induced liver injury. In the context of chronic liver disease, it has been implicated in alcoholic liver disease, liver fibrosis, nonalcoholic steatohepatitis, and hepatocellular carcinoma. Recently, specific posttranslational modifications have been identified that could condition the effects of the protein in the liver. Here, we provide a detailed review of how HMGB1 signaling participates in acute liver injury and chronic liver disease.
Collapse
Affiliation(s)
- Harriet Gaskell
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Xiaodong Ge
- Department of Pathology University of Illinois at Chicago Chicago IL
| | - Natalia Nieto
- Department of Pathology University of Illinois at Chicago Chicago IL.,Department of Medicine University of Illinois at Chicago Chicago IL
| |
Collapse
|
7
|
Amemiya R, Shinoda M, Yamada M, Ueno Y, Shimada K, Fujieda H, Yagi H, Mizota T, Nishiyama R, Oshima G, Yamada S, Matsubara K, Abe Y, Hibi T, Kitago M, Obara H, Itano O, Kitagawa Y. Hemoadsorption of high-mobility-group box 1 using a porous polymethylmethacrylate fiber in a swine acute liver failure model. Int J Artif Organs 2018. [PMID: 29528759 DOI: 10.1177/0391398817752296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND High-mobility-group box chromosomal protein 1 has been identified as an important mediator of various kinds of acute and chronic inflammation. In this study, we aimed to develop a column that effectively adsorbs high-mobility-group box chromosomal protein 1 by altering the pore size of the fiber. MATERIALS AND METHODS First, we produced three types of porous polymethylmethacrylate fiber by altering the concentration of polymethylmethacrylate dissolved in dimethylsulfoxide. We then selected a fiber based on the results of an in vitro incubation test of high-mobility-group box chromosomal protein 1 adsorption. Using the selected fiber, we constructed a new column and tested its high-mobility-group box chromosomal protein 1 adsorption capacity during 4-h extracorporeal hemoperfusion in a swine acute liver failure model. RESULTS Electron microscope observation showed that the three types of fibers had different pore sizes on the surface and in cross section, which were dependent on the concentration of polymethylmethacrylate. In the in vitro incubation test, fiber with moderate-sized pores demonstrated the highest adsorption capacity. In the in vivo hemoperfusion study, the ratio of the high-mobility-group box chromosomal protein 1 concentration at the outlet versus the inlet of the column was significantly lower with the new column than with the control column during 4-h extracorporeal hemoperfusion. The normalized plasma level of high-mobility-group box chromosomal protein 1 at 12 h after the completion of hemoperfusion was significantly lower with the new column than with the control column. CONCLUSION The newly developed polymethylmethacrylate column adsorbs high-mobility-group box chromosomal protein 1 during hemoperfusion in swine ALF model.
Collapse
Affiliation(s)
- Ryusuke Amemiya
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Shinoda
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Yamada
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Yoshiyuki Ueno
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Kaoru Shimada
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Hiroaki Fujieda
- 2 Medical Devices & Materials Research Unit, Advanced Materials Research Laboratories, Toray Industries, Inc., Shiga, Japan
| | - Hiroshi Yagi
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Takamasa Mizota
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Nishiyama
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Go Oshima
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shingo Yamada
- 3 Central Institute, Shino-Test Corporation, Kanagawa, Japan
| | - Kentaro Matsubara
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Taizo Hibi
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hideaki Obara
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Osamu Itano
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- 1 Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Abstract
Although acute liver failure (ALF) is a rare disease, it continues to have high mortality and morbidity rates due to its many causes. High mobility group box 1 (HMGB1), originally reported as a ubiquitous non-histone chromosomal protein, is a multi-functional protein with varying functions depending on its location, such as in the nucleus, cytoplasm and extracellular space. The role of extracellular HMGB1 as an inflammatory mediator has been well studied, and the elevation of serum HMGB1 has been reported in several diseases that are closely associated with ALF. Areas covered: In this review, we focus on the relationship between causes of acute liver failure, such as viral infection, drug-induced liver injury, ischemia/reperfusion injury, and acute-on-chronic liver failure, and the role of HMGB1. Furthermore, we also consolidate and summarize the current reports of HMGB1-targeting therapies in hepatic injury models. Expert commentary: HMGB1 could be a novel therapeutic candidate for ALF, and the clinical testing of HMGB1-targeting therapies for ALF patients is expected.
Collapse
Affiliation(s)
- Tetsu Yamamoto
- a Department of Digestive and General Surgery , Shimane University Faculty of Medicine , Izumo , Japan
| | - Yoshitsugu Tajima
- a Department of Digestive and General Surgery , Shimane University Faculty of Medicine , Izumo , Japan
| |
Collapse
|
9
|
Therapeutic potential of recombinant thrombomodulin for lung injury after pneumonectomy via inhibition of high-mobility group box 1 in mice. J Trauma Acute Care Surg 2017; 81:868-875. [PMID: 27504958 DOI: 10.1097/ta.0000000000001208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Surgical acute respiratory distress syndrome (ARDS) is an extremely critical condition which may occur after major lung resection. Despite advances in minimally invasive surgical procedures and progress in the therapeutic management of this disease, prognosis remains poor. In this study, we investigated the contribution of high-mobility group box 1 (HMGB1) in a surgical ARDS model and evaluated the possible therapeutic effect of recombinant thrombomodulin (rTM) for the treatment of surgical ARDS. METHODS C57BL/6J mice underwent left pneumonectomy. rTM was injected at 12 hours before surgery, followed by 12 hours for 3 days after surgery. Lipopolysaccharide (LPS) was administered at 2 hours after surgery. We conducted a histologic analysis and measured HMGB1, IL-6, IL-1β, and TNF-α in bronchoalveolar lavage fluid on day 3 after pneumonectomy. Data were compared between the treatment groups. RESULTS On histologic analysis, left pneumonectomy followed by LPS administration induced both severe inflammatory cellular infiltration and alveolar wall congestion with hemorrhage. rTM administration rescued these histologic changes. The level of HMGB1, IL-6, IL-1β, and TNF-α in bronchoalveolar lavage fluid was significantly increased by LPS administration after pneumonectomy and significantly decreased by rTM administration with LPS and pneumonectomy (p < 0.001). Also, LPS alone showed no statistical differences in HMGB1 or proinflammatory cytokine level compared with pneumonectomy (PNX) group. In addition, the survival outcome was also improved by rTM administration. CONCLUSIONS LPS administration after left pneumonectomy could induce the severe lung injury. PNX and LPS have similar contribution to this model and may play a synergistic role in this process. rTM may have the potential therapeutic effect for surgical ARDS via suppression of HMGB1 and the secretion of proinflammatory cytokines induced by the administration of LPS after left pneumonectomy.
Collapse
|
10
|
Li X, Liu HC, Yao QY, Xu BL, Zhang SC, Tu CT. Quercetin Protects Mice from ConA-Induced Hepatitis by Inhibiting HMGB1-TLR Expression and Down-Regulating the Nuclear Factor Kappa B Pathway. Inflammation 2016; 39:96-106. [PMID: 26267064 DOI: 10.1007/s10753-015-0227-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The dietary flavonoid quercetin has hepatoprotective effects. We analyzed the effects of quercetin on concanavalin A (ConA)-induced hepatitis in mice and its underlying molecular mechanisms of action. Mice were administered quercetin (50 mg/kg body weight, i.p.) or vehicle 30 min before intravenous administration of ConA. Quercetin pretreatment significantly reduced the ConA-induced elevations in plasma aminotransferase concentrations and liver necrosis, as well as reducing serum concentrations of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, interferon-γ, and interleukin-4. Quercetin pretreatment also reduced expression of high-mobility group box 1 protein (HMGB1) and toll-like receptor (TLR)-2 and TLR-4 messenger RNA (mRNA) and protein in liver tissues. Quercetin pretreatment significantly inhibited degradation of inhibitory kappa B alpha and modulated ConA-induced nuclear translocation in the liver of nuclear factor kappa B (NF-κB) p65. These results demonstrate that quercetin protects against ConA-mediated hepatitis in mice by attenuating the HMGB1-TLRs-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xi Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong-Chun Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Qun-Yan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Bei-Li Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Shun-Cai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Liver Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Chuan-Tao Tu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Institute of Liver Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
11
|
Larsen FS, Schmidt LE, Bernsmeier C, Rasmussen A, Isoniemi H, Patel VC, Triantafyllou E, Bernal W, Auzinger G, Shawcross D, Eefsen M, Bjerring PN, Clemmesen JO, Hockerstedt K, Frederiksen HJ, Hansen BA, Antoniades CG, Wendon J. High-volume plasma exchange in patients with acute liver failure: An open randomised controlled trial. J Hepatol 2016; 64:69-78. [PMID: 26325537 DOI: 10.1016/j.jhep.2015.08.018] [Citation(s) in RCA: 406] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Revised: 07/25/2015] [Accepted: 08/11/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Acute liver failure (ALF) often results in cardiovascular instability, renal failure, brain oedema and death either due to irreversible shock, cerebral herniation or development of multiple organ failure. High-volume plasma exchange (HVP), defined as exchange of 8-12 or 15% of ideal body weight with fresh frozen plasma in case series improves systemic, cerebral and splanchnic parameters. METHODS In this prospective, randomised, controlled, multicentre trial we randomly assigned 182 patients with ALF to receive either standard medical therapy (SMT; 90 patients) or SMT plus HVP for three days (92 patients). The baseline characteristics of the groups were similar. The primary endpoint was liver transplantation-free survival during hospital stay. Secondary-endpoints included survival after liver transplantation with or without HVP with intention-to-treat analysis. A proof-of-principle study evaluating the effect of HVP on the immune cell function was also undertaken. RESULTS For the entire patient population, overall hospital survival was 58.7% for patients treated with HVP vs. 47.8% for the control group (hazard ratio (HR), with stratification for liver transplantation: 0.56; 95% confidence interval (CI), 0.36-0.86; p=0.0083). HVP prior to transplantation did not improve survival compared with patients who received SMT alone (CI 0.37 to 3.98; p=0.75). The incidence of severe adverse events was similar in the two groups. Systemic inflammatory response syndrome (SIRS) and sequential organ failure assessment (SOFA) scores fell in the treated group compared to control group, over the study period (p<0.001). CONCLUSIONS Treatment with HVP improves outcome in patients with ALF by increasing liver transplant-free survival. This is attributable to attenuation of innate immune activation and amelioration of multi-organ dysfunction.
Collapse
Affiliation(s)
| | | | | | - Allan Rasmussen
- Department of Surgery and Liver Transplantation C, Rigshospitalet, Copenhagen, Denmark
| | - Helena Isoniemi
- Transplantation and Liver Surgery Clinic, Helsinki University Hospital, Finland
| | - Vishal C Patel
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | | | - William Bernal
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Georg Auzinger
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Debbie Shawcross
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Martin Eefsen
- Department of Hepatology, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Krister Hockerstedt
- Transplantation and Liver Surgery Clinic, Helsinki University Hospital, Finland
| | | | | | - Charalambos G Antoniades
- Institute of Liver Studies, King's College Hospital, London, United Kingdom; Section of Hepatology, St. Mary's Hospital, Imperial College London, London, UK
| | - Julia Wendon
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| |
Collapse
|
12
|
Farnesyltransferase inhibitor, tipifarnib, prevents galactosamine/lipopolysaccharide-induced acute liver failure. Shock 2015; 42:570-577. [PMID: 25046541 DOI: 10.1097/shk.0000000000000239] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Acute liver failure (ALF) is a fatal syndrome associated with massive hepatocyte death. There is no cure for ALF except liver transplantation. Protein farnesylation is a lipid modification of cysteine residues that is catalyzed by farnesyltransferase (FTase) and has been proposed as an integral component of acute inflammation. Previously, we have demonstrated that FTase inhibitors improve survival in mouse models of endotoxemia and sepsis. Here we studied the effects of FTase inhibitor, tipifarnib, on galactosamine (GalN)/lipopolysaccharide (LPS)-induced ALF. The effects of tipifarnib (10 mg/kg, i.p.) were studied in GalN (400 mg/kg, i.p.)- and LPS (3 μg/kg)-challenged mice by histological and biochemical analyses. Galactosamine/LPS administration caused prominent liver injury characterized by the increased plasma alanine aminotransferase and aspartic aminotransferase levels, leading to significant mortality in mice. Tipifarnib inhibited GalN/LPS-induced caspase 3 activation, inflammatory cytokine production, and c-Jun N-terminal kinase phosphorylation in the liver. On the other hand, tipifarnib upregulated antiapoptotic protein, Bcl-xL, in the liver after GalN/LPS challenge. Tipifarnib also protected primary hepatocytes from GalN/tumor necrosis factor α-induced cell death by inhibiting caspase 3 activation and upregulating antiapoptotic proteins. Galactosamine/LPS-induced liver injury was associated with increased protein farnesylation in the liver. Tipifarnib prevented protein farnesylation in the liver and markedly attenuated liver injury and mortality in GalN/LPS-challenged mice. These results suggest that protein farnesylation is a novel potential molecular target to prevent hepatocyte death and acute inflammatory liver failure in fulminant hepatitis.
Collapse
|
13
|
Thrombomodulin improves rat survival after extensive hepatectomy. J Surg Res 2015; 194:375-382. [DOI: 10.1016/j.jss.2014.10.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 10/11/2014] [Accepted: 10/29/2014] [Indexed: 01/08/2023]
|
14
|
Tanaka M, Shinoda M, Takayanagi A, Oshima G, Nishiyama R, Fukuda K, Yagi H, Hayashida T, Masugi Y, Suda K, Yamada S, Miyasho T, Hibi T, Abe Y, Kitago M, Obara H, Itano O, Takeuchi H, Sakamoto M, Tanabe M, Maruyama I, Kitagawa Y. Gene transfer of high-mobility group box 1 box-A domain in a rat acute liver failure model. J Surg Res 2015; 194:571-580. [PMID: 25498512 DOI: 10.1016/j.jss.2014.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 10/13/2014] [Accepted: 11/13/2014] [Indexed: 01/18/2023]
|
15
|
The sterile inflammation in the exacerbation of HBV-associated liver injury. Mediators Inflamm 2015; 2015:508681. [PMID: 25892853 PMCID: PMC4393905 DOI: 10.1155/2015/508681] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/25/2014] [Accepted: 10/08/2014] [Indexed: 12/20/2022] Open
Abstract
Exacerbation of hepatitis B virus-associated liver injury is characterized by abnormal immune response which not only mobilizes specific antiviral effects but also poses a potentially lethal nonspecific sterile inflammation to the host. How nonspecific sterile inflammation is triggered after the preexisting injury caused by specific immune injury remains elusive. In the setting of sterile inflammation, endogenous damage-associated molecular patterns are released by stressed and dying hepatocytes, which alarm the immune system through their potential pattern recognition receptors and related signaling pathways, orchestrate the influx of diverse cytokines, and ultimately amplify liver destruction. This review highlights current knowledge about the sterile hepatic inflammation in the exacerbation of chronic hepatitis B.
Collapse
|
16
|
Protective effect of neutralization of the extracellular high-mobility group box 1 on renal ischemia-reperfusion injury in miniature swine. Transplantation 2015; 98:937-43. [PMID: 25136847 DOI: 10.1097/tp.0000000000000358] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Strategies that reduce ischemia-reperfusion injury (IRI) have the potential to expand the numbers of available organs for transplantation. Recent reports in rodent models have demonstrated that high-mobility group box 1 (HMGB1) acts as an alarm in initiating the inflammatory response resulting from ischemic injury. The aim of this study was to evaluate the cytoprotective effects of anti-HMGB1 antibodies on renal IRI in preclinical large animals. METHODS One hundred twenty minutes of warm and 60 min of cold renal ischemia were induced in 8 CLAWN miniature swine. Three of eight animals received intravenous anti-HMGB1 antibody at 1 mg/kg just before the reperfusion of renal blood flow. Renal function was assessed by serum creatinine and renal biopsy. Serum levels of interleukin (IL)-1β, IL-6, and HMGB1 were measured. RESULTS The concentration of HMGB1 increased as early as 30 min after reperfusion and before the elevation of IL-1β and IL-6. Serum creatinine levels were markedly elevated, peaking at a median of 5 days (peak creatinine levels: 11.6 ± 1.6 mg/dL) and recovering by day 14. Anti-HMGB1 antibody injection dramatically decreased renal damage as well as serum levels of HMGB1 associated with IRI. Renal function returned to near normal by day 9, and peak creatinine levels were markedly lower (7.4 ± 0.2 mg/dL), and biopsies possessed fewer pathologic changes when compared to the control group. CONCLUSION In this study, we demonstrated the beneficial effects of perioperative administration of anti-HMGB1 antibody in reducing renal IRI in a clinically relevant, large animal model.
Collapse
|
17
|
Li X, Gou C, Yang H, Qiu J, Gu T, Wen T. Echinacoside ameliorates D-galactosamine plus lipopolysaccharide-induced acute liver injury in mice via inhibition of apoptosis and inflammation. Scand J Gastroenterol 2014; 49:993-1000. [PMID: 24797709 DOI: 10.3109/00365521.2014.913190] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE This study aimed to investigate the protective effects of echinacoside, one of the phenylethanoids isolated from the stems of Cistanche salsa, a Chinese herbal medicine, on D-galactosamine (GalN) and lipopolysaccharide (LPS)-induced acute liver injury in mice. METHODS We administered GalN (650 mg/kg) together with LPS (30 μg/kg) to mice by intraperitoneal injection to induce acute liver damage. Echinacoside (60 mg/kg) was given intraperitoneally to mice at 1 h prior to GalN/LPS exposure. Mice were sacrificed at different time points following GalN/LPS treatment, and the liver and blood samples were collected for future analysis. RESULTS It showed that GalN/LPS treatment produced severe hepatic injury, evidenced by significantly elevated plasma alanine aminotransferase (ALT) levels and abnormal histological changes such as hepatocyte necrosis or apoptosis, hemorrhage, fatty degeneration, and neutrophil infiltration. Notably, pretreatment with echinacoside remarkably improved the survival rate of GalN/LPS-treated mice and attenuated acute hepatotoxicity, as demonstrated by decreased ALT levels and improved histological signs. Echinacoside shows both anti-apoptotic and anti-inflammatory properties, characterized by a substantial inhibition of hepatocyte apoptosis and a significant reduction in the inflammatory markers, including myeloperoxidase, extracellular nucleosomes, high-mobility group box 1, and inflammatory cytokines in the plasma of mice, which may be important mechanisms related to its protective effect. CONCLUSION Our results suggest that echinacoside can provide a pronounced protection against GalN/LPS-induced acute liver injury in mice, which may complement the available strategies for management of acute liver damage in clinical settings.
Collapse
Affiliation(s)
- Xiuhui Li
- Department of Integrated TCM and Western Medicine, Beijing Youan Hospital Affiliated with Capital Medical University , Beijing, 100069 , P.R.China
| | | | | | | | | | | |
Collapse
|
18
|
Abstract
BACKGROUND Acute liver failure (ALF) is a serious clinical syndrome with high mortality. Sodium butyrate has been shown to alleviate organ injury in a wide variety of preclinical models of critical diseases. The aim of this study was to investigate the protective effect of sodium butyrate on ALF in rats. METHODS All rats were randomly divided into control, model and sodium butyrate treatment groups. Except the control group, the rats were induced ALF animal model by subcutaneous injection of human serum albumin+ D-galactosamine+lipopolysaccharide. After induction of ALF, the rats in the treatment group received sodium butyrate (500 mg/kg) at 12-hour or 24-hour time point. Fourty-eight hours after ALF induction, the animals were sacrificed and samples were harvested. Serum endotoxin, high mobility group box-1 (HMGB1), liver function parameters, tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) were measured. The expression of HMGB1 and nuclear factor-kappa B (NF-kappaB) p65 protein in liver tissue was detected by Western blotting. The histological changes of liver and intestine were examined. The survival duration was also observed. RESULTS Serum endotoxin, alanine aminotransferase, HMGB1, TNF-alpha and IFN-gamma were significantly increased and the liver histology showed more severe histopathological injury in the model group compared with the control group (P<0.05). Compared to the model group, sodium butyrate treatment significantly improved the histopathological changes in the liver and intestine, reduced serum endotoxin and inflammatory cytokines, suppressed HMGB1 and NF-kappaB p65 proteins in liver tissue, and prolonged the survival duration regardless of treatment at 12 hours or 24 hours after induction of ALF (P<0.05). CONCLUSIONS Sodium butyrate protected the liver from toxin-induced ALF in rats. The mechanisms may be due to direct hepatoprotection and decreased intestinal permeability.
Collapse
|
19
|
Abstract
INTRODUCTION Sepsis refers to the host's deleterious and non-resolving systemic inflammatory response to microbial infections and represents the leading cause of death in the intensive care unit. The pathogenesis of sepsis is complex, but partly mediated by a newly identified alarmin molecule, the high mobility group box 1 (HMGB1). AREAS COVERED Here we review the evidence that support extracellular HMGB1 as a late mediator of experimental sepsis with a wider therapeutic window and discuss the therapeutic potential of HMGB1-neutralizing antibodies and small molecule inhibitors (herbal components) in experimental sepsis. EXPERT OPINION It will be important to evaluate the efficacy of HMGB1-targeting strategies for the clinical management of human sepsis in the future.
Collapse
Affiliation(s)
- Haichao Wang
- The Feinstein Institute for Medical Research and North Shore University Hospital, The Hofstra North Shore - LIJ School of Medicine, Laboratory of Emergency Medicine, North Shore-LIJ Health System , 350 Community Drive, Manhasset, NY 11030 , USA +1 516 562 2823 ; +1 516 562 1022 ;
| | | | | |
Collapse
|
20
|
Kikuchi K, Tancharoen S, Ito T, Morimoto-Yamashita Y, Miura N, Kawahara KI, Maruyama I, Murai Y, Tanaka E. Potential of the angiotensin receptor blockers (ARBs) telmisartan, irbesartan, and candesartan for inhibiting the HMGB1/RAGE axis in prevention and acute treatment of stroke. Int J Mol Sci 2013; 14:18899-924. [PMID: 24065095 PMCID: PMC3794813 DOI: 10.3390/ijms140918899] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/07/2013] [Accepted: 09/09/2013] [Indexed: 12/29/2022] Open
Abstract
Stroke is a major cause of mortality and disability worldwide. The main cause of stroke is atherosclerosis, and the most common risk factor for atherosclerosis is hypertension. Therefore, antihypertensive treatments are recommended for the prevention of stroke. Three angiotensin receptor blockers (ARBs), telmisartan, irbesartan and candesartan, inhibit the expression of the receptor for advanced glycation end-products (RAGE), which is one of the pleiotropic effects of these drugs. High mobility group box 1 (HMGB1) is the ligand of RAGE, and has been recently identified as a lethal mediator of severe sepsis. HMGB1 is an intracellular protein, which acts as an inflammatory cytokine when released into the extracellular milieu. Extracellular HMGB1 causes multiple organ failure and contributes to the pathogenesis of hypertension, hyperlipidemia, diabetes mellitus, atherosclerosis, thrombosis, and stroke. This is the first review of the literature evaluating the potential of three ARBs for the HMGB1-RAGE axis on stroke therapy, including prevention and acute treatment. This review covers clinical and experimental studies conducted between 1976 and 2013. We propose that ARBs, which inhibit the HMGB1/RAGE axis, may offer a novel option for prevention and acute treatment of stroke. However, additional clinical studies are necessary to verify the efficacy of ARBs.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, 6 Yothe Road, Rajthevee, Bangkok 10400, Thailand; E-Mails: (K.K.); (S.T.)
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
- Department of Neurosurgery, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, 6 Yothe Road, Rajthevee, Bangkok 10400, Thailand; E-Mails: (K.K.); (S.T.)
| | - Takashi Ito
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan; E-Mails: (T.I.); (I.M.)
| | - Yoko Morimoto-Yamashita
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan; E-Mail:
| | - Naoki Miura
- Laboratory of Diagnostic Imaging, Department of Veterinary Science, Faculty of Agriculture, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan; E-Mail:
| | - Ko-ichi Kawahara
- Laboratory of Functional Foods, Department of Biomedical Engineering Osaka Institute of Technology, 5-16-1 Omiya, Asahi Ward, Osaka 535-8585, Japan; E-Mail:
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan; E-Mails: (T.I.); (I.M.)
| | - Yoshinaka Murai
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
| | - Eiichiro Tanaka
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan; E-Mail:
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-942-31-7542; Fax: +81-942-31-7695
| |
Collapse
|
21
|
Curcumin protects against concanavalin A-induced hepatitis in mice through inhibiting the cytoplasmic translocation and expression of high mobility group box 1. Inflammation 2013; 36:206-15. [PMID: 22948514 DOI: 10.1007/s10753-012-9536-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The aims of this study were to examine the anti-inflammatory effect of curcumin on concanavalin A (ConA) induced hepatitis in mice, and to elucidate its underlying molecular mechanisms. Mice received curcumin by gavage before ConA intravenous administration. The results showed that curcumin pretreatment attenuated ConA-induced hepatitis. Enzyme linked immunosorbent assay (ELISA) results showed that serum levels of high mobility group box 1 (HMGB1) increased at 4 h and reached its peak value at 12 h after challenge with ConA; but this increase was significantly inhibited by curcumin. Furthermore, curcumin significantly decreased the HMGB1 translocation from nucleus to cytoplasm of hepatocytes in ConA-induced mice. The levels of HMGB1 mRNA and protein expression in the liver were also significantly lowered in curcumin-treated mice. In addition, curcumin inhibited intrahepatic expression of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 protein. In conclusion, the results indicated that curcumin protected against ConA-induced hepatitis in mice; and the beneficial effects may be partly through inhibition of HMGB1 translocation in hepatocytes, release into the plasma and expression in livers.
Collapse
|
22
|
Fujimura N, Obara H, Suda K, Takeuchi H, Miyasho T, Kawasako K, Du W, Yamada S, Ono S, Matsumoto K, Matsuda S, Yagi H, Kitago M, Shinoda M, Itano O, Tanabe M, Sakamoto M, Maruyama I, Kitagawa Y. Neutrophil elastase inhibitor improves survival rate after ischemia reperfusion injury caused by supravisceral aortic clamping in rats. J Surg Res 2013; 180:e31-6. [PMID: 22595015 DOI: 10.1016/j.jss.2012.04.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/27/2012] [Accepted: 04/17/2012] [Indexed: 11/16/2022]
|
23
|
Wang W, Sun L, Deng Y, Tang J. Synergistic effects of antibodies against high-mobility group box 1 and tumor necrosis factor-α antibodies on D-(+)-galactosamine hydrochloride/lipopolysaccharide-induced acute liver failure. FEBS J 2013; 280:1409-19. [PMID: 23331758 DOI: 10.1111/febs.12132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 12/19/2012] [Accepted: 01/14/2013] [Indexed: 12/18/2022]
Abstract
High-mobility group box 1 (HMGB1) protein is released into the serum after tissue damage, and serves as a warning signal to enhance the inflammatory response. Acute liver injury is one of the diseases that starts with tissue damage and ends with systemic inflammation. We used D-(+)-galactosamine hydrochloride (D-GalN)/lipopolysaccharide (LPS)-treated mice as an acute liver injury model to explore the functions of HMGB1 in more detail. HMGB1 is released into the serum at a very early stage of D-GalN/LPS-induced acute liver injury. It upregulates the expression of tumor necrosis factor-α (TNF-α), interleukin-6, inducible nitric oxide synthase, and tissue factor. TNF-α and HMGB1 form a positive feedback loop to amplify the downstream signals. mAbs against HMGB1 and TNF-α have synergistic effects in protecting mice from D-GalN/LPS-induced acute liver failure. The results suggest that HMGB1 is a key mediator in D-GalN/LPS-induced acute liver injury. Tissue damage and cell necrosis shortly after administration of D-GalN and LPS lead to early HMGB1 release, and HMGB1 acts synergistically with TNF-α to promote pathological processes in acute liver failure.
Collapse
Affiliation(s)
- Wei Wang
- Center for Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | | | | | | |
Collapse
|
24
|
Oshima G, Shinoda M, Tanabe M, Ebinuma H, Nishiyama R, Takano K, Yamada S, Miyasho T, Masugi Y, Matsuda S, Suda K, Fukunaga K, Matsubara K, Hibi T, Yagi H, Hayashida T, Yamagishi Y, Obara H, Itano O, Takeuchi H, Kawachi S, Saito H, Hibi T, Maruyama I, Kitagawa Y. Increased plasma levels of high mobility group box 1 in patients with acute liver failure. ACTA ACUST UNITED AC 2012; 48:154-62. [PMID: 22585050 DOI: 10.1159/000338363] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 03/07/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) is a monocyte-derived late-acting inflammatory mediator, which is released in conditions such as shock, tissue injury and endotoxin-induced lethality. In this study, we determined the plasma and hepatic tissue levels of HMGB1 in patients with acute liver failure (ALF). PATIENTS AND METHODS We determined the plasma levels of HMGB1 and aspartate aminotransferase (AST) in 7 healthy volunteers (HVs), 40 patients with liver cirrhosis (LC), 37 patients with chronic hepatitis (CH), 18 patients with severe acute hepatitis (AH), and 14 patients with fulminant hepatitis (FH). The 14 patients with FH were divided into two subgroups depending upon the history of plasma exchange (PE) before their plasma sample collection. The hepatic levels of HMGB1 were measured in tissue samples from 3 patients with FH who underwent living-donor liver transplantation and from 3 healthy living donors. Hepatic tissue samples were also subjected to immunohistochemical examination for HMGB1. RESULTS The plasma levels of HMGB1 (ng/ml) were higher in patients with liver diseases, especially in FH patients with no history of PE, than in HVs (0.3 ± 0.3 in HVs, 4.0 ± 2.0 in LC, 5.2 ± 2.6 in CH, 8.6 ± 4.8 in severe AH, 7.8 ± 2.7 in FH with a history of PE, and 12.5 ± 2.6 in FH with no history of PE, p < 0.05 in each comparison). There was a strong and statistically significant relationship between the mean plasma HMGB1 level and the logarithm of the mean AST level (R = 0.900, p < 0.05). The hepatic tissue levels of HMGB1 (ng/mg tissue protein) were lower in patients with FH than in healthy donors (539 ± 116 in FH vs. 874 ± 81 in healthy donors, p < 0.05). Immunohistochemical staining for HMGB1 was strong and clear in the nuclei of hepatocytes in liver sections from healthy donors, but little staining in either nuclei or cytoplasm was evident in specimens from patients with FH. CONCLUSION We confirmed that plasma HMGB1 levels were increased in patients with ALF. Based on a comparison between HMGB1 contents in normal and ALF livers, it is very likely that HMGB1 is released from injured liver tissue.
Collapse
Affiliation(s)
- G Oshima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Adebayo D, Mookerjee RP, Jalan R. Mechanistic biomarkers in acute liver injury: are we there yet? J Hepatol 2012; 56:1003-1005. [PMID: 22322232 DOI: 10.1016/j.jhep.2012.01.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/04/2022]
Affiliation(s)
- Danielle Adebayo
- Liver Failure Group, UCL Institute of Hepatology, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, United Kingdom
| | - Rajeshwar P Mookerjee
- Liver Failure Group, UCL Institute of Hepatology, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, United Kingdom
| | - Rajiv Jalan
- Liver Failure Group, UCL Institute of Hepatology, UCL Medical School, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, United Kingdom.
| |
Collapse
|
26
|
Kikuchi K, Uchikado H, Miura N, Morimoto Y, Ito T, Tancharoen S, Miyata K, Sakamoto R, Kikuchi C, Iida N, Shiomi N, Kuramoto T, Miyagi N, Kawahara KI. HMGB1 as a therapeutic target in spinal cord injury: A hypothesis for novel therapy development. Exp Ther Med 2011; 2:767-770. [PMID: 22977572 DOI: 10.3892/etm.2011.310] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/29/2011] [Indexed: 12/20/2022] Open
Abstract
Historically, clinical outcomes following spinal cord injury (SCI) have been dismal. Severe SCI leads to devastating neurological deficits, and there is no treatment available that restores the injury-induced loss of function to a degree that an independent life can be guaranteed. To address all the issues associated with SCI, a multidisciplinary approach is required, as it is unlikely that a single approach, such as surgical intervention, pharmacotherapy or cellular transplantation, will suffice. High mobility group box 1 (HMGB1) is an inflammatory cytokine. Various studies have shown that HMGB1 plays a critical role in SCI and that inhibition of HMGB1 release may be a novel therapeutic target for SCI and may support spinal cord repair. In addition, HMGB1 has been associated with graft rejection in the early phase. Therefore, HMGB1 may be a promising therapeutic target for SCI transplant patients. We hypothesize that inhibition of HMGB1 release rescues patients with SCI. Taken together, our findings suggest that anti-HMGB1 monoclonal antibodies or short hairpin RNA-mediated HMGB1 could be administered for spinal cord repair in SCI patients.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Department of Neurosurgery, Yame Public General Hospital, Yame 834-0034
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
HMGB1 cytoplasmic translocation in patients with acute liver failure. BMC Gastroenterol 2011; 11:21. [PMID: 21406085 PMCID: PMC3063205 DOI: 10.1186/1471-230x-11-21] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 03/15/2011] [Indexed: 12/12/2022] Open
Abstract
Background High-mobility group box 1 (HMGB1) is a late mediator of lethal systemic inflammation. Acute liver failure (ALF) has been shown to trigger systemic inflammation in clinical and animal studies. To evaluate the possibility of HMGB1 cytoplasmic translocation in ALF, we determined whether HMGB1 is released in hepatocytes and end organ in patients with liver failure/injury. Methods HepG2 cell were stimulated with LPS or TNF-α, the increase of HMGB1 extracellularly in the culture medium and intracellularly in various cellular fractions were determined by western blot or immunocytochemistry. To observe sub-cellular location of HMGB1 in hepatocytes, liver specimens were obtained from 6 patients with ALF caused by HBV infection, 10 patients with chronic viral hepatitis B, 6 healthy controls, as well as animals model of ALF by intraperitoneal administration of D-GalN (600 mg/kg) and LPS (0.5 mg/kg). Results In HepG2 cell culture, LPS or TNF actively induced HMGB1 cytoplasmic translocation and release in a time- and dose-dependent fashion. In animal model of ALF, cytoplasmic HMGB1 translocation was observed in hepatocyts as early as 3 hours post onset of ALF. In patients with ALF caused by HBV infection, cytoplasmic HMGB1 translocation was similarly observed in some hepatocytes of the liver specimen. Conclusions Cytoplasmic HMGB1 translocation may occur during ALF, which may potentially contribute to the pathogenesis of liver inflammatory diseases.
Collapse
|
28
|
What's New in Shock, December 2010? Shock 2010; 34:545-7. [PMID: 21079492 DOI: 10.1097/shk.0b013e3181f9e4ae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|