351
|
Gou X, Ying J, Yue Y, Qiu X, Hu P, Qu Y, Li J, Mu D. The Roles of High Mobility Group Box 1 in Cerebral Ischemic Injury. Front Cell Neurosci 2020; 14:600280. [PMID: 33384585 PMCID: PMC7770223 DOI: 10.3389/fncel.2020.600280] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that plays an important role in stabilizing nucleosomes and DNA repair. HMGB1 can be passively released from necrotic neurons or actively secreted by microglia, macrophages/monocytes, and neutrophils. Cerebral ischemia is a major cause of mortality and disability worldwide, and its outcome depends on the number of neurons dying due to hypoxia in the ischemic area. HMGB1 contributes to the pathogenesis of cerebral ischemia via mediating neuroinflammatory responses to cerebral ischemic injury. Extracellular HMGB1 regulates many neuroinflammatory events by interacting with its different cell surface receptors, such as receptors for advanced glycation end products, toll-like receptor (TLR)-2, and TLR-4. Additionally, HMGB1 can be redox-modified, thus exerting specific cellular functions in the ischemic brain and has different roles in the acute and late stages of cerebral ischemic injury. However, the role of HMGB1 in cerebral ischemia is complex and remains unclear. Herein, we summarize and review the research on HMGB1 in cerebral ischemia, focusing especially on the role of HMGB1 in hypoxic ischemia in the immature brain and in white matter ischemic injury. We also outline the possible mechanisms of HMGB1 in cerebral ischemia and the main strategies to inhibit HMGB1 pertaining to its potential as a novel critical molecular target in cerebral ischemic injury.
Collapse
Affiliation(s)
- Xiaoyun Gou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Peng Hu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Jinhui Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
352
|
Sun YD, Zhang H, Chen YQ, Wu CX, Zhang JB, Xu HR, Liu JZ, Han JJ. HMGB1, the Next Predictor of Transcatheter Arterial Chemoembolization for Liver Metastasis of Colorectal Cancer? Front Oncol 2020; 10:572418. [PMID: 33473353 PMCID: PMC7812918 DOI: 10.3389/fonc.2020.572418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
HMGB1 is an important mediator of inflammation during ischemia-reperfusion injury on organs. The serum expression of HMGB1 was increased significantly on the 1st day after TACE and decreased significantly which was lower on the 30th day after TACE. Tumor markers of post-DEB-TACE decreased significantly. The correlational analysis showed that patients with low HMGB1 expression had lower risks of fever and liver injury compared those with the higher expression, while the ORR is relatively worse. Patients with lower expression of HMGB1 had longer PFS, better efficacy, and higher quality of life. With the high post-expression, the low expression had lower incidence of fever and liver injury too. There was no statistical difference in the one-year survival among the different groups. The quality of life of all patients was improved significantly. The over-expression of HMGB1 in LMCRC is an adverse prognostic feature and a positive predictor of response to TACE.
Collapse
Affiliation(s)
- Yuan-dong Sun
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Hao Zhang
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Ye-qiang Chen
- Maternal and Child Health Care Hospital of Shandong Province, Ji’nan, China
| | - Chun-xue Wu
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
- School of Medicine and Life Sciences, University of Ji’nan-Shandong Academy of Medical Sciences, Ji’nan, China
| | - Jian-bo Zhang
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Hui-rong Xu
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Jing-zhou Liu
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| | - Jian-jun Han
- Interventional Medicine Department, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, China
| |
Collapse
|
353
|
Chen R, Huang Y, Quan J, Liu J, Wang H, Billiar TR, Lotze MT, Zeh HJ, Kang R, Tang D. HMGB1 as a potential biomarker and therapeutic target for severe COVID-19. Heliyon 2020; 6:e05672. [PMID: 33313438 PMCID: PMC7720697 DOI: 10.1016/j.heliyon.2020.e05672] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/26/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has attracted global attention due to its rapid spread around the world with substantial morbidity and associated mortality. Severe COVID-19 can be complicated by the acute respiratory distress syndrome, sepsis and septic shock leading to death. These complications are thought to result from an overactivation of the immune system, leading to a cytokine storm syndrome associated with multiple organ failure. Here, we report that high mobility group box 1 (HMGB1), a prototypical damage-associated molecular pattern (DAMP) and a central mediator of lethal inflammation, could be a potential target for innovative therapeutic strategies for COVID-19. Serum HMGB1 in severe COVID-19 patients is elevated (189.40 ± 140.88 ng/ml). Exogenous HMGB1 induces the expression of SARS-CoV-2 entry receptor ACE2 in alveolar epithelial cells in an AGER-dependent manner. Importantly, genetic (using AGER siRNA) or pharmacological (using glycyrrhizin, chloroquine, hydroxychloroquine, and FPS-ZM1) inhibition of the HMGB1-AGER pathway blocks ACE2 expression. Thus, HMGB1 inhibitors are likewise promising drug candidates for the treatment of patients suffering from COVID-19.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jun Quan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangdong 510600, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, North Shore University Hospital, Feinstein Institute for Medical Research, Manhasset, New York 11030, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15219, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
354
|
Bailly C, Vergoten G. Interaction of fumigaclavine C with High Mobility Group Box 1 protein (HMGB1) and its DNA complex: A computational approach. Comput Biol Chem 2020; 89:107409. [DOI: 10.1016/j.compbiolchem.2020.107409] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/20/2020] [Accepted: 10/25/2020] [Indexed: 12/28/2022]
|
355
|
Bailly C, Vergoten G. Esculentosides: Insights into the potential health benefits, mechanisms of action and molecular targets. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 79:153343. [PMID: 33002830 DOI: 10.1016/j.phymed.2020.153343] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 08/14/2020] [Accepted: 09/10/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Esculentosides and related phytolaccosides form a group of oleanene-type saponins isolated from plants of the Phytolaccaceae family, essentially Phytolacca esculenta, P. americana and P. acinosa. This chemical family offers a diversity of glycosylated compounds, including molecules with a mono-, di- or tri-saccharide unit at position C-3, and with or without a glucose residue at position C-28. The esculentosides, which derive essentially from the sapogenin jaligonic acid or its 30-methyl ester phytolaccagenin, exhibit anti-inflammatory, antifungal and anticancer activities. PURPOSE The objective of the review was to identify the 26 esculentosides (ES) and phytolaccosides known to date, including 16 monodesmosidic and 10 bidesmosidic saponins, and to review their pharmacological properties and molecular targets. METHODOLOGY The retrieval of potentially relevant studies was done by systematically searching of scientific databases like Google Scholar and PubMed in January-May 2020. The main keywords used as search terms were related to esculentosides, phytolaccosides and Phytolaccaceae. The systematic search retrieved about 110 papers that were potentially relevant and after an abstract-based selection, 68 studies were analyzed in details and discussed. RESULTS The structural relationship between the compounds and their sapogenin precursors has been studied. In addition, the pharmacological properties of the main ES, such as ES-A, -B and -H, have been analyzed to highlight their mode of action and potential targets. ES-A is a potent inhibitor of the release of cytokines and this anti-inflammatory activity contributes to the anticancer effects observed in vitro and in vivo. Potential molecular targets of ES-A/B include the enzymes cyclooxygenase 2 (COX-2) and casein kinase 2 (CK2). In addition, the targeting of the protein high-mobility group box 1 (HGMB1) by ES-A/B is proposed, based on molecular modeling and the structural analogy with the related saponin glycyrrhizin, a potent HGMB1 alarmin inhibitor. CONCLUSION More work is needed to properly characterize the molecular targets but otherwise compounds like ES-A and ES-H emerge as potent anti-inflammatory and anticancer agents and ES-B as an antifungal agent. A preclinical development of these three compounds should be considered.
Collapse
Affiliation(s)
| | - Gérard Vergoten
- University of Lille, Inserm, INFINITE - U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, BP-83, F-59006, Lille, France
| |
Collapse
|
356
|
Kaya MA, Erin N, Bozkurt O, Erkek N, Duman O, Haspolat S. Changes of HMGB-1 and sTLR4 levels in cerebrospinal fluid of patients with febrile seizures. Epilepsy Res 2020; 169:106516. [PMID: 33276244 DOI: 10.1016/j.eplepsyres.2020.106516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/23/2020] [Accepted: 11/26/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Fever-induced inflammatory processes and pro-inflammatory cytokines have gained importance in recent years in the pathogenesis of febrile convulsion. Increased levels of HMGB1 (high mobility group box 1), one of the most important pro-inflammatory cytokines, are associated with prolongation of seizure duration, recurrence of seizures and the development of epilepsy. Changes in the sTLR4 level (soluble toll-like receptor 4) in the cerebrospinal fluid (CSF) are thought to be associated with memory and learning functions. In our study, we aimed to evaluate changes in HMGB1 and sTLR4 levels in patients who had febrile seizures between 6 months and 6 years. METHODS Forty patients who were admitted to Akdeniz University Medical Faculty Hospital between April 2016 and April 2018 with a complaint of febrile seizure and 45 patients whose CSF samples were taken for complaints other than febrile convulsion (control group) were included in our study. RESULTS Comparison of the CSF HMGB1 levels of the febrile convulsion group and control group revealed a statistically significant increase in patients with febrile convulsions (p: 0.001). Comparison of the subgroups revealed that the mean value of CSF HMGB1 level was highest in the complex FS group with a mean value of 3363.9 ± 835,47 pg/mL. Comparison of the patient and control groups revealed that the changes in CSF sTLR4 levels were not statistically significant. CONCLUSION HMGB1 level, a key inflammatory molecule, was significantly higher in the CSF of children with febrile seizures. Our data suggest that the HMGB1 network may contribute to the generation of febrile seizures in children.
Collapse
Affiliation(s)
- Mehmet Akif Kaya
- Department of Pediatrics of Medicine, Akdeniz University Hospital, Antalya, Turkey
| | - Nuray Erin
- Department of Medical Pharmacology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Oznur Bozkurt
- Department of Pediatric Neurology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Nilgun Erkek
- Department of Pediatric Emergency, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Ozgur Duman
- Department of Pediatric Neurology, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Senay Haspolat
- Department of Pediatric Neurology, Faculty of Medicine, Akdeniz University, Antalya, Turkey.
| |
Collapse
|
357
|
Niu M, Jiang Z, Xin X, Zhu J, Yang J, Diao M, Qi G, Qi B. Effect of HMGB1 on monocyte immune function in respiratory syncytial virus bronchiolitis. Exp Ther Med 2020; 21:75. [PMID: 33365075 PMCID: PMC7716648 DOI: 10.3892/etm.2020.9507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022] Open
Abstract
Expression of high mobility group protein box 1 (HMGB1) in children with respiratory syncytial virus bronchiolitis and its effect on the inflammatory function of monocytes were investigated. A total of 30 cases of respiratory syncytial viral bronchitis and 30 cases of healthy persons from physical examination were collected from January 2017 to September 2019 in the pediatric department of Xuzhou Children's Hospital, Xuzhou Medical University. HMGB1 expression level in plasma was detected by ELISA. All participants in the study were followed up for 18 months. Human recombinant respiratory syncytial virus (RSV)-A2 virus was used to infect human bronchial epithelial cell line 16HBE, and cell culture supernatant was collected to detect HMGB1. Transwell plate was used to co-culture infected or no-infection groups of epithelial cells and monocytes THP-1. Western blot was used to detect the level of Toll-like receptor (TLR)4 and TLR7 in monocytes. HMGB1 expression level in peripheral blood of children with bronchiolitis was significantly increased compared with that in healthy controls (P<0.0001), and was significantly correlated with the severity of the children's condition (P<0.01). The expression level of HMGB1 was significantly correlated with the number of monocytes, lymphocytes and CRP expression level. HMGB1 was also significantly increased in cell culture supernatant compared with no-infection group (P<0.0001). TLR4 expression in monocytes could be activated by the virus infected cell lines. Follow-up results showed that children with bronchiolitis had a higher incidence of asthma within 18 months (P<0.05). The independent risk factors for children to develop asthma were age, number of monocytes and HMGB1 level. HMGB1 is highly expressed in peripheral blood of children with respiratory syncytial virus bronchitis, and RSV epithelial cells can activate TLR4 expression in monocytes, suggesting that HMGB1 plays an important role in monocyte mediated immune inflammation. HMGB1 expression level is related to the development of asthma in children, which is of great significance for understanding the pathogenesis of bronchiolitis and suggesting the prognosis of children.
Collapse
Affiliation(s)
- Mingyang Niu
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Zhen Jiang
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xin Xin
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Junling Zhu
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Jia Yang
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Min Diao
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Gongjian Qi
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Boxiang Qi
- Department of Critical Care Medicine, Xuzhou Children's Hospital, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
358
|
Niu L, Yang W, Duan L, Wang X, Li Y, Xu C, Liu C, Zhang Y, Zhou W, Liu J, Zhao Q, Han Y, Hong L, Fan D. Biological functions and theranostic potential of HMGB family members in human cancers. Ther Adv Med Oncol 2020; 12:1758835920970850. [PMID: 33224279 PMCID: PMC7659026 DOI: 10.1177/1758835920970850] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/08/2020] [Indexed: 12/14/2022] Open
Abstract
The high mobility group box (HMGB) protein family consists of four members: HMGB1, 2, 3, and 4. They share similar amino acid sequences and identical functional regions, especially HMGB1, 2, and 3. The homology in structure may lead to similarity in function. In fact, though their targets may be different, they all possess the fundamental function of binding and distorting target DNAs. However, further research confirmed they are distributed differently in tissues and involved in various distinct physiological and pathological cellular processes, including cell proliferation, division, migration, and differentiation. Recently, the roles of HMGB family members in carcinogenesis has been widely investigated; however, systematic discussion on their functions and clinical values in malignant tumors is limited. In this review, we mainly review and summarize recent advances in knowledge of HMGB family members in terms of structure, distribution, biochemical cascades, and specific mechanisms regarding tumor progression. Importantly, the diagnostic, prognostic, and therapeutic value of these proteins in cancers is discussed. Finally, we envisage the orientation and challenges of this field in further studies.
Collapse
Affiliation(s)
- Liaoran Niu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wanli Yang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Lili Duan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiaoqian Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yiding Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Chengchao Xu
- 94719 Military Hospital, Ji'an, Jiangxi Province, China
| | - Chao Liu
- School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yujie Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wei Zhou
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jinqiang Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Qingchuan Zhao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yu Han
- Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, 710032, China
| | - Liu Hong
- State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Shaanxi Province, 710032, China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
359
|
Molecular Correlates of Hemorrhage and Edema Volumes Following Human Intracerebral Hemorrhage Implicate Inflammation, Autophagy, mRNA Splicing, and T Cell Receptor Signaling. Transl Stroke Res 2020; 12:754-777. [PMID: 33206327 PMCID: PMC8421315 DOI: 10.1007/s12975-020-00869-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022]
Abstract
Intracerebral hemorrhage (ICH) and perihematomal edema (PHE) volumes are major determinants of ICH outcomes as is the immune system which plays a significant role in damage and repair. Thus, we performed whole-transcriptome analyses of 18 ICH patients to delineate peripheral blood genes and networks associated with ICH volume, absolute perihematomal edema (aPHE) volume, and relative PHE (aPHE/ICH; rPHE). We found 440, 266, and 391 genes correlated with ICH and aPHE volumes and rPHE, respectively (p < 0.005, partial-correlation > |0.6|). These mainly represented inflammatory pathways including NF-κB, TREM1, and Neuroinflammation Signaling-most activated with larger volumes. Weighted Gene Co-Expression Network Analysis identified seven modules significantly correlated with these measures (p < 0.05). Most modules were enriched in neutrophil, monocyte, erythroblast, and/or T cell-specific genes. Autophagy, apoptosis, HIF-1α, inflammatory and neuroinflammatory response (including Toll-like receptors), cell adhesion (including MMP9), platelet activation, T cell receptor signaling, and mRNA splicing were represented in these modules (FDR p < 0.05). Module hub genes, potential master regulators, were enriched in neutrophil-specific genes in three modules. Hub genes included NCF2, NCF4, STX3, and CSF3R, and involved immune response, autophagy, and neutrophil chemotaxis. One module that correlated negatively with ICH volume correlated positively with rPHE. Its genes and hubs were enriched in T cell-specific genes including hubs LCK and ITK, Src family tyrosine kinases whose modulation improved outcomes and reduced BBB dysfunction following experimental ICH. This study uncovers molecular underpinnings associated with ICH and PHE volumes and pathophysiology in human ICH, where knowledge is scarce. The identified pathways and hub genes may represent novel therapeutic targets.
Collapse
|
360
|
Increase in Epithelial Permeability and Cell Metabolism by High Mobility Group Box 1, Inflammatory Cytokines and TPEN in Caco-2 Cells as a Novel Model of Inflammatory Bowel Disease. Int J Mol Sci 2020; 21:ijms21228434. [PMID: 33182652 PMCID: PMC7696423 DOI: 10.3390/ijms21228434] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/24/2022] Open
Abstract
High mobility group box 1 protein (HMGB1) is involved in the pathogenesis of inflammatory bowel disease (IBD). Patients with IBD develop zinc deficiency. However, the detailed roles of HMGB1 and zinc deficiency in the intestinal epithelial barrier and cellular metabolism of IBD remain unknown. In the present study, Caco-2 cells in 2D culture and 2.5D Matrigel culture were pretreated with transforming growth factor-β (TGF-β) type 1 receptor kinase inhibitor EW-7197, epidermal growth factor receptor (EGFR) kinase inhibitor AG-1478 and a TNFα antibody before treatment with HMGB1 and inflammatory cytokines (TNFα and IFNγ). EW-7197, AG-1478 and the TNFα antibody prevented hyperpermeability induced by HMGB1 and inflammatory cytokines in 2.5D culture. HMGB1 affected cilia formation in 2.5D culture. EW-7197, AG-1478 and the TNFα antibody prevented the increase in cell metabolism induced by HMGB1 and inflammatory cytokines in 2D culture. Furthermore, ZnSO4 prevented the hyperpermeability induced by zinc chelator TPEN in 2.5D culture. ZnSO4 and TPEN induced cellular metabolism in 2D culture. The disruption of the epithelial barrier induced by HMGB1 and inflammatory cytokines contributed to TGF-β/EGF signaling in Caco-2 cells. The TNFα antibody and ZnSO4 as well as EW-7197 and AG-1478 may have potential for use in therapy for IBD.
Collapse
|
361
|
Sharma A, Kaur S, Sarkar M, Sarin BC, Changotra H. The AGE-RAGE Axis and RAGE Genetics in Chronic Obstructive Pulmonary Disease. Clin Rev Allergy Immunol 2020; 60:244-258. [PMID: 33170477 DOI: 10.1007/s12016-020-08815-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2020] [Indexed: 12/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a heterogeneous group of lung diseases limiting the airflow due to narrowing of airways, chronic bronchitis and emphysema that leads to difficulties in breathing. Chronic inflammation is another important characteristic of COPD which leads to immune cell infiltration and helps in the alveolar destruction. Pathology of COPD is driven by various environmental and genetic factors. COPD is mainly associated with the inhalation of toxic agents mainly the cigarette smoke. Receptor for advanced glycation end products (RAGE) has emerged as a pattern recognition receptor and is a multiligand receptor expressed moderately in various cells, tissues and highly in the lungs throughout life. RAGE recognizes various ligands produced by cigarette smoke and its role has been implicated in the pathogenesis of COPD. RAGE ligands have been reported to accumulate in the lungs of patients with COPD. RAGE is a membrane receptor but its truncated form i.e. soluble RAGE (sRAGE) mainly functions as a contender of RAGE and inhibits various RAGE dependent cell signalling. Among the various ligands of RAGE, advanced glycation end products (AGEs) are majorly linked with COPD. Accumulated AGE triggers downstream RAGE-AGE axis in COPD. Moreover, RAGE genetics has long been known to play a vital role in the pathology of various airway diseases including COPD and this gene contains an associated locus. A reliable biomarker is needed for the management of this disease. sRAGE has an inverse correlation with the RAGE showed its importance as a valuable marker in COPD. This review is focused on the role of RAGE, sRAGE, RAGE axis and RAGE genetics in COPD.
Collapse
Affiliation(s)
- Ambika Sharma
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Sargeet Kaur
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India
| | - Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla, Himachal Pradesh, 171 001, India
| | - B C Sarin
- Department of Chest and TB, Sri Guru Ram Das Institute of Medical Sciences and Research, Vallah, Amritsar, 143 501, India
| | - Harish Changotra
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan, Himachal Pradesh, 173 234, India.
| |
Collapse
|
362
|
Tsai CY, Chou HC, Chen CM. Perinatal nicotine exposure alters lung development and induces HMGB1-RAGE expression in neonatal mice. Birth Defects Res 2020; 113:570-578. [PMID: 33166075 DOI: 10.1002/bdr2.1840] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 10/17/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Maternal nicotine exposure during gestation and lactation adversely affect lung development of their children. High-mobility group box 1 (HMGB1) is the encoded non-histone, nuclear DNA-binding protein that regulates transcription, and is involved in organization of DNA. Receptors for advanced glycation end products (RAGE) is a receptor for HMGB1 and activates nuclear factor-κB (NF-κB) signaling. Animal and human studies have found cigarette smoke exposure upregulates RAGE expression, suggesting that the HMGB1-RAGE pathway might be involved in maternal nicotine-induced lung injury. METHODS This study evaluated prenatal and perinatal nicotine effects on lung development and HMGB1 and RAGE expression in mouse offspring. Nicotine was administered to pregnant mice by subcutaneous osmotic mini-pump at a dose of 6 mg kg-1 day-1 from gestational Day 14 to birth (prenatal) or to postnatal Day 21 (perinatal). A control group received an equal volume of saline by the same route. Three study groups were obtained: prenatal normal saline (NS), prenatal nicotine, and perinatal nicotine groups. The mice were euthanized on postnatal Day 21, and the lung tissues were collected for histological and Western blot analyses. RESULTS Mice exposed to prenatal nicotine exhibited significantly higher lung mean chord length and oxidative stress marker 8-hydroxy-2'-deoxyguanosine and NF-κB expression compared to mice exposed to NS. Perinatal nicotine exposure further enhanced these harmful effects. These perinatal nicotine effects on lung development were associated with increased HMGB1 and RAGE expression. CONCLUSIONS HMGB1-RAGE pathway may be involved in the pathogenesis of altered lung development induced by perinatal nicotine exposure.
Collapse
Affiliation(s)
- Chin-Yen Tsai
- Department of Pediatrics, Yuan's General Hospital, Kaohsiung, Taiwan
| | - Hsiu-Chu Chou
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Ming Chen
- Department of Pediatrics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pediatrics, Taipei Medical University Hospital, Taipei, Taiwan
| |
Collapse
|
363
|
Wang Y, Yu Z, Yuan H, Chen H, Xie N, Wang Z, Sun Q, Zhang W. Structure-based design of glycyrrhetinic acid derivatives as potent anti-sepsis agents targeting high-mobility group box-1. Bioorg Chem 2020; 106:104461. [PMID: 33223202 DOI: 10.1016/j.bioorg.2020.104461] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/15/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
Novel Glycyrrhetinic Acid (GA) derivatives with fused heterocycles on A ring were structure-based designed and synthesized. Their potential anti-inflammatory effects were investigated by a classical LPS stimulated macrophage model. Surface plasmon resonance (SPR) was used to verify the binding of GA analogues with HMGB1. A preliminary structure-activity relationship was summarized and an analogue GA-60 with ortho-methoxybenzyl pyrozole showed stronger anti-inflammatory effect and higher affinity for HMGB1 with a Kd value of 12.5 μM. In addition, this compound exhibited excellent inhibitory functions on NO (96%), TNF-α (94%), and IL-6 (100%), by interfering with phosphorylation of p38, ERK, JNK MAPKs, as well as that of NF-κB p65 and IKKα/β. Moreover, GA-60 extended the survival of either the classic CLP-induced or LPS-induced sepsis mouse models. Molecular modeling predictions further supported these findings, clearly indicating that inhibiting HMGB1 release, using fused heterocyclic GA derivatives, is a promising strategy for treatment of sepsis.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zongmin Yu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; Department of Medical Service Training Center, No. 965 Hospital, Joint Logistics Support Force of PLA, Jilin 132011, China
| | - Hu Yuan
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Hao Chen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China; State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Ning Xie
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Jiangxi 341000, China
| | - Zhibin Wang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| | - Qingyan Sun
- Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China.
| | - Weidong Zhang
- Interdisciplinary Science Research Institute, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, China; School of Pharmacy, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
364
|
Wang R, Xiao Y, Zhang Q, Bai L, Wang W, Zhao S, Liu E. Upregulation of HMGB1 secretion in lungs of pigs infected by highly pathogenic porcine reproductive and respiratory syndrome virus. Vet Microbiol 2020; 252:108922. [PMID: 33221069 DOI: 10.1016/j.vetmic.2020.108922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/01/2020] [Indexed: 01/18/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) remains a major driver for substantial economic losses to the swine industry across the world. Pulmonary inflammatory injury is a common manifestation in infected pigs. Previous studies reported that PRRS virus (PRRSV) induces secretion of high mobility group box 1 (HMGB1), a proinflammatory factor, in cultured cells. The objective of this study was to evaluate whether HMGB1 secretion is associated with PRRSV-induced pulmonary inflammatory responses in the early stage of infection in vivo. Three-week-old piglets were inoculated with either HuN4, a highly pathogenic PRRSV (HP-PRRSV) strain, or CH1R, an avirulent PRRSV vaccine strain. Necropsy was performed at 7 days post-infection. The results showed that HuN4 significantly induced the secretion of HMGB1 and inflammatory cytokines (IL-1β, IL-6) into the bronchoalveolar lavage fluid (BALF). HuN4 infection induced severe interstitial pneumonia in the pigs. In contrast, pigs infected by CH1R had mild lung inflammation with minimal HMGB1 secretion. In addition, high viral load of HuN4 was detected in both pulmonary alveolar macrophages (PAMs) and lung tissue, whereas viral RNA of CH1R was confined to PAMs. In consistent with the pneumonia development, HuN4 induced inflammatory cytokines in both PAMs and lung tissue, while their expression in CH1R-infected pigs confined only to PAMs. These results indicate that the HuN4-induced HMGB1 secretion into BALF may enhance the pulmonary inflammatory response and exacerbate the lung injury. This finding provides insights to the inflammatory response and pathogenesis of the HP-PRRSV infection.
Collapse
Affiliation(s)
- Rong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China.
| | - Yueqiang Xiao
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, Shandong, China
| | - Qian Zhang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, Shandong, China
| | - Liang Bai
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Weirong Wang
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Sihai Zhao
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Enqi Liu
- Laboratory Animal Center, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| |
Collapse
|
365
|
Prantner D, Nallar S, Vogel SN. The role of RAGE in host pathology and crosstalk between RAGE and TLR4 in innate immune signal transduction pathways. FASEB J 2020; 34:15659-15674. [PMID: 33131091 DOI: 10.1096/fj.202002136r] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
Although the innate immune receptor protein, Receptor for Advanced Glycation End products (RAGE), has been extensively studied, there has been renewed interest in RAGE for its potential role in sepsis, along with a host of other inflammatory diseases of chronic, noninfectious origin. In contrast to other innate immune receptors, for example, Toll-like receptors (TLRs), that recognize ligands derived from pathogenic organisms that are collectively known as "pathogen-associated molecular patterns" (PAMPs) or host-derived "damage-associated molecular patterns" (DAMPs), RAGE has been shown to recognize a broad collection of DAMPs exclusively. Historically, these DAMPs have been shown to be pro-inflammatory in nature. Early studies indicated that the adaptor molecule, MyD88, might be important for this change. More recent studies have explored further the mechanisms underlying this inflammatory change. Overall, the newer results have shown that there is extensive crosstalk between RAGE and TLRs. The three canonical RAGE ligands, Advanced Glycation End products (AGEs), HMGB1, and S100 proteins, have all been shown to activate both TLRs and RAGE to varying degrees in order to induce inflammation in in vitro models. As with any field that delves deeply into innate signaling, obstacles of reagent purity may be a cause of some of the discrepancies in the literature, and we have found that commercial antibodies that have been widely used exhibit a high degree of nonspecificity. Nonetheless, the weight of published evidence has led us to speculate that RAGE may be physically interacting with TLRs on the cell surface to elicit inflammation via MyD88-dependent signaling.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Shreeram Nallar
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
366
|
Chen L, Zhu H, Su S, Harshfield G, Sullivan J, Webb C, Blumenthal JA, Wang X, Huang Y, Treiber FA, Kapuku G, Li W, Dong Y. High-Mobility Group Box-1 Is Associated With Obesity, Inflammation, and Subclinical Cardiovascular Risk Among Young Adults: A Longitudinal Cohort Study. Arterioscler Thromb Vasc Biol 2020; 40:2776-2784. [PMID: 32814439 PMCID: PMC7578115 DOI: 10.1161/atvbaha.120.314599] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE We aimed to characterize circulating HMGB1 (high-mobility group box-1) levels, one of the better-characterized damage-associated molecular patterns, with respect to age, sex, and race in the general population, and investigate the longitudinal associations of HMGB1 with inflammatory markers, obesity, and preclinical markers of cardiovascular disease. Approach and Results: The analyses included 489 participants (50% Blacks, aged 24.6±3.3 years at the first visit) with up to 4 follow-up visits (1149 samples) over a maximum of 8.5 years. Systolic blood pressure, diastolic blood pressure, carotid-femoral pulse wave velocity, and carotid intima-media thickness together with plasma HMGB1, hs-CRP (high-sensitivity C-reactive protein), IFN-γ (interferon-γ), IL-6 (interleukin-6), IL-10 (interleukin-10), and TNF-α (tumor necrosis factor-α) were measured at each visit. At baseline, plasma HMGB1 concentrations were higher in Blacks compared with Whites (3.86 versus 3.20 ng/mL, P<0.001), and in females compared with males (3.75 versus 3.30 ng/mL, P=0.005). HMGB1 concentrations increased with age (P=0.007), and higher levels of obesity measures (P<0.001). Without adjustment for age, sex, race, and body mass index, HMGB1 concentrations were positively associated with hs-CRP, IL-6, TNF-α, systolic blood pressure, diastolic blood pressure, and carotid-femoral pulse wave velocity (P<0.05) but not IL-10, IFN-γ or carotid intima-media thickness. After covariate adjustments, the associations of HMGB1 with hs-CRP, and carotid-femoral pulse wave velocity remained statistically significant (P<0.05). CONCLUSIONS This study demonstrates the age, sex, and race differences in circulating HMGB1. The increasing circulating concentrations of HMGB1 with age suggest a potential role of HMGB1 in the pathogenesis of chronic low-grade inflammation, obesity, and subclinical cardiovascular disease risk.
Collapse
Affiliation(s)
- Li Chen
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Haidong Zhu
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Shaoyong Su
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Gregory Harshfield
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Jennifer Sullivan
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Clinton Webb
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - James A. Blumenthal
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Xiaoling Wang
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ying Huang
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Frank A. Treiber
- College of Nursing, Medical University of South Carolina, Charleston, SC, USA
- College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Gaston Kapuku
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Wenjun Li
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Yanbin Dong
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| |
Collapse
|
367
|
Liu L, Zhang J, Zhang X, Cheng P, Liu L, Huang Q, Liu H, Ren S, Wei P, Wang C, Dou C, Chen L, Liu X, Zhang H, Chen M. HMGB1: an important regulator of myeloid differentiation and acute myeloid leukemia as well as a promising therapeutic target. J Mol Med (Berl) 2020; 99:107-118. [PMID: 33128580 PMCID: PMC7782413 DOI: 10.1007/s00109-020-01998-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
Abstract High mobility group box 1 (HMGB1) is a non-histone nuclear protein which has been intensively studied in various physiological and pathological processes including leukemia. Here in this study, we further demonstrated that HMGB1 presents higher expression in the bone marrow mononuclear cells of acute myeloid leukemia (AML) patients compared with the normal controls and contributes to the AML pathogenesis and progression by inhibiting apoptosis, facilitating proliferation, and inducing myeloid differentiation blockade of AML cells. Mechanistic investigation revealed that transforming growth factor beta-induced (TGFBI) acts as a potential downstream target of HMGB1 and lentivirus-mediated knockdown of TGFBI expression impaired phorbol-12-myristate-13-acetate (PMA) and all-trans retinoic acid (ATRA)–induced myeloid differentiation of AML cell lines. On the other hand, chidamide, an orally histone deacetylase inhibitor, decreases HMGB1 expression significantly in AML cells with concomitant upregulation of TGFBI expression, and confers therapeutic effect on AML by inducing cell differentiation, apoptosis and inhibiting cell proliferation. In conclusion, our findings provide additional insights that HMGB1 is a promising therapeutic target of AML, and also present experimental evidence for the clinical application of chidamide as a novel agent in AML therapy by downregulating HMGB1 expression. Key messages HMGB1 induces cell proliferation and myeloid differentiation blockade and inhibits apoptosis of AML cells. TGFBI acts as a potential target of HMGB1. Chidamide, a selective HDAC inhibitor, confers promising therapeutic effect for AML via downregulating HMGB1 expression.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-020-01998-5.
Collapse
Affiliation(s)
- Lulu Liu
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Jingjing Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Xianning Zhang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Panpan Cheng
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Lei Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Qian Huang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Haihui Liu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Saisai Ren
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Peng Wei
- Department of Radiation Oncology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Cuiling Wang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Cuiyun Dou
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Lulu Chen
- Department of Graduate School, Jining Medical University, Jining, 272000, Shandong Province, China
| | - Xin Liu
- Department of Graduate School, Jining Medical University, Jining, 272000, Shandong Province, China
| | - Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China
| | - Mingtai Chen
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong Province, China.
| |
Collapse
|
368
|
Chen L, Li J, Ye Z, Sun B, Wang L, Chen Y, Han J, Yu M, Wang Y, Zhou Q, Seidler U, Tian D, Xiao F. Anti-High Mobility Group Box 1 Neutralizing-Antibody Ameliorates Dextran Sodium Sulfate Colitis in Mice. Front Immunol 2020; 11:585094. [PMID: 33193406 PMCID: PMC7661783 DOI: 10.3389/fimmu.2020.585094] [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: 07/19/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous nuclear protein in mammals. When released into the extracellular space, it acts as a damage-associated molecular pattern. This study investigates whether increased HMGB1 levels are found in the intestinal mucosa of ulcerative colitis (UC) patients, and whether an anti-HMGB1 neutralizing-antibody (HnAb) can inhibit the intestinal inflammation elicited by dextran sulfate sodium (DSS) in mice. Because toll-like receptor 4 (TLR4) is implicated in HMGB1-mediated immune cell activation, DSS colitis was also elicited in TLR4-deficient mice in the presence and absence of HnAb. The expression of HMGB1 in UC patients was examined. HnAb was administered via intraperitoneal injection to TLR4 deficient mice and their wild-type littermates, both being induced to colitis with DSS. Finally, the protective effect of HnAb and TLR4 deficiency were evaluated. In UC patients, HMGB1 was up-regulated in the inflamed colon. When administered during DSS application, HnAb alleviated the severity of colitis with a lower disease activity index, limited histological damages, and reduced production of proinflammatory cytokines. This antibody also limited colonic barrier loss, decreased colonic lamina propria macrophages and partially reversed the DSS treatment-associated dysbiosis. The protective effect of this antibody was enhanced in TLR4-deficient mice in some aspects, indicating that both additional HMGB1-mediated as well as TLR4-mediated inflammatory signaling pathways were involved in the induction of colitis by DSS. HnAb ameliorated colitis via macrophages inhibition and colonic barrier protection. It may therefore be a novel treatment option in colitis.
Collapse
Affiliation(s)
- Liping Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenghao Ye
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binghua Sun
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.,Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yu Chen
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meiping Yu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wang
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Zhou
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ursula Seidler
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - De'an Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Xiao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
369
|
Sitapara RA, Gauthier AG, Patel VS, Lin M, Zur M, Ashby CR, Mantell LL. The α7 nicotinic acetylcholine receptor agonist GTS-21 improves bacterial clearance in mice by restoring hyperoxia-compromised macrophage function. Mol Med 2020; 26:98. [PMID: 33126860 PMCID: PMC7596622 DOI: 10.1186/s10020-020-00224-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/07/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mechanical ventilation, in combination with supraphysiological concentrations of oxygen (i.e., hyperoxia), is routinely used to treat patients with respiratory distress, such as COVID-19. However, prolonged exposure to hyperoxia compromises the clearance of invading pathogens by impairing macrophage phagocytosis. Previously, we have shown that the exposure of mice to hyperoxia induces the release of the nuclear protein high mobility group box-1 (HMGB1) into the pulmonary airways. Furthermore, extracellular HMGB1 impairs macrophage phagocytosis and increases the mortality of mice infected with Pseudomonas aeruginosa (PA). The aim of this study was to determine whether GTS-21 (3-(2,4-dimethoxybenzylidene) anabaseine), an α7 nicotinic acetylcholine receptor (α7nAChR) agonist, could (1) inhibit hyperoxia-induced HMGB1 release into the airways; (2) enhance macrophage phagocytosis and (3) increase bacterial clearance from the lungs in a mouse model of ventilator-associated pneumonia. METHOD GTS-21 (0.04, 0.4, and 4 mg/kg) or saline were administered by intraperitoneal injection to mice that were exposed to hyperoxia (≥ 99% O2) and subsequently challenged with PA. RESULTS The systemic administration of 4 mg/kg i.p. of GTS-21 significantly increased bacterial clearance, decreased acute lung injury and decreased accumulation of airway HMGB1 compared to the saline control. To determine the mechanism of action of GTS-21, RAW 264.7 cells, a macrophage-like cell line, were incubated with different concentrations of GTS-21 in the presence of 95% O2. The phagocytic activity of macrophages was significantly increased by GTS-21 in a dose-dependent manner. In addition, GTS-21 significantly inhibited the cytoplasmic translocation and release of HMGB1 from RAW 264.7 cells and attenuated hyperoxia-induced NF-κB activation in macrophages and mouse lungs exposed to hyperoxia and infected with PA. CONCLUSIONS Our results indicate that GTS-21 is efficacious in improving bacterial clearance and reducing acute lung injury via enhancing macrophage function by inhibiting the release of nuclear HMGB1. Therefore, the α7nAChR represents a possible pharmacological target to improve the clinical outcome of patients on ventilators by augmenting host defense against bacterial infections.
Collapse
Affiliation(s)
- Ravikumar A Sitapara
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Alex G Gauthier
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Vivek S Patel
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Mosi Lin
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Michelle Zur
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Lin L Mantell
- Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 8000 Utopia Parkway, Queens, NY, 11439, USA. .,The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, NY, 11030, USA.
| |
Collapse
|
370
|
Ghoneim MES, Abdallah DM, Shebl AM, El-Abhar HS. The interrupted cross-talk of inflammatory and oxidative stress trajectories signifies the effect of artesunate against hepatic ischemia/reperfusion-induced inflammasomopathy. Toxicol Appl Pharmacol 2020; 409:115309. [PMID: 33130049 DOI: 10.1016/j.taap.2020.115309] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
The antimalarial drug artesunate (Art) has proven its beneficial effects against ischemia/reperfusion (I/R) injury in diverse organs, but its potential role against hepatic I/R is still obscure. This study, hence, examined whether treatment with Art alone or in combination with rapamycin (Rapa), an mTOR inhibitor, can ameliorate hepatic I/R injury via targeting the NLRP3 inflammasome signaling pathway. Rats were divided into hepatic sham- and I/R-operated rats. The latter were either left untreated (I/R group) or treated with Art, Rapa, or their combination. On the molecular level, all treatment regimens succeeded to hinder inflammasome assembly and activation, assessed as NLRP3, ASC, cleaved caspase-1, caspase-11, N-terminal cleaved gasdermin-D (GSDMD-N), IL-1β, and IL-18. This effect was associated by the inhibition in the harmful signaling pathways HMGB1/RAGE and TLR4/MyD88/TRAF6 to inactivate the transcription factor NF-κB and the production of its pro-inflammatory cytokines IL-1β, IL-18, IL-6, and TNF-α. Additionally, this effect entailed the inhibition of ICAM-1/MPO/ROS cascade, which in turn hampered cell demise induced by apoptosis, manifested as correction of the imbalanced Bcl2/Bax, as well as pyroptosis (LDH, cleaved caspase-1, caspase-11, GSDMD-N, IL-1β, and IL-18), and necrosis. The corrected pathways were reflected on the improved liver function (serum ALT, AST, and LDH) and microscopical hepatic architecture. Noteworthy, the effect of Art on all parameters exceeded significantly that of Rapa and even improved the effect of the latter in the combination group. In conclusion, our results suggest novel roles for Art in abating functional and structural I/R-induced hepatic abnormalities via several traversing cross-talking pathways that succeeded to abate NLRP3 inflammasome and cell death.
Collapse
Affiliation(s)
- Mai El-Sayed Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City (USC), Menoufia, Egypt.
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., 11562 Cairo, Egypt.
| | | | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology and Biochemistry, Faculty of Pharmaceutical Sciences and Pharmaceutical Industries, Future University in Egypt (FUE), 11835 Cairo, Egypt
| |
Collapse
|
371
|
Alarmins and c-Jun N-Terminal Kinase (JNK) Signaling in Neuroinflammation. Cells 2020; 9:cells9112350. [PMID: 33114371 PMCID: PMC7693759 DOI: 10.3390/cells9112350] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/08/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation is involved in the progression or secondary injury of multiple brain conditions, including stroke and neurodegenerative diseases. Alarmins, also known as damage-associated molecular patterns, are released in the presence of neuroinflammation and in the acute phase of ischemia. Defensins, cathelicidin, high-mobility group box protein 1, S100 proteins, heat shock proteins, nucleic acids, histones, nucleosomes, and monosodium urate microcrystals are thought to be alarmins. They are released from damaged or dying cells and activate the innate immune system by interacting with pattern recognition receptors. Being principal sterile inflammation triggering agents, alarmins are considered biomarkers and therapeutic targets. They are recognized by host cells and prime the innate immune system toward cell death and distress. In stroke, alarmins act as mediators initiating the inflammatory response after the release from the cellular components of the infarct core and penumbra. Increased c-Jun N-terminal kinase (JNK) phosphorylation may be involved in the mechanism of stress-induced release of alarmins. Putative crosstalk between the alarmin-associated pathways and JNK signaling seems to be inherently interwoven. This review outlines the role of alarmins/JNK-signaling in cerebral neurovascular inflammation and summarizes the complex response of cells to alarmins. Emerging anti-JNK and anti-alarmin drug treatment strategies are discussed.
Collapse
|
372
|
Ghosh SS, Ghosh S. HMGB1 (High-Mobility Group Box-1): A Common Link Determining the Consequences of Tissue Injury, Sterile/Microbial and Low-Grade Chronic Inflammation. Arterioscler Thromb Vasc Biol 2020; 40:2561-2563. [PMID: 33085519 DOI: 10.1161/atvbaha.120.315189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Siddhartha S Ghosh
- Department of Internal Medicine, VCU Medical Center, Richmond, VA (S.S.G., S.G.)
| | - Shobha Ghosh
- Hunter Homes McGuire VA Medical Center, Richmond, VA (S.G.)
| |
Collapse
|
373
|
Miao J, Ye P, Lan J, Ye S, Zhong J, Gresham A, Li S, You A, Chen X, Liu X, Li H. Paeonol promotes the phagocytic ability of macrophages through confining HMGB1 to the nucleus. Int Immunopharmacol 2020; 89:107068. [PMID: 33091813 DOI: 10.1016/j.intimp.2020.107068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
Phagocytosis is a basic immune response to the pathogens invading. Immunosuppression may occur in diseases like sepsis and cancer, and cause a low phagocytic ability of phagocytes. High mobility group protein B1 (HMGB1) is a DNA chaperone which is closely related to the phagocytosis. Nonetheless, its influence on phagocytosis is still controversial. We found that paeonol could inhibit the translocation of HMGB1 from the nucleus to the cytoplasm, it may have an impact on phagocytosis. In the present study, we performed in vivo and in vitro experiments to investigate the influence of paeonol on phagocytosis. Zymosan was used to test the phagocytic function of macrophages. Our results showed that paeonol promotes the phagocytosis of macrophages through confining HMGB1 to the nucleus. Through interacting with P53, the nuclear HMGB1 keep it in the nucleus and decrease the negative influence of P53 on the phosphorylation of Focal Adhesion Kinase (FAK). The increasing of phosphorylated FAK promotes the formation of pseudopod and enhances the phagocytic ability of macrophages.
Collapse
Affiliation(s)
- Jifei Miao
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peng Ye
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Sen Ye
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jun Zhong
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Siyan Li
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aijia You
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianjie Chen
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liu
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Li
- Research Center of Integrative Medicine, School Basic Medical sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
374
|
Zhou B, Liu J, Zeng L, Zhu S, Wang H, Billiar TR, Kroemer G, Klionsky DJ, Zeh HJ, Jiang J, Tang D, Kang R. Extracellular SQSTM1 mediates bacterial septic death in mice through insulin receptor signalling. Nat Microbiol 2020; 5:1576-1587. [PMID: 33077977 DOI: 10.1038/s41564-020-00795-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022]
Abstract
Sepsis is the most common cause of death for patients in intensive care worldwide due to a dysregulated host response to infection. Here, we investigate the role of sequestosome-1 (SQSTM1/p62), an autophagy receptor that functions as a regulator of innate immunity, in sepsis. We find that lipopolysaccharide elicits gasdermin D-dependent pyroptosis to enable passive SQSTM1 release from macrophages and monocytes, whereas transmembrane protein 173-dependent TANK-binding kinase 1 activation results in the phosphorylation of SQSTM1 at Ser403 and subsequent SQSTM1 secretion from macrophages and monocytes. Moreover, extracellular SQSTM1 binds to insulin receptor, which in turn activates a nuclear factor kappa B-dependent metabolic pathway, leading to aerobic glycolysis and polarization of macrophages. Intraperitoneal injection of anti-SQSTM1-neutralizing monoclonal antibodies or conditional depletion of Insr in myeloid cells using the Cre-loxP system protects mice from lethal sepsis (caecal ligation and puncture or infection by Escherichia coli or Streptococcus pneumoniae) and endotoxaemia. We also report that circulating SQSTM1 and the messenger RNA expression levels of SQSTM1 and INSR in peripheral blood mononuclear cells are related to the severity of sepsis in 40 patients. Thus, extracellular SQSTM1 has a pathological role in sepsis and could be targeted to develop therapies for sepsis.
Collapse
Affiliation(s)
- Borong Zhou
- The Third Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Jiao Liu
- The Third Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China
| | - Ling Zeng
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Shan Zhu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, North Shore University Hospital and the Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guido Kroemer
- Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale U1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Herbert J Zeh
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China.
| | - Daolin Tang
- The Third Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, China. .,Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
375
|
Xu X, Piao HN, Aosai F, Zeng XY, Cheng JH, Cui YX, Li J, Ma J, Piao HR, Jin X, Piao LX. Arctigenin protects against depression by inhibiting microglial activation and neuroinflammation via HMGB1/TLR4/NF-κB and TNF-α/TNFR1/NF-κB pathways. Br J Pharmacol 2020; 177:5224-5245. [PMID: 32964428 DOI: 10.1111/bph.15261] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/19/2020] [Accepted: 09/03/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Arctigenin, a major bioactive component of Fructus arctii, has been reported to have antidepressant-like effects. However, the mechanisms underlying these effects are still unclear. Neuroinflammation can be caused by excessive production of proinflammatory cytokines in microglia via high-mobility group box 1 (HMGB1)/TLR4/NF-κB and TNF-α/TNFR1/NF-κB signalling pathways, leading to depression. In this study, we have investigated the antidepressant mechanism of arctigenin by conducting in vitro and in vivo studies. EXPERIMENTAL APPROACH The effects of chronic unpredictable mild stress (CUMS) on wild-type (WT) and TLR4-/- mice were examined. Antidepressant-like effects of arctigenin were tested using the CUMS-induced model of depression in WT mice. The effects of arctigenin were assessed on the HMGB1/TLR4/NF-κB and TNF-α/TNFR1/NF-κB signalling pathways in the prefrontal cortex (PFC) of mouse brain and HMGB1- or TNF-α-stimulated primary cultured microglia. The interaction between HMGB1 and TLR4 or TNF-α and TNFR1 with or without arctigenin was examined by localized surface plasmon resonance (LSPR) and co-immunoprecipitation assays. KEY RESULTS The immobility times in the tail suspension test (TST) and forced swimming test (FST) were reduced in TLR4-/- mice, compared with WT mice. Arctigenin exhibited antidepressant-like effects. Arctigenin also inhibited microglia activation and inflammatory responses in the PFC of mouse brain. Arctigenin inhibited HMGB1 and TLR4 or TNF-α and TNFR1 interactions, and suppressed both HMGB1/TLR4/NF-κB and TNF-α/TNFR1/NF-κB signalling pathways. CONCLUSIONS AND IMPLICATIONS Arctigenin has antidepressant-like effects by attenuating excessive microglial activation and neuroinflammation through the HMGB1/TLR4/NF-κB and TNF-α/TNFR1/NF-κB signalling pathways. This suggests that arctigenin has potential as a new drug candidate suitable for clinical trials to treat depression.
Collapse
Affiliation(s)
- Xiang Xu
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Hu-Nan Piao
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Fumie Aosai
- Department of Infection and Host Defense, Graduate School of Medicine, Shinshu University, Matsumoto, Japan
| | - Xiao-Yu Zeng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Jia-Hui Cheng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Yue-Xian Cui
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Jing Li
- Department of Neurology, Affliated Hospital of Yanbian University, Yanji, Jilin, China
| | - Juan Ma
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Hu-Ri Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Xuejun Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| | - Lian-Xun Piao
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, Jilin, China
| |
Collapse
|
376
|
Chen L, Chi K, Xiang H, Yang Y. Circ_0032821 Facilitates Gastric Cancer Cell Proliferation, Migration, Invasion and Glycolysis by Regulating MiR-1236-3p/HMGB1 Axis. Cancer Manag Res 2020; 12:9965-9976. [PMID: 33116853 PMCID: PMC7567569 DOI: 10.2147/cmar.s270164] [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: 06/30/2020] [Accepted: 09/10/2020] [Indexed: 01/03/2023] Open
Abstract
Background Circular RNAs (circRNAs) play an essential role in the pathogenesis of malignant tumors, including gastric cancer (GC). However, the effect of circ_0032821 on GC remains largely unknown. Methods QRT-PCR assay was employed to examine the levels of circ_0032821, CEP128 mRNA and miR-1236-3p. RNase R digestion assay was utilized to verify the feature of circ_0032821. Cell Counting Kit-8 (CCK-8) assay and transwell assay were adopted to evaluate cell proliferation and metastasis. The level of glycolysis was evaluated through detecting ECAR, OCR, lactate production, glucose uptake and ATP synthesis. Dual-luciferase reporter assay and RIP assay were conducted to analyze the relationship between miR-1236-3p and circ_0032821 or HMGB1. Western blot assay was adopted for high mobility group box 1 (HMGB1) level. Murine xenograft model assay was utilized for the effect of circ_0032821 in vivo. Results High level of circ_0032821 was observed in GC tissues and cells. Silencing of circ_0032821 markedly repressed cell proliferation, metastasis and glycolysis in GC cells in vitro and blocked tumorigenesis of GC in vivo. For mechanism analysis, circ_0032821 was identified as the sponge of miR-1236-3p and HMGB1 was the target gene of miR-1236-3p. Moreover, miR-1236-3p suppression restored the influences of circ_0032821 deficiency on GC cell proliferation, metastasis and glycolysis. Overexpression of miR-1236-3p relieved the malignant behaviors of GC cells by targeting HMGB1. Conclusion Circ_0032821 accelerated GC development through elevating HMGB1 expression via sponging miR-1236-3p.
Collapse
Affiliation(s)
- Lei Chen
- Department of Emergency Medicine, Xiang'an Hospital of Xiamen University, Xiamen, 361101, People's Republic of China
| | - Kun Chi
- Department of Nursing, Qingdao Municipal Hospital (Group), Qingdao 266071, People's Republic of China
| | - Huaguo Xiang
- Medical Laboratory, Shenzhen Baoan District Fuyong People's Hospital, Shenzhen 518103, People's Republic of China
| | - Yan Yang
- Department of Gastroenterology, Central People's Hospital of Tengzhou, Tengzhou 277500, People's Republic of China
| |
Collapse
|
377
|
Nakajima S, Ogawa N, Yokoue N, Tachibana H, Tamada K, Okazawa M, Sato A, Oyama T, Abe H, Kamiya T, Yoshimori A, Yoshizawa K, Inoue S, Yokomizo T, Uchiumi F, Abe T, Tanuma SI. Trimebutine attenuates high mobility group box 1-receptor for advanced glycation end-products inflammatory signaling pathways. Biochem Biophys Res Commun 2020; 533:1155-1161. [PMID: 33041002 DOI: 10.1016/j.bbrc.2020.09.126] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 01/10/2023]
Abstract
We previously identified papaverine as an inhibitor of receptor for advanced glycation end-products (RAGE) and showed its suppressive effect on high mobility group box 1 (HMGB1)-mediated responses to inflammation. Here, we found trimebutine to be a 3D pharmacophore mimetics of papaverine. Trimebutine was revealed to have more potent suppressive effects on HMGB1-induced production of pro-inflammatory cytokines, such as interleukin-6 and tumor necrosis factor-α in macrophage-like RAW264.7 cells and mouse bone marrow primarily differentiated macrophages than did papaverine. However, the inhibitory effect of trimebutine on the interaction of HMGB1 and RAGE was weaker than that of papaverine. Importantly, mechanism-of-action analyses revealed that trimebutine strongly inhibited the activation of RAGE downstream inflammatory signaling pathways, especially the activation of extracellular signal-regulated kinase 1 and 2 (ERK1/2), which are mediator/effector kinases recruited to the intracellular domain of RAGE. Consequently, the activation of Jun amino terminal kinase, which is an important effector kinase for the up-regulation of pro-inflammatory cytokines, was inhibited. Taken together, these results suggest that trimebutine may exert its suppressive effect on the HMGB1-RAGE inflammatory signal pathways by strongly blocking the recruitment of ERK1/2 to the intracellular tail domain of RAGE in addition to its weak inhibition of the extracellular interaction of HMGB1 with RAGE. Thus, trimebutine may provide a unique scaffold for the development of novel dual inhibitors of RAGE for inflammatory diseases.
Collapse
Affiliation(s)
- Shingo Nakajima
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Natsumi Ogawa
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Natsuki Yokoue
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Haruki Tachibana
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Kenya Tamada
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Miwa Okazawa
- Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Akira Sato
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Takahiro Oyama
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Hideaki Abe
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Takanori Kamiya
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Atsushi Yoshimori
- Institute for Theoretical Medicine Inc., Fujisawa, Kanagawa, 251-0012, Japan
| | - Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Shigeaki Inoue
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Isehara, Kanagawa, 259-1193, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Fumiaki Uchiumi
- Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Takehiko Abe
- Hinoki Shinyaku Co. Ltd., Chiyoda-ku, Tokyo, 102-0084, Japan
| | - Sei-Ichi Tanuma
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan; Department of Genomic Medicinal Science, Research Institute for Science and Technology, Organization for Research Advancement, Tokyo University of Science, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
378
|
Molinaro C, Martoriati A, Pelinski L, Cailliau K. Copper Complexes as Anticancer Agents Targeting Topoisomerases I and II. Cancers (Basel) 2020; 12:E2863. [PMID: 33027952 PMCID: PMC7601307 DOI: 10.3390/cancers12102863] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
Organometallics, such as copper compounds, are cancer chemotherapeutics used alone or in combination with other drugs. One small group of copper complexes exerts an effective inhibitory action on topoisomerases, which participate in the regulation of DNA topology. Copper complexes inhibitors of topoisomerases 1 and 2 work by different molecular mechanisms, analyzed herein. They allow genesis of DNA breaks after the formation of a ternary complex, or act in a catalytic mode, often display DNA intercalative properties and ROS production, and sometimes display dual effects. These amplified actions have repercussions on the cell cycle checkpoints and death effectors. Copper complexes of topoisomerase inhibitors are analyzed in a broader synthetic view and in the context of cancer cell mutations. Finally, new emerging treatment aspects are depicted to encourage the expansion of this family of highly active anticancer drugs and to expend their use in clinical trials and future cancer therapy.
Collapse
Affiliation(s)
- Caroline Molinaro
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| | - Alain Martoriati
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| | - Lydie Pelinski
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181-UCCS-Unité de Catalyse et Chimie du Solide, F-59000 Lille, France;
| | - Katia Cailliau
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| |
Collapse
|
379
|
Molecular insights into the therapeutic promise of targeting HMGB1 in depression. Pharmacol Rep 2020; 73:31-42. [PMID: 33015736 DOI: 10.1007/s43440-020-00163-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/09/2020] [Accepted: 09/19/2020] [Indexed: 12/17/2022]
Abstract
Depression is a common psychiatric disorder, the exact pathogenesis of which is still elusive. Studies have proposed that immunity disproportion and enhancement in proinflammatory cytokines might be linked with the development of depression. HMGB1 (High-mobility group box (1) protein has obtained more interest as an essential factor in inherent immune reactions and a regulating factor in various inflammation-related diseases. HMGB1 is a ubiquitous chromatin protein and is constitutively expressed in nucleated mammalian cells. HMGB1 is released by glial cells and neurons upon inflammasome activation and act as a pro-inflammatory cytokine. HMGB1 is a late mediator of inflammation and has been indicated as a major mediator in various neuroinflammatory diseases. Microglia, which is the brain immune cell, is stimulated by HMGB1 and released inflammatory mediators and induces chronic neurodegeneration in the CNS (central nervous system). In the current review, we aimed to investigate the role of HMGB1 in the pathogenesis of depression. The studies found that HMGB1 functions as proinflammatory cytokines primarily via binding receptors like RAGE (receptor for advanced glycation end product), TLR2 and TLR4 (Toll-like receptor 2 and 4). Further, HMGB1 added to the preparing impacts of stress-pretreatment and assumed a major function in neurodegenerative conditions through moderating neuroinflammation. Studies demonstrated that neuroinflammation played a major role in the development of depression. The patients of depression generally exhibited an elevated amount of proinflammatory cytokines in the serum, microglia activation and neuronal deficit in the CNS.
Collapse
|
380
|
Chou YE, Yang PJ, Lin CY, Chen YY, Chiang WL, Lin PX, Huang ZY, Huang M, Ho YC, Yang SF. The Impact of HMGB1 Polymorphisms on Prostate Cancer Progression and Clinicopathological Characteristics. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17197247. [PMID: 33023053 PMCID: PMC7579148 DOI: 10.3390/ijerph17197247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer is one of the major cancers of the genitourinary tract. High-mobility group box 1 (HMGB1) was suggested as a promising therapeutic target for prostate cancer. In this study, we aim to elucidate the associations of HMGB1 single nucleotide polymorphisms (SNPs) with prostate cancer susceptibility and clinicopathological characteristics. The HMGB1 SNPs rs1412125, rs2249825, rs1045411, and rs1360485 in 579 prostate cancer patients and 579 cancer-free controls were analyzed with real-time polymerase chain reactions (real-time PCR). All of the data were evaluated with SAS statistical software. Our results showed that the HMGB1 rs1045411 T allele genotype was significantly associated with advanced pathologic T stage (odds ratio (OR) = 1.433, 95% confidence interval (CI) = 1.021–2.012; p = 0.037) and pathologic N1 stage (OR = 2.091, 95% CI = 1.160–3.767; p = 0.012), and the rs1360485 polymorphic CT + TT genotype was associated with pathologic Gleason grade group (4 + 5) (OR = 1.583, 95% CI = 1.017–2.462; p = 0.041), pathologic T stage (3 + 4) (OR = 1.482, 95% CI = 1.061–2.070; p = 0.021), and pathologic N1 stage (OR = 2.131, 95% CI = 1.178–3.852; p = 0.011) compared with their wild-type carriers. In conclusion, our results revealed that the HMGB1 SNPs were associated with the clinical status of prostate cancer. The HMGB1 SNPs may have the potential to predict prostate cancer disease progression.
Collapse
Affiliation(s)
- Ying-Erh Chou
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-E.C.); (P.-J.Y.)
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Po-Jen Yang
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan; (Y.-E.C.); (P.-J.Y.)
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Chia-Yen Lin
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan;
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Yen-Yu Chen
- School of Medical Applied Chemistry, Chung Shan Medical University, Taichung 402, Taiwan;
| | - Whei-Ling Chiang
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan; (W.-L.C.); (P.-X.L.); (Z.-Y.H.)
| | - Pei-Xuan Lin
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan; (W.-L.C.); (P.-X.L.); (Z.-Y.H.)
| | - Zih-Yun Huang
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan; (W.-L.C.); (P.-X.L.); (Z.-Y.H.)
| | - Matthew Huang
- White Oaks Secondary School, Oakville, ON L6H 1Z5, Canada;
| | - Yung-Chuan Ho
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- School of Medical Applied Chemistry, Chung Shan Medical University, Taichung 402, Taiwan;
- Correspondence: (Y.-C.H.); (S.-F.Y.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (Y.-C.H.); (S.-F.Y.)
| |
Collapse
|
381
|
Lan J, Luo H, Wu R, Wang J, Zhou B, Zhang Y, Jiang Y, Xu J. Internalization of HMGB1 (High Mobility Group Box 1) Promotes Angiogenesis in Endothelial Cells. Arterioscler Thromb Vasc Biol 2020; 40:2922-2940. [PMID: 32998518 DOI: 10.1161/atvbaha.120.315151] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE In patients with peripheral artery disease, blockages in arterioles <1 mm cannot be treated surgically, and there are currently few effective medicines. Studies have shown that inflammation in ischemic tissue is related to injury recovery and angiogenesis, but insufficient attention has been paid to this area. Studies have suggested that HMGB1 (high mobility group protein 1), which is released by ischemic tissue, promotes angiogenesis, but the mechanism is not entirely clear. In this study, we tested the internalization of HMGB1 in endothelial cells and investigated a novel proangiogenic pathway. Approach and Results: Using green fluorescent protein-tagged HMGB1 to stimulate endothelial cells, we demonstrated HMGB1 internalization via dynamin and RAGE (receptor for advanced glycation end products)-dependent signaling. Using a fluorescence assay, we detected internalized protein fusion to lysosomes, followed by activation of CatB (cathepsin B) and CatL (cathepsin L). The latter promoted the release of VEGF (vascular endothelial growth factor)-A and endoglin and upregulated the capacities of cell migration, proliferation, and tube formation in endothelial cells. We identified that the cytokine-induced fragment-a key functional domain in HMGB1-mediates the internalization and angiogenic function of HMGB1. We further confirmed that HMGB1 internalization also occurs in vivo in endothelial cells and promotes angiogenesis in mouse femoral artery ligation. CONCLUSIONS In this study, we identified a novel pathway of HMGB1 internalization-induced angiogenesis in endothelial cells. This finding sheds light on the regulatory role of inflammatory factors in angiogenesis through cell internalization and opens a new door to understand the relationship between inflammation and angiogenesis in ischemic diseases.
Collapse
Affiliation(s)
- Jiaoli Lan
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rong Wu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Juan Wang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Biying Zhou
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yun Zhang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jia Xu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failured Research, Department of Pathophysiology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
382
|
Asbestos induces mesothelial cell transformation via HMGB1-driven autophagy. Proc Natl Acad Sci U S A 2020; 117:25543-25552. [PMID: 32999071 DOI: 10.1073/pnas.2007622117] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Asbestos causes malignant transformation of primary human mesothelial cells (HM), leading to mesothelioma. The mechanisms of asbestos carcinogenesis remain enigmatic, as exposure to asbestos induces HM death. However, some asbestos-exposed HM escape cell death, accumulate DNA damage, and may become transformed. We previously demonstrated that, upon asbestos exposure, HM and reactive macrophages releases the high mobility group box 1 (HMGB1) protein that becomes detectable in the tissues near asbestos deposits where HMGB1 triggers chronic inflammation. HMGB1 is also detectable in the sera of asbestos-exposed individuals and mice. Searching for additional biomarkers, we found higher levels of the autophagy marker ATG5 in sera from asbestos-exposed individuals compared to unexposed controls. As we investigated the mechanisms underlying this finding, we discovered that the release of HMGB1 upon asbestos exposure promoted autophagy, allowing a higher fraction of HM to survive asbestos exposure. HMGB1 silencing inhibited autophagy and increased asbestos-induced HM death, thereby decreasing asbestos-induced HM transformation. We demonstrate that autophagy was induced by the cytoplasmic and extracellular fractions of HMGB1 via the engagement of the RAGE receptor and Beclin 1 pathway, while nuclear HMGB1 did not participate in this process. We validated our findings in a novel unique mesothelial conditional HMGB1-knockout (HMGB1-cKO) mouse model. Compared to HMGB1 wild-type mice, mesothelial cells from HMGB1-cKO mice showed significantly reduced autophagy and increased cell death. Autophagy inhibitors chloroquine and desmethylclomipramine increased cell death and reduced asbestos-driven foci formation. In summary, HMGB1 released upon asbestos exposure induces autophagy, promoting HM survival and malignant transformation.
Collapse
|
383
|
Maysinger D, Zhang Q, Kakkar A. Dendrimers as Modulators of Brain Cells. Molecules 2020; 25:E4489. [PMID: 33007959 PMCID: PMC7582352 DOI: 10.3390/molecules25194489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/19/2022] Open
Abstract
Nanostructured hyperbranched macromolecules have been extensively studied at the chemical, physical and morphological levels. The cellular structural and functional complexity of neural cells and their cross-talk have made it rather difficult to evaluate dendrimer effects in a mixed population of glial cells and neurons. Thus, we are at a relatively early stage of bench-to-bedside translation, and this is due mainly to the lack of data valuable for clinical investigations. It is only recently that techniques have become available that allow for analyses of biological processes inside the living cells, at the nanoscale, in real time. This review summarizes the essential properties of neural cells and dendrimers, and provides a cross-section of biological, pre-clinical and early clinical studies, where dendrimers were used as nanocarriers. It also highlights some examples of biological studies employing dendritic polyglycerol sulfates and their effects on glia and neurons. It is the aim of this review to encourage young scientists to advance mechanistic and technological approaches in dendrimer research so that these extremely versatile and attractive nanostructures gain even greater recognition in translational medicine.
Collapse
Affiliation(s)
- Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Qiaochu Zhang
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
- Department of Chemistry, McGill University, 801 Sherbrooke St West, Montreal, QC H3A 0B8, Canada
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke St West, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
384
|
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
|
385
|
Saito Reis CA, Padron JG, Norman Ing ND, Kendal-Wright CE. High-mobility group box 1 is a driver of inflammation throughout pregnancy. Am J Reprod Immunol 2020; 85:e13328. [PMID: 32851715 DOI: 10.1111/aji.13328] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/10/2020] [Accepted: 08/18/2020] [Indexed: 12/15/2022] Open
Abstract
A proinflammatory response driven by high-mobility group box 1 (HMGB1) is important for the success of both the early stages of pregnancy and parturition initiation. However, the tight regulation of HMGB1 within these two stages is critical, as increased HMGB1 can manifest into pregnancy-related pathologies. Although during the early stages of pregnancy HMGB1 is critical for the development and implantation of the embryo, and uterine decidualization, high levels within the uterine cavity have been linked to pregnancy failure. In addition, chronic inflammation, resultant from increased HMGB1 within the maternal circulation and gestational tissues, also increases the risk for preterm labor, preterm birth, or infant mortality. Due to the link between HMGB1 and several pregnancy pathologies, the possibility of leveraging HMGB1 as a biomarker has been assessed. However, data are limited that demonstrate how known HMGB1 inhibitors could reduce inflammation within pregnancy. Thus, further research is warranted to improve our understanding of the potential of HMGB1 as a therapeutic target to reduce inflammation within pregnancy. This review aims to describe what is understood about the role of HMGB1 that drives inflammation throughout pregnancy and highlight its potential as a biomarker and therapeutic target within this context.
Collapse
Affiliation(s)
- Chelsea A Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Justin G Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA
| | - Nainoa D Norman Ing
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA
| | - Claire E Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, USA.,Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Manoā, Honolulu, HI, USA.,Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawai'I at Manoā, Honolulu, HI, USA
| |
Collapse
|
386
|
Tang N, Dong Y, Xiao T, Zhao H. LncRNA TUG1 promotes the intervertebral disc degeneration and nucleus pulposus cell apoptosis though modulating miR-26a/HMGB1 axis and regulating NF-κB activation. Am J Transl Res 2020; 12:5449-5464. [PMID: 33042430 PMCID: PMC7540166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
AIMS This study was to investigate the effect of TUG1 on apoptosis and ECM degradation of human degenerative intervertebral disc nucleus pulposus cells (NPCs) and its mechanism. METHODS Human degenerative intervertebral disc NP tissues were obtained from 10 patients with lumbar disc herniation (LDH) who underwent lumbar spine surgery (IDD group), normal intervertebral disc NP tissues were obtained from 10 patients with lumbar vertebrae fractures (LVF group). RESULTS The expression of TUG1 and HMGB1 protein in human degenerative disc NP tissues and NPCs was significantly increased, while the level of miR-26a was significantly decreased. Overexpression of TUG1 inhibited the proliferation while promoted apoptosis and ECM degradation of human degenerative intervertebral disc NPCs. Simultaneously, the effect of TUG1 knockdown on NPCs was opposite. Interestingly, TUG1 acted as an endogenous sponge to down-regulate the expression of miR-26a in NPCs by direct binding to miR-26a. Overexpression of miR-26a reversed the effects of TUG1 overexpression on apoptosis and ECM degradation. Additionally, HMGB1 was a target gene of miR-26a. The increased expression of HMGB1 induced by TUG1 overexpression could be reversed by the introduction of miR-26a mimic. Overexpression of TUG1 significantly upregulated the expression of p65 in the nucleus, while overexpression of TUG1 partially abolished the inhibition of NF-κB by QNZ pretreatment. CONCLUSION TUG1 could promote the apoptosis and ECM degradation of degenerated intervertebral disc NPCs by regulating the miR-26a/HMGB1, which may be involved in the activation of NF-κB pathway.
Collapse
Affiliation(s)
- Ning Tang
- Department of Orthopedic, Chinese Academy of Medical Sciences Peking Union Medical College HospitalBeijing, China
| | - Yulei Dong
- Department of Orthopedic, Chinese Academy of Medical Sciences Peking Union Medical College HospitalBeijing, China
| | - Tinghui Xiao
- Department of Orthopedic, Shenzhen People’s Hospital First Affiliated Hospital of Southern University of Science and TechnologyShenzhen, China
| | - Hong Zhao
- Department of Orthopedic, Chinese Academy of Medical Sciences Peking Union Medical College HospitalBeijing, China
| |
Collapse
|
387
|
Chibaatar E, Le K, Abdoulaye IA, Wu S, Guo Y. Melatonin Ameliorates Lipopolysaccharide-Induced Microglial Inflammation via Triggering SIRT1/HMGB1 Signaling Axis. J Mol Neurosci 2020; 71:691-701. [PMID: 32910356 DOI: 10.1007/s12031-020-01699-1] [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: 06/09/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
Stroke is one of the highest incidence neurological disorder with great morbidity and mortality rate. The secondary neuroinflammation contributed by microglial activation is a consequential response observed in the pathogenesis of stroke. High-mobility group box 1, a non-histone nuclear protein, interacts with immune cells, such as microglia, and leads to a cascade amplification of the secondary neuroinflammatory responses, which are related to neuronal damage later. Melatonin is a neurohormone, well-known as its anti-oxidative and anti-inflammatory effects. However, until now, more findings are required for better understanding about anti-inflammatory effect of melatonin on HMGB1 and HMGB1-triggered pathway in LPS-induced microglial activation. Melatonin effect on the viability of BV2 microglial cells was measured by CCK-8 assay; mRNA levels of HMGB1 and other inflammatory cytokines were determined by quantitative real-time polymerase chain reaction assay or enzyme-linked immunosorbent assays; the protein expression levels of TLR4/MyD88/NF-κB and SIRT1 were detected by Western blot, and HMGB1 translocation and release from BV2 microglial cells were examined by immunofluorescence assay. The results of this study demonstrated that melatonin suppressed LPS-triggered BV2 microglial activation-mediated inflammation by inhibiting high expression and release of HMGB1 and moderating the activation of subsequent TLR4/MyD88/NF-κB signaling pathway, which was activated by SIRT1 elevation. Furthermore, LPS-induced expression of pro-inflammatory cytokines (i.e., TNF-α, IL-6, and IL-1β) was notably reversed by melatonin pre-treatment. In summary, our findings suggest that melatonin may act as a promising therapeutic agent for reducing post-stroke neuroinflammation by targeting HMGB1 and the subsequent signaling axis.
Collapse
Affiliation(s)
- Enkhmurun Chibaatar
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Idriss Ali Abdoulaye
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Shanshan Wu
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Yijing Guo
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
388
|
Dymova MA, Taskaev SY, Richter VA, Kuligina EV. Boron neutron capture therapy: Current status and future perspectives. Cancer Commun (Lond) 2020; 40:406-421. [PMID: 32805063 PMCID: PMC7494062 DOI: 10.1002/cac2.12089] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/09/2020] [Accepted: 08/09/2020] [Indexed: 12/11/2022] Open
Abstract
The development of new accelerators has given a new impetus to the development of new drugs and treatment technologies using boron neutron capture therapy (BNCT). We analyzed the current status and future directions of BNCT for cancer treatment, as well as the main issues related to its introduction. This review highlights the principles of BNCT and the key milestones in its development: new boron delivery drugs and different types of charged particle accelerators are described; several important aspects of BNCT implementation are discussed. BCNT could be used alone or in combination with chemotherapy and radiotherapy, and it is evaluated in light of the outlined issues. For the speedy implementation of BCNT in medical practice, it is necessary to develop more selective boron delivery agents and to generate an epithermal neutron beam with definite characteristics. Pharmacological companies and research laboratories should have access to accelerators for large-scale screening of new, more specific boron delivery agents.
Collapse
Affiliation(s)
- Mayya Alexandrovna Dymova
- Laboratory of BiotechnologyInstitute of Chemical Biology and Fundamental MedicineSiberian Branch of the Russian Academy of SciencesLavrentjeva Av. 8Novosibirsk630090Russia
| | - Sergey Yurjevich Taskaev
- Budker Institute of Nuclear PhysicsSiberian Branch of the Russian Academy of SciencesLavrentjeva Av. 11Novosibirsk630090Russia
- Laboratory of Boron Neutron Capture TherapyNovosibirsk State UniversityPirogova str. 1Novosibirsk630090Russia
| | - Vladimir Alexandrovich Richter
- Laboratory of BiotechnologyInstitute of Chemical Biology and Fundamental MedicineSiberian Branch of the Russian Academy of SciencesLavrentjeva Av. 8Novosibirsk630090Russia
| | - Elena Vladimirovna Kuligina
- Laboratory of BiotechnologyInstitute of Chemical Biology and Fundamental MedicineSiberian Branch of the Russian Academy of SciencesLavrentjeva Av. 8Novosibirsk630090Russia
| |
Collapse
|
389
|
|
390
|
Fu B, Lu X, Zhao EY, Wang JX, Peng SM. HMGB1-induced autophagy promotes extracellular matrix degradation leading to intervertebral disc degeneration. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2240-2248. [PMID: 33042328 PMCID: PMC7539879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 01/19/2020] [Indexed: 06/11/2023]
Abstract
Intervertebral disc degeneration (IDD) remains a leading cause of adult disability. High mobility group box l (HMGB1) is a nuclear DNA-binding protein and acts as a central mediator of inflammation. The purpose of this study was to investigate the effect of HMGB1 in IDD. In our study, IDD intervertebral disc tissues were collected and nucleus pulposus cells (NPCs) were primarily cultured. The HMGB1 expression and the effect of HMGB1 on NPCs and extracellular matrix and autophagy were all evaluated. Results showed that HMGB1 was markedly overexpressed in IDD (P<0.05), and upregulated expression of HMGB1 can inhibit NPC proliferation and promote NPC apoptosis (P<0.05), promote extracellular matrix degradation, and activate cell autophagy (P<0.05). Therefore, we concluded that HMGB1 was up-regulated in IDD and HMGB1-induced autophagy can promotes extracellular matrix degradation and thus lead to intervertebral disc degeneration. In brief, HMGB1 may serve as a novel diagnostic biomarker and therapeutic target for IDD.
Collapse
Affiliation(s)
- Biao Fu
- Department of Neurosurgery, The People Hospital of Xin ChangNo. 117 Gushan Middle Road, Nanming Street, Xin Chang 312500, Zhejiang, China
| | - Xia Lu
- Department of Neurosurgery, The People Hospital of Xin ChangNo. 117 Gushan Middle Road, Nanming Street, Xin Chang 312500, Zhejiang, China
| | - Er-Yang Zhao
- Department of Neurosurgery, The People Hospital of Xin ChangNo. 117 Gushan Middle Road, Nanming Street, Xin Chang 312500, Zhejiang, China
| | - Ji-Xin Wang
- Department of Spinal Surgery, Zhejiang Quhua HospitalNo. 62 Wenchang Road, Quzhou 475000, Zhejiang, China
| | - Si-Min Peng
- Department of Neurosurgery, The People Hospital of Xin ChangNo. 117 Gushan Middle Road, Nanming Street, Xin Chang 312500, Zhejiang, China
| |
Collapse
|
391
|
Huang XY, Li H. Correlation of blood flow in hepatocellular carcinoma with serum high mobility group box protein 1 and microvascular invasion: A preliminary study. Shijie Huaren Xiaohua Zazhi 2020; 28:807-812. [DOI: 10.11569/wcjd.v28.i16.807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The occurrence and development of tumors are closely related to microcirculation and high mobility group box protein 1 (HMGB1) expression levels. It is very important to predict the microvascular invasion (MVI) of hepatocellular carcinoma (HCC) before operation.
AIM To quantitatively analyze the blood flow characteristics of HCC by contrast-enhanced ultrasound (CEUS), and to explore the correlation of the blood flow with HMGB1 and MVI.
METHODS Seventy-eight patients with HCC who underwent surgical resection were included. According to the postoperative pathological results, the patients were divided into either an MVI group (45 cases) or a non-MVI group (33 cases). All patients underwent CEUS 3 d before operation. The enhancement intensity (EI) and area under the curve (AUC) in HCC were measured by time-intensity curve analysis. Serum HMGB1 and vascular endothelial growth factor (VEGF) were measured by enzyme-linked immunosorbent assay. Microvessel density was measured after operation. The correlation between EI, AUC, and serum HMGB1 was analyzed.
RESULTS EI and AUC in the MVI group were significantly higher than those in the non-MVI group (P < 0.05). Serum HMGB1 and VEGF were significantly higher than those of the non-MVI group (P < 0.05). EI and AUC were positively correlated with serum HMGB1 (r = 0.729 and 0.759, P < 0.05).
CONCLUSION CEUS can quantitatively analyze the blood flow in HCC, and its blood flow parameters have a good correlation with serum HMGB1, which can provide reference for noninvasive assessment of MVI before operation.
Collapse
Affiliation(s)
- Xiao-Yi Huang
- Department of Laboratory Medicine, Hangzhou Yuhang District Hospital of Traditional Chinese Medicine, Hangzhou 311106, Zhejiang Province, China
| | - Hao Li
- Department of Ultrasonography, Traditional Chinese Medicine Hospital of Hangzhou, Hangzhou 310013, Zhejiang Province, China
| |
Collapse
|
392
|
Somayajulu M, Ekanayaka S, McClellan SA, Bessert D, Pitchaikannu A, Zhang K, Hazlett LD. Airborne Particulates Affect Corneal Homeostasis and Immunity. Invest Ophthalmol Vis Sci 2020; 61:23. [PMID: 32301974 PMCID: PMC7401652 DOI: 10.1167/iovs.61.4.23] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Purpose To determine the effects of airborne particulate matter (PM) <2.5 µm in vitro and on the normal and Pseudomonas aeruginosa (PA)-infected cornea. Methods An MTT viability assay tested the effects of PM2.5 on mouse corneal epithelial cells (MCEC) and human corneal epithelial cells (HCET). MCEC were tested for reactive oxygen species using a 2′,7′-dichlorodihydrofluorescein assay; RT-PCR determined mRNA levels of inflammatory and oxidative stress markers in MCEC (HMGB1, toll-like receptor 2, IL-1β, CXCL2, GPX1, GPX2, GR1, superoxide dismutase 2, and heme oxygenase 1) and HCET (high mobility group box 1, CXCL2, and IL-1β). C57BL/6 mice also were infected and after 6 hours, the PM2.5 was topically applied. Disease was graded by clinical score and evaluated by histology, plate count, myeloperoxidase assay, RT-PCR, ELISA, and Western blot. Results After PM2.5 (25–200 µg/mL), 80% to 90% of MCEC and HCET were viable and PM exposure increased reactive oxygen species in MCEC and mRNA expression levels for inflammatory and oxidative stress markers in mouse and human cells. In vivo, the cornea of PA+PM2.5 exposed mice exhibited earlier perforation over PA alone (confirmed histologically). In cornea, plate counts were increased after PA+PM2.5, whereas myeloperoxidase activity was significantly increased after PA+PM2.5 over other groups. The mRNA levels for several proinflammatory and oxidative stress markers were increased in the cornea in the PA+PM2.5 over other groups; protein levels were elevated for high mobility group box 1, but not toll-like receptor 4 or glutathione reductase 1. Uninfected corneas treated with PM2.5 did not differ from normal. Conclusions PM2.5 triggers reactive oxygen species, upregulates mRNA levels of oxidative stress, inflammatory markers, and high mobility group box 1 protein, contributing to perforation in PA-infected corneas.
Collapse
|
393
|
Protective effects of mild hypothermia against hepatic injury in rats with acute liver failure. Ann Hepatol 2020; 18:770-776. [PMID: 31422029 DOI: 10.1016/j.aohep.2019.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 01/25/2019] [Accepted: 12/17/2018] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES Acute liver failure (ALF) is a severe disease which is associated with a high mortality rate. As mild hypothermia has been shown to have protective effects on the brain, this study aimed to determine whether it also provides protection to the liver in rats with ALF and to explore its underlying mechanism. MATERIALS AND METHODS In total, 72 rats were divided into 3 groups: control group (CG, treated with normal saline), normothermia group (NG, treated with d-galactosamine and lipopolysaccharide; d-GalN/LPS), and mild hypothermia group (MHG, treated with d-GalN/LPS and kept in a state of mild hypothermia, defined as an anal temperature of 32-35°C). The rats were examined at 4, 8, and 12h after treatment. RESULTS Mild hypothermia treatment significantly reduced serum alanine transaminase and aspartate transaminase levels and improved the liver condition of rats with d-GalN/LPS-induced ALF at 12h. Serum tumor necrosis factor-alpha levels were significantly lower in the MHG than in the NG at 4h, but no significant differences were observed in the interleukin-10 levels between the NG and MHG at any time. The serum and hepatic levels of high mobility group box 1 were significantly lower in the MHG than in the NG at 8 and 12h. The protein expression levels of cytochrome C and cleaved-caspase 3 in hepatic tissues were significantly lower in the MHG than in the NG at 8h. CONCLUSION Mild hypothermia improved the liver conditions of rats with ALF via its anti-inflammatory and anti-apoptotic effects.
Collapse
|
394
|
Wang B, Huang X, Pan X, Zhang T, Hou C, Su WJ, Liu LL, Li JM, Wang YX. Minocycline prevents the depressive-like behavior through inhibiting the release of HMGB1 from microglia and neurons. Brain Behav Immun 2020; 88:132-143. [PMID: 32553784 DOI: 10.1016/j.bbi.2020.06.019] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Our previous study reports the causal role of high mobility group box 1 (HMGB1) in the development of depression; and we find glycyrrhizic acid (GZA) can be a potential treatment for major depressive disorder (MDD) considering its inhibition of HMGB1 activity. This study aims to further explore the exact cell types that release HMGB1 in the hippocampus. METHODS We detected the effects of microglia conditioned medium on primary astrocytes and neurons. The effects of minocycline on depressive-like behaviors were tested in BABLB/c mice after four weeks of chronic unpredictable mild stress (CUMS) exposure. Furthermore, the immunofluorescence (IF) assays, hematoxylin-eosin (HE) and TUNEL staining were used to observe hippocampal slices to evaluate the release of HMGB1. The cytoplasmic translocations of HMGB1 protein were assayed by western-blot. RESULTS Exposure to CUMS caused an active release of HMGB1 from microglia and neurons in the hippocampus. After minocycline administration for inhibiting the activation of microglia, both microglia and neurons reduced the release of HMGB1 and the protein level of central and peripheral HMGB1 recovered accordingly. Along with blocking the release of HMGB1, behavioral and cognitive deficits induced by CUMS were improved significantly by minocycline. In addition, the supernatant of primary microglia stimulated the secretion of HMGB1 in primary neurons, not in astrocytes, at 24 h after 4 h-LPS treatment. CONCLUSION All the evidence supported our hypotheses that microglia and neurons are the main cell sources of HMGB1 release under CUMS condition, and that the release of HMGB1 by microglia may play an important role in the development of depressive-like behavior.
Collapse
Affiliation(s)
- Bo Wang
- Department of Nautical Psychology, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China; Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China; Department of Medicine, The Unit 31641 of PLA, Xishuangbanna 666100, China
| | - Xiao Huang
- Department of Anaesthesiology, South Campus, Ren Ji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 201112, China
| | - Xiao Pan
- Department of Medical Psychology, Changzheng Hospital, Navy Medical University, Shanghai 200003, China
| | - Ting Zhang
- Department of Nautical Psychology, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China
| | - Cheng Hou
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China
| | - Lin-Lin Liu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China
| | - Jia-Mei Li
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China
| | - Yun-Xia Wang
- Department of Nautical Psychology, Faculty of Psychology, Second Military Medical University, Shanghai 200433, China; Department of Medical Psychology, Changzheng Hospital, Navy Medical University, Shanghai 200003, China.
| |
Collapse
|
395
|
Kaur I, Behl T, Bungau S, Kumar A, Mehta V, Setia D, Uddin MS, Zengin G, Aleya L, Arora S. Exploring the therapeutic promise of targeting HMGB1 in rheumatoid arthritis. Life Sci 2020; 258:118164. [PMID: 32739467 DOI: 10.1016/j.lfs.2020.118164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/25/2020] [Accepted: 07/25/2020] [Indexed: 12/22/2022]
Abstract
High mobility group box-1 (HMGB1) protein is a diverse, single polypeptide moiety, present in mammalian eukaryotic cells. In response to stimuli, this nuclear protein is actively secreted in to the extracellular compartment or passively released by the necrotic cells, in order to mediate inflammatory responses, by forming complexes with IL-1α, IL-1β, LPS and other moieties, and binding to RAGE, TLR and other receptor ligands, initiating downstream, signaling processes. This molecule acts as a proinflammatory cytokine and contributes to the progression of diseases like, acute lung injury, autoimmune liver damage, graft rejection immune response and arthritis. Small concentrations of HMGB1 are released during apoptosis, which facilitates oxidative regulation on Cys106, and propagates immune inactivating tolerogenic signals in the body. The review portrays the role of HMGB1 in rheumatoid arthritis, evidently supported by pre-clinical and clinical investigations, demonstrating extensive HMGB1 expression in synovial tissue and fluid as well as serum, excessive expression of transduction receptor signaling molecules, bone remodeling and uncontrolled expression of bone destroying osteoclastogenesis, resulting in destruction of articular cartilage, bone deformation and synovial proliferation, alleviating the pathogenesis in RA disease. Moreover, the review highlights the therapeutic regime targeting HMGB1, facilitating inhibition of its actions and release into the extracellular compartment, to ameliorate the destructive events that prevail in rheumatoid arthritis.
Collapse
Affiliation(s)
- Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine of Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, Distt. Shimla, Himachal Pradesh, India
| | - Dhruv Setia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey
| | - Lotfi Aleya
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey; Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
396
|
Nebbioso M, Lambiase A, Armentano M, Tucciarone G, Bonfiglio V, Plateroti R, Alisi L. The Complex Relationship between Diabetic Retinopathy and High-Mobility Group Box: A Review of Molecular Pathways and Therapeutic Strategies. Antioxidants (Basel) 2020; 9:antiox9080666. [PMID: 32722545 PMCID: PMC7464385 DOI: 10.3390/antiox9080666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a protein that is part of a larger family of non-histone nuclear proteins. HMGB1 is a ubiquitary protein with different isoforms, linked to numerous physiological and pathological pathways. HMGB1 is involved in cytokine and chemokine release, leukocyte activation and migration, tumorigenesis, neoangiogenesis, and the activation of several inflammatory pathways. HMGB1 is, in fact, responsible for the trigger, among others, of nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), toll-like receptor-4 (TLR-4), and vascular endothelial growth factor (VEGF) pathways. Diabetic retinopathy (DR) is a common complication of diabetes mellitus (DM) that is rapidly growing in number. DR is an inflammatory disease caused by hyperglycemia, which determines the accumulation of oxidative stress and cell damage, which ultimately leads to hypoxia and neovascularization. Recent evidence has shown that hyperglycemia is responsible for the hyperexpression of HMGB1. This protein activates numerous pathways that cause the development of DR, and HMGB1 levels are constantly increased in diabetic retinas in both proliferative and non-proliferative stages of the disease. Several molecules, such as glycyrrhizin (GA), have proven effective in reducing diabetic damage to the retina through the inhibition of HMGB1. The main focus of this review is the growing amount of evidence linking HMGB1 and DR as well as the new therapeutic strategies involving this protein.
Collapse
Affiliation(s)
- Marcella Nebbioso
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Alessandro Lambiase
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
- Correspondence: ; Tel.: +39-06-4997-5357; Fax: +39-06-4997-5425
| | - Marta Armentano
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Giosuè Tucciarone
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Vincenza Bonfiglio
- Department of Ophthalmology, University of Catania, Via S. Sofia 76, 95100 Catania, Italy;
| | - Rocco Plateroti
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Ludovico Alisi
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| |
Collapse
|
397
|
Late Peaks of HMGB1 and Sepsis Outcome: Evidence For Synergy With Chronic Inflammatory Disorders. Shock 2020; 52:334-339. [PMID: 30239421 DOI: 10.1097/shk.0000000000001265] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
High mobility group box 1 (HMGB1) is released from macrophages as a late biomarker of sepsis. Conditions associated with pre-existing macrophage activation may modify HMGB1 expression. This study aimed to assess the impact of HMGB1 kinetics on 28-day mortality. In a sub-study of a previous randomized clinical trial among patients with systemic inflammatory response syndrome and gram-negative infections, patients were classified in early and late HMGB1 peak groups. Serial measurements of HMGB1, ferritin and interferon-gamma (IFNγ) were performed in all available sera. Two hundred ten patients were included; 118 (46.5%) had at least one inflammatory disease (diabetes, chronic obstructive pulmonary disease, chronic heart failure, or chronic renal disease). Mortality after 28 days was higher among patients with a late peak of HMGB1 (OR 2.640; P = 0.026). Co-existence of late peak and inflammatory disease synergistically impacted mortality (odds ratio of logistic regression analysis 3.17; P: 0.027). Late peak was concomitantly associated with higher values of ferritin (P = 0.035), and IFNγ (P = 0.002) among patients with hyperferritinemia. It is concluded that late HMGB1 peak was associated with worse prognosis, especially in patients with underlying chronic inflammatory conditions.
Collapse
|
398
|
Non-Thermal Plasma Application in Tumor-Bearing Mice Induces Increase of Serum HMGB1. Int J Mol Sci 2020; 21:ijms21145128. [PMID: 32698492 PMCID: PMC7404183 DOI: 10.3390/ijms21145128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022] Open
Abstract
The application of cold atmospheric plasma (CAP) in cancer therapy could be one of the new anticancer strategies. In the current work, we used cold atmospheric plasma jet for the treatment of cultured cells and mice. We showed that CAP induced the death of MX-7 mouse rhabdomyosarcoma cells with the hallmarks of immunogenic cell death (ICD): calreticulin and heat shock protein 70 (HSP70) externalization and high-mobility group box 1 protein (HMGB1) release. The intensity of HMGB1 release after the CAP treatment correlated directly with the basal extracellular HMGB1 level. Releasing from dying cells, HMGB1 can act as a proinflammatory cytokine. Our in vivo study demonstrated that cold atmospheric plasma induces a short-term two-times increase in serum HMGB1 level only in tumor-bearing mice with no effect in healthy mice. These findings support our hypothesis that CAP-dependent HMGB1 release from dying cancer cells can change the serum HMGB1 level. At the same time, we showed a weak cytokine response to CAP irradiation in healthy mice that can characterize CAP as an immune-safety physical antitumor approach.
Collapse
|
399
|
Chen Q, Zhang Y, Yue ZD, Zhao HW, Wang L, Fan ZH, Liu FQ. High-mobility group protein B1: A predictive biomarker for hepatic encephalopathy after transjugular intrahepatic portosystemic shunt. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2020; 27:522-530. [PMID: 32463549 DOI: 10.1002/jhbp.770] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/28/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The aim of the present study was to investigate whether portal level of high-mobility group protein B1 (HMGB1) is associated with hepatic encephalopathy (HE) after transjugular intrahepatic portosystemic shunt (TIPS). METHODS We enrolled 127 consecutive patients who underwent TIPS and collected portal and peripheral blood samples in our department from December 2017 to May 2019. HMGB1 levels were determined using enzyme-linked immunosorbent assay kits. HMGB1 and other HE related parameters were estimated by competing risk analysis, receiver operating characteristic (ROC) analysis and Kaplan-Meier analysis. RESULTS Patients with HE after TIPS were older (P = .019) and had higher portal HMGB1 level (P = .038) than those without. Univariate competing risk analysis: age (sHR 1.025, P = .026), hepatorenal syndrome (sHR 3.149, P = .010), model for end-of-stage liver disease (MELD) score (sHR 1.055, P = .024), prior HE (sHR 4.029, P = .0005), portal HMGB1 before TIPS (sHR 1.177, P = .001) reached statistical significance. Multivariate analysis: age (sHR 1.025, P = .037), MELD score (sHR 1.062, P = .011), prior HE (sHR 2.492, P = .030) and portal HMGB1 level before TIPS (sHR 1.217, P = .0002) were significantly different. ROC analyses and Kaplan-Meier curve showed portal HMGB1 level changes before and after TIPS (ΔHMGB1) had good predictive value in the cut-off 0.012 ng/mL (AUC = 0.748, P < .001, Sensitivity = 0.743, Specificity = 0.655). CONCLUSIONS Portal HMGB1 may be a therapeutic target for post-TIPS HE.
Collapse
Affiliation(s)
- Quan Chen
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Yu Zhang
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Zhen-Dong Yue
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Hong-Wei Zhao
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Lei Wang
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Zhen-Hua Fan
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| | - Fu-Quan Liu
- Department of Interventional Therapy, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Beijing, China
| |
Collapse
|
400
|
Yuan S, Liu Z, Xu Z, Liu J, Zhang J. High mobility group box 1 (HMGB1): a pivotal regulator of hematopoietic malignancies. J Hematol Oncol 2020; 13:91. [PMID: 32660524 PMCID: PMC7359022 DOI: 10.1186/s13045-020-00920-3] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin-associated protein that has been widely reported to play a pivotal role in the pathogenesis of hematopoietic malignancies. As a representative damage-associated molecular pattern (DAMP), HMGB1 normally exists inside cells but can be secreted into the extracellular environment through passive or active release. Extracellular HMGB1 binds with several different receptors and interactors to mediate the proliferation, differentiation, mobilization, and senescence of hematopoietic stem cells (HSCs). HMGB1 is also involved in the formation of the inflammatory bone marrow (BM) microenvironment by activating proinflammatory signaling pathways. Moreover, HMGB1-dependent autophagy induces chemotherapy resistance in leukemia and multiple myeloma. In this review, we systematically summarize the emerging roles of HMGB1 in carcinogenesis, progression, prognosis, and potential clinical applications in different hematopoietic malignancies. In summary, targeting the regulation of HMGB1 activity in HSCs and the BM microenvironment is highly beneficial in the diagnosis and treatment of various hematopoietic malignancies.
Collapse
Affiliation(s)
- Shunling Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaoping Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhenru Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Jing Liu
- Hunan Province Key Laboratory of Basic and Applied Hematology, Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Ji Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|